1
|
Motta I, Soccio M, Guidotti G, Lotti N, Pasquinelli G. Hydrogels for Cardio and Vascular Tissue Repair and Regeneration. Gels 2024; 10:196. [PMID: 38534614 DOI: 10.3390/gels10030196] [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: 01/17/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Cardiovascular disease (CVD), the leading cause of death globally, affects the heart and arteries with a variety of clinical manifestations, the most dramatic of which are myocardial infarction (MI), abdominal aortic aneurysm (AAA), and intracranial aneurysm (IA) rupture. In MI, necrosis of the myocardium, scar formation, and loss of cardiomyocytes result from insufficient blood supply due to coronary artery occlusion. Beyond stenosis, the arteries that are structurally and functionally connected to the cardiac tissue can undergo pathological dilation, i.e., aneurysmal dilation, with high risk of rupture. Aneurysms of the intracranial arteries (IAs) are more commonly seen in young adults, whereas those of the abdominal aorta (AAA) are predominantly seen in the elderly. IAs, unpredictably, can undergo rupture and cause life-threatening hemorrhage, while AAAs can result in rupture, internal bleeding and high mortality rate. In this clinical context, hydrogels, three-dimensional networks of water-seizing polymers, have emerged as promising biomaterials for cardiovascular tissue repair or protection due to their biocompatibility, tunable properties, and ability to encapsulate and release bioactive molecules. This review provides an overview of the current state of research on the use of hydrogels as an innovative platform to promote cardiovascular-specific tissue repair in MI and functional recovery or protection in aneurysmal dilation.
Collapse
Affiliation(s)
- Ilenia Motta
- Alma Mater Institute on Healthy Planet, University of Bologna, Via Massarenti 11, 40138 Bologna, Italy
| | - Michelina Soccio
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Giulia Guidotti
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Nadia Lotti
- Civil, Chemical, Environmental and Materials Engineering Department, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Gianandrea Pasquinelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
2
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
3
|
Chen X, Zhu L, Wang X, Xiao J. Insight into Heart-Tailored Architectures of Hydrogel to Restore Cardiac Functions after Myocardial Infarction. Mol Pharm 2023; 20:57-81. [PMID: 36413809 DOI: 10.1021/acs.molpharmaceut.2c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
With permanent heart muscle injury or death, myocardial infarction (MI) is complicated by inflammatory, proliferation and remodeling phases from both the early ischemic period and subsequent infarct expansion. Though in situ re-establishment of blood flow to the infarct zone and delays of the ventricular remodeling process are current treatment options of MI, they fail to address massive loss of viable cardiomyocytes while transplanting stem cells to regenerate heart is hindered by their poor retention in the infarct bed. Equipped with heart-specific mimicry and extracellular matrix (ECM)-like functionality on the network structure, hydrogels leveraging tissue-matching biomechanics and biocompatibility can mechanically constrain the infarct and act as localized transport of bioactive ingredients to refresh the dysfunctional heart under the constant cyclic stress. Given diverse characteristics of hydrogel including conductivity, anisotropy, adhesiveness, biodegradability, self-healing and mechanical properties driving local cardiac repair, we aim to investigate and conclude the dynamic balance between ordered architectures of hydrogels and the post-MI pathological milieu. Additionally, our review summarizes advantages of heart-tailored architectures of hydrogels in cardiac repair following MI. Finally, we propose challenges and prospects in clinical translation of hydrogels to draw theoretical guidance on cardiac repair and regeneration after MI.
Collapse
Affiliation(s)
- Xuerui Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Liyun Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xu Wang
- Hangzhou Medical College, Binjiang Higher Education Park, Binwen Road 481, Hangzhou 310053, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
4
|
Nanogels: Update on the methods of synthesis and applications for cardiovascular and neurological complications. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
5
|
Tapeinos C, Gao H, Bauleth-Ramos T, Santos HA. Progress in Stimuli-Responsive Biomaterials for Treating Cardiovascular and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200291. [PMID: 35306751 DOI: 10.1002/smll.202200291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) describe abnormal vascular system conditions affecting the brain and heart. Among these, ischemic heart disease and ischemic stroke are the leading causes of death worldwide, resulting in 16% and 11% of deaths globally. Although several therapeutic approaches are presented over the years, the continuously increasing mortality rates suggest the need for more advanced strategies for their treatment. One of these strategies lies in the use of stimuli-responsive biomaterials. These "smart" biomaterials can specifically target the diseased tissue, and after "reading" the altered environmental cues, they can respond by altering their physicochemical properties and/or their morphology. In this review, the progress in the field of stimuli-responsive biomaterials for CCVDs in the last five years, aiming at highlighting their potential as early-stage therapeutics in the preclinical scenery, is described.
Collapse
Affiliation(s)
- Christos Tapeinos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Han Gao
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Tomás Bauleth-Ramos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
6
|
Wen X, Peng Y, Gao M, Zhu Y, Zhu Y, Yu F, Zhou T, Shao J, Feng L, Ma X. Endothelial Transient Receptor Potential Canonical Channel Regulates Angiogenesis and Promotes Recovery After Myocardial Infarction. J Am Heart Assoc 2022; 11:e023678. [PMID: 35253458 PMCID: PMC9075314 DOI: 10.1161/jaha.121.023678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background
Transient receptor potential canonical (TRPC) channels play a role in angiogenesis. However, the involvement of TRPC1 in myocardial infarction (MI) remains unclear. The present study was aimed at investigating whether TRPC1 can improve the recovery of cardiac function via prompting angiogenesis following MI.
Methods and Results
In vitro, coronary artery endothelial cells from floxed TRPC1 mice and endothelial cell‐specific TRPC1 channel knockout mice were cultured to access EC angiogenesis. Both EC tube formation and migration were significantly suppressed in mouse coronary artery endothelial cells from endothelial cell‐specific TRPC1 channel knockout mice. In vivo, coronary artery endothelial cells from floxed TRPC1 and endothelial cell‐specific TRPC1 channel knockout mice were subjected to MI, then echocardiography, triphenyltetrazolium chloride staining and immunofluorescence were performed to assess cardiac repair on day 28. Endothelial cell‐specific TRPC1 channel knockout mice had higher ejection fraction change, larger myocardial infarct size, and reduced capillary density in the infarct area compared with coronary artery endothelial cells from floxed TRPC1 mice. Furthermore, we found underlying regulation by HIF‐1α (hypoxic inducible factor‐1α) and MEK‐ERK (mitogen‐activated protein kinase/extracellular signal‐regulated kinase) that could be the mechanism for the angiogenetic action of TRPC1. Significantly, treatment with dimethyloxaloylglycine, an activator of HIF‐1α, induced cardiac improvement via the HIF‐1α‐TRPC1‐MEK/ERK pathway in MI mice.
Conclusions
Our study demonstrated TRPC1 improves cardiac function after MI by increasing angiogenesis via the upstream regulator HIF‐1α and downstream MEK/ERK, and dimethyloxaloylglycine treatment has protective effect on MI through the HIF‐1α‐TRPC1‐MEK/ERK pathway.
Collapse
Affiliation(s)
- Xin Wen
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Yidi Peng
- School of Pharmaceutical Sciences Jiangnan University Wuxi China
| | - Mengru Gao
- School of Pharmaceutical Sciences Jiangnan University Wuxi China
| | - Yuzhong Zhu
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Yifei Zhu
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Fan Yu
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Tingting Zhou
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Jing Shao
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Lei Feng
- Wuxi School of Medicine Jiangnan University Wuxi China
| | - Xin Ma
- Wuxi School of Medicine Jiangnan University Wuxi China
- School of Pharmaceutical Sciences Jiangnan University Wuxi China
| |
Collapse
|
7
|
Cardiac-derived stem cell engineered with constitutively active HIF-1α gene enhances blood perfusion of hindlimb ischemia. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.09.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
8
|
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: 1.0] [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
|
9
|
Zhang Y, Yin WH, Yang F, An YQ, Zhou W, Yu H, Xie H, Zhang YL, Zhu Y, Shen XC, Tian R. VEGF121 Mediates Post-Hypoxia Cardioprotective Effects Via CaSR and Mitochondria-Dependent Protease Pathway. Arq Bras Cardiol 2021; 117:476-483. [PMID: 34550233 PMCID: PMC8462959 DOI: 10.36660/abc.20190902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/24/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cardiovascular disease is the major cause of death worldwide. Hypoxia-mediated apoptosis in cardiomyocytes is a major cause of cardiovascular disorders. Treatment with vascular endothelial growth factor (VEGF) protein has been tested but operational difficulties have limited its use. However, with the advancements of gene therapy, interest has risen in VEGF-based gene therapy in cardiovascular disorders. However, the precise mechanism by which VEGF replenishment rescues post-hypoxia damage in cardiomyocytes is not known. OBJECTIVES To investigate the effect of post-hypoxia VEGF121 expression using neonatal rat cardiomyocytes. METHODS Cardiomyocytes isolated from neonatal rats were used to establish an in vitro model of hypoxia-induced cardiac injury. The effect of VEGF overexpression, alone or in combination with small-molecule inhibitors targeting calcium channel, calcium sensitive receptors (CaSR), and calpain on cell growth and proliferation on hypoxia-induced cardiomyocyte injury were determined using an MTT assay, TUNEL staining, Annexin V/PI staining, lactate dehydrogenase and caspase activity. For statistical analysis, a value of P<0.05 was considered to be significant. RESULTS The effect of VEGF121 was found to be mediated by CaSR and calpain but was not dependent on calcium channels. CONCLUSIONS Our findings, even though using an in vitro setting, lay the foundation for future validation and pre-clinical testing of VEGF-based gene therapy in cardiovascular diseases.
Collapse
Affiliation(s)
- Yan Zhang
- Hospital of Guizhou Medical University, Guiyang - China
| | - Wei-Hua Yin
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing - China
| | - Fan Yang
- Hospital of Guizhou Medical University, Guiyang - China
| | - Yun-Qiang An
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing - China
| | - Wei Zhou
- Hospital of Guizhou Medical University, Guiyang - China
| | - Hui Yu
- Hospital of Guizhou Medical University, Guiyang - China
| | - Hong Xie
- Hospital of Guizhou Medical University, Guiyang - China
| | | | - Yue Zhu
- Hospital of Guizhou Medical University, Guiyang - China
| | | | - Ruiqing Tian
- The First People's Hospital of Guiyang, Guiyang - China
| |
Collapse
|
10
|
Fan Z, Wei Y, Yin Z, Huang H, Liao X, Sun L, Liu B, Liu F. Near-Infrared Light-Triggered Unfolding Microneedle Patch for Minimally Invasive Treatment of Myocardial Ischemia. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40278-40289. [PMID: 34424666 DOI: 10.1021/acsami.1c09658] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
It is hard to achieve safe, effective, and minimally invasive therapies on myocardial infarction (MI) via conventional treatments. To address this challenge, a vascular endothelial growth factor (VEGF)-loaded and near-infrared (NIR)-triggered self-unfolding graphene oxide (GO)-poly(vinyl alcohol) (PVA) microneedle (MN) patch was designed and fabricated to treat MI through a minimally invasive surgery (MIS). The folded MN patch can be easily placed into the chest cavity through a small cut (4 mm) and quickly recover to its original shape with 10 s of irradiation of NIR light (1.5 W/cm2, beam diameter = 0.5 cm), thanks to its excellent shape memory effect and fast shape recovery ability. Meanwhile, the unfolded MN patch can be readily punctured into the heart and wrap the heart tightly, thanks to its sufficient mechanical strength and adjustable morphological structure, thus ensuring a high fixation strength to withstand the high-frequency pulsation of the heart. In addition, the prepared MN patch has low cytotoxicity and controllable and sustainable release of VEGF. More importantly, the MN patch can effectively promote neovascularization, reduce myocardial fibrosis, and restore cardiac function, which indicates its promising application prospects in MIS.
Collapse
Affiliation(s)
- Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, Lanzhou University, Lanzhou 730000, People's Republic of China
- School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Yuan Wei
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, Lanzhou University, Lanzhou 730000, People's Republic of China
- School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Zhengrong Yin
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, Lanzhou University, Lanzhou 730000, People's Republic of China
- School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Haofei Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, Lanzhou University, Lanzhou 730000, People's Republic of China
- School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xiaozhu Liao
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, Lanzhou University, Lanzhou 730000, People's Republic of China
- School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Luyi Sun
- Polymer Program, Institute of Materials Science and Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, Lanzhou University, Lanzhou 730000, People's Republic of China
- School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Fengzhen Liu
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng 252000, People's Republic of China
| |
Collapse
|
11
|
Hemalatha T, Aarthy M, Pandurangan S, Kamini NR, Ayyadurai N. A deep dive into the darning effects of biomaterials in infarct myocardium: current advances and future perspectives. Heart Fail Rev 2021; 27:1443-1467. [PMID: 34342769 DOI: 10.1007/s10741-021-10144-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/21/2022]
Abstract
Myocardial infarction (MI) occurs due to the obstruction of coronary arteries, a major crux that restricts blood flow and thereby oxygen to the distal part of the myocardium, leading to loss of cardiomyocytes and eventually, if left untreated, leads to heart failure. MI, a potent cardiovascular disorder, requires intense therapeutic interventions and thereby presents towering challenges. Despite the concerted efforts, the treatment strategies for MI are still demanding, which has paved the way for the genesis of biomaterial applications. Biomaterials exhibit immense potentials for cardiac repair and regeneration, wherein they act as extracellular matrix replacing scaffolds or as delivery vehicles for stem cells, protein, plasmids, etc. This review concentrates on natural, synthetic, and hybrid biomaterials; their function; and interaction with the body, mechanisms of repair by which they are able to improve cardiac function in a MI milieu. We also provide focus on future perspectives that need attention. The cognizance provided by the research results certainly indicates that biomaterials could revolutionize the treatment paradigms for MI with a positive impact on clinical translation.
Collapse
Affiliation(s)
- Thiagarajan Hemalatha
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Mayilvahanan Aarthy
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Suryalakshmi Pandurangan
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Numbi Ramudu Kamini
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Niraikulam Ayyadurai
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India.
| |
Collapse
|
12
|
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: 8.0] [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
|
13
|
Biotherapeutic-loaded injectable hydrogels as a synergistic strategy to support myocardial repair after myocardial infarction. J Control Release 2021; 335:216-236. [PMID: 34022323 DOI: 10.1016/j.jconrel.2021.05.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
Myocardial infarction (MI) has been considered as the leading cause of cardiovascular-related deaths worldwide. Although traditional therapeutic agents including various bioactive species such as growth factors, stem cells, and nucleic acids have demonstrated somewhat usefulness for the restoration of cardiac functions, the therapeutic efficiency remains unsatisfactory most likely due to the off-target-associated side effects and low localized retention of the used therapeutic agents in the infarcted myocardium, which constitutes a substantial barrier for the effective treatment of MI. Injectable hydrogels are regarded as a minimally invasive technology that can overcome the clinical and surgical limitations of traditional stenting by a modulated sol-gel transition and localized transport of a variety of encapsulated cargoes, leading to enhanced therapeutic efficiency and improved patient comfort and compliance. However, the design of injectable hydrogels for myocardial repair and the mechanism of action of bioactive substance-loaded hydrogels for MI repair remain unclear. To elucidate these points, we summarized the recent progresses made on the use of injectable hydrogels for encapsulation of various therapeutic substances for MI treatment with an emphasis on the mechanism of action of hydrogel systems for myocardial repair. Specifically, the pathogenesis of MI and the rational design of injectable hydrogels for myocardial repair were presented. Next, the mechanisms of various biotherapeutic substance-loaded injectable hydrogels for myocardial repair was discussed. Finally, the potential challenges and future prospects for the use of injectable hydrogels for MI treatment were proposed for the purpose of drawing theoretical guidance on the development of novel therapeutic strategies for efficient treatment of MI.
Collapse
|
14
|
Yang Q, Fang J, Lei Z, Sluijter JPG, Schiffelers R. Repairing the heart: State-of the art delivery strategies for biological therapeutics. Adv Drug Deliv Rev 2020; 160:1-18. [PMID: 33039498 DOI: 10.1016/j.addr.2020.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/23/2022]
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality worldwide. It is caused by an acute imbalance between oxygen supply and demand in the myocardium, usually caused by an obstruction in the coronary arteries. The conventional therapy is based on the application of (a combination of) anti-thrombotics, reperfusion strategies to open the occluded artery, stents and bypass surgery. However, numerous patients cannot fully recover after these interventions. In this context, new therapeutic methods are explored. Three decades ago, the first biologicals were tested to improve cardiac regeneration. Angiogenic proteins gained popularity as potential therapeutics. This is not straightforward as proteins are delicate molecules that in order to have a reasonably long time of activity need to be stabilized and released in a controlled fashion requiring advanced delivery systems. To ensure long-term expression, DNA vectors-encoding for therapeutic proteins have been developed. Here, the nuclear membrane proved to be a formidable barrier for efficient expression. Moreover, the development of delivery systems that can ensure entry in the target cell, and also correct intracellular trafficking towards the nucleus are essential. The recent introduction of mRNA as a therapeutic entity has provided an attractive intermediate: prolonged but transient expression from a cytoplasmic site of action. However, protection of the sensitive mRNA and correct delivery within the cell remains a challenge. This review focuses on the application of synthetic delivery systems that target the myocardium to stimulate cardiac repair using proteins, DNA or RNA.
Collapse
Affiliation(s)
- Qiangbing Yang
- Division LAB, CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Juntao Fang
- Division Heart & Lungs, Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Zhiyong Lei
- Division LAB, CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands; Division Heart & Lungs, Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division Heart & Lungs, Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands; Regenerative Medicine Utrecht, Circulatory Health Laboratory, Utrecht University, Utrecht, the Netherlands
| | - Raymond Schiffelers
- Division LAB, CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
15
|
Mikrani R, Li C, Naveed M, Li C, Baig MMFA, Zhang Q, Wang Y, Peng J, Zhao L, Zhou X. Pharmacokinetic Advantage of ASD Device Promote Drug Absorption through the Epicardium. Pharm Res 2020; 37:173. [DOI: 10.1007/s11095-020-02898-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/28/2020] [Indexed: 01/03/2023]
|
16
|
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: 69] [Impact Index Per Article: 17.3] [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
|
17
|
|
18
|
Liao X, Yang X, Deng H, Hao Y, Mao L, Zhang R, Liao W, Yuan M. Injectable Hydrogel-Based Nanocomposites for Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 8:251. [PMID: 32296694 PMCID: PMC7136457 DOI: 10.3389/fbioe.2020.00251] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs), including a series of pathological disorders, severely affect millions of people all over the world. To address this issue, several potential therapies have been developed for treating CVDs, including injectable hydrogels as a minimally invasive method. However, the utilization of injectable hydrogel is a bit restricted recently owing to some limitations, such as transporting the therapeutic agent more accurately to the target site and prolonging their retention locally. This review focuses on the advances in injectable hydrogels for CVD, detailing the types of injectable hydrogels (natural or synthetic), especially that complexed with stem cells, cytokines, nano-chemical particles, exosomes, genetic material including DNA or RNA, etc. Moreover, we summarized the mainly prominent mechanism, based on which injectable hydrogel present excellent treating effect of cardiovascular repair. All in all, it is hopefully that injectable hydrogel-based nanocomposites would be a potential candidate through cardiac repair in CVDs treatment.
Collapse
Affiliation(s)
- Xiaoshan Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xushan Yang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hong Deng
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lianzhi Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Rongjun Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
19
|
Wen Y, Li XY, Li ZY, Wang ML, Chen PP, Liu Y, Zhang XZ, Jiang XJ. Intra-myocardial Delivery of a Novel Thermosensitive Hydrogel Inhibits Post-infarct Heart Failure After Degradation in Rat. J Cardiovasc Transl Res 2020; 13:677-685. [PMID: 32020504 DOI: 10.1007/s12265-019-09941-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
Whether intra-myocardial delivery of hydrogel can prevent post-infarct heart failure (HF) in a long follow-up period, especially after it is degraded, remains unclear. In this study, Dex-PCL-HEMA/PNIPAAm (DPHP) hydrogel was delivered into peri-infarct myocardium of rat when coronary artery was ligated, while PBS was employed as control. Twelve weeks later, compared with control, left ventricle remodeling was attenuated and cardiac function was preserved; serum brain natriuretic peptide, cardiac aldosterone, and pulmonary congestion were suppressed in hydrogel group. Pro-fibrogenic mRNA increased in infarct area while decreased in remote zone, as well as hypertrophic mRNA. These data proves DPHP hydrogel suppresses ventricular remodeling and HF by promoting fibrotic healing in infarct area and inhibiting reactive fibrosis and hypertrophy in remote zone. Timely intra-myocardial hydrogel implantation is an effective strategy to inhibit post-infarct cardiac remodeling and have a long-term beneficial effect even after it has been biodegraded.
Collapse
Affiliation(s)
- Ying Wen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Xiao-Yan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
| | - Ze-Yong Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Meng-Long Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
| | - Pan-Pan Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,People's Hospital of Fangcheng County, Nanyang, Henan, China
| | - Yang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Xue-Jun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.
| |
Collapse
|
20
|
Liao Q, Qu S, Tang LX, Li LP, He DF, Zeng CY, Wang WE. Irisin exerts a therapeutic effect against myocardial infarction via promoting angiogenesis. Acta Pharmacol Sin 2019; 40:1314-1321. [PMID: 31061533 DOI: 10.1038/s41401-019-0230-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/14/2019] [Indexed: 02/08/2023] Open
Abstract
Irisin, a myokine, is cleaved from the extracellular portion of fibronectin domain-containing 5 protein in skeletal muscle and myocardium and secreted into circulation as a hormone during exercise. Irisin has been found to exert protective effects against lung and heart injuries. However, whether irisin influences myocardial infarction (MI) remains unclear. In this study we investigated the therapeutic effects of irisin in an acute MI model and its underlying mechanisms. Adult C57BL/6 mice were subjected to ligation of the left anterior descending coronary artery and treated with irisin for 2 weeks after MI. Cardiac function was assessed using echocardiography. We found that irisin administration significantly alleviated MI-induced cardiac dysfunction and ventricular dilation at 4 weeks post-MI. Irisin significantly reduced infarct size and fibrosis in post-MI hearts. Irisin administration significantly increased angiogenesis in the infarct border zone and decreased cardiomyocyte apoptosis, but did not influence cardiomyocyte proliferation. In human umbilical vein endothelial cells (HUVEC), irisin significantly increased the phosphorylation of ERK, and promoted the migration of HUVEC detected in wound-healing and transwell chamber migration assay. The effects of irisin were blocked by the ERK inhibitor U0126. In conclusion, irisin improves cardiac function and reduces infarct size in post-MI mouse heart. The therapeutic effect is associated with its pro-angiogenic function through activating ERK signaling pathway.
Collapse
|
21
|
Nam K, Dean SM, Brown CT, Smith RJ, Lei P, Andreadis ST, Baker OJ. Synergistic effects of laminin-1 peptides, VEGF and FGF9 on salivary gland regeneration. Acta Biomater 2019; 91:186-194. [PMID: 31028910 DOI: 10.1016/j.actbio.2019.04.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/25/2019] [Accepted: 04/23/2019] [Indexed: 01/19/2023]
Abstract
Hyposalivation is associated with radiation therapy, Sjögren's syndrome and/or aging, and is a significant clinical problem that decreases oral health and overall health in many patients and currently lacks effective treatment. Hence, methods to regenerate salivary glands and restore saliva secretion are urgently needed. To this end, this study describes the modification of fibrin hydrogels with a combination of laminin-1 peptides (YIGSR and A99) and human growth factors (vascular endothelial growth factor and fibroblast growth factor 9) to enhance regeneration in a salivary gland injury mouse model. Our results indicate that these fortified hydrogels enhanced angiogenesis and neurogenesis while promoting formation of acinar structures, thereby leading to enhanced saliva secretion. Such functional recovery indicates salivary gland regeneration and suggests that our technology may be useful in promoting gland regeneration and reversing hyposalivation in a clinical setting. STATEMENT OF SIGNIFICANCE: We engineered Fibrin Hydrogels (FH) to contain multiple regenerative cues including laminin-1 peptides (L1p) and growth factors (GFs). L1p and GF modified FH were used to induce salivary gland regeneration in a wounded mouse model. Treatment with L1p and GF modified FH promoted salivary epithelial tissue regeneration, vascularization, neurogenesis and healing as compared to L1p-FH or FH alone. Results indicate that L1p and GF modified FH can be used for future therapeutic applications.
Collapse
Affiliation(s)
- Kihoon Nam
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States
| | - Spencer M Dean
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States
| | - Callie T Brown
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States
| | - Randall J Smith
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States
| | - Pedro Lei
- Department of Chemical and Biological Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States; Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States; Center of Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States.
| | - Olga J Baker
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States.
| |
Collapse
|
22
|
Kuraitis D, Hosoyama K, Blackburn NJR, Deng C, Zhong Z, Suuronen EJ. Functionalization of soft materials for cardiac repair and regeneration. Crit Rev Biotechnol 2019; 39:451-468. [PMID: 30929528 DOI: 10.1080/07388551.2019.1572587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Coronary artery disease is a leading cause of death in developed nations. As the disease progresses, myocardial infarction can occur leaving areas of dead tissue in the heart. To compensate, the body initiates its own repair/regenerative response in an attempt to restore function to the heart. These efforts serve as inspiration to researchers who attempt to capitalize on the natural regenerative processes to further augment repair. Thus far, researchers are exploiting these repair mechanisms in the functionalization of soft materials using a variety of growth factor-, ligand- and peptide-incorporating approaches. The goal of functionalizing soft materials is to best promote and direct the regenerative responses that are needed to restore the heart. This review summarizes the opportunities for the use of functionalized soft materials for cardiac repair and regeneration, and some of the different strategies being developed.
Collapse
Affiliation(s)
- Drew Kuraitis
- a Division of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , Canada
| | - Katsuhiro Hosoyama
- a Division of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , Canada
| | - Nick J R Blackburn
- a Division of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , Canada
| | - Chao Deng
- b Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , Suzhou , People's Republic of China
| | - Zhiyuan Zhong
- b Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , Suzhou , People's Republic of China
| | - Erik J Suuronen
- a Division of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , Canada
| |
Collapse
|
23
|
Johnson T, Zhao L, Manuel G, Taylor H, Liu D. Approaches to therapeutic angiogenesis for ischemic heart disease. J Mol Med (Berl) 2018; 97:141-151. [PMID: 30554258 DOI: 10.1007/s00109-018-1729-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022]
Abstract
Ischemic heart disease (IHD) is caused by the narrowing of arteries that work to provide blood, nutrients, and oxygen to the myocardial tissue. The worldwide epidemic of IHD urgently requires innovative treatments despite the significant advances in medical, interventional, and surgical therapies for this disease. Angiogenesis is a physiological and pathophysiological process that initiates vascular growth from pre-existing blood vessels in response to a lack of oxygen. This process occurs naturally over time and has encouraged researchers and clinicians to investigate the outcomes of accelerating or enhancing this angiogenic response as an alternative IHD therapy. Therapeutic angiogenesis has been shown to revascularize ischemic heart tissue, reduce the progression of tissue infarction, and evade the need for invasive surgical procedures or tissue/organ transplants. Several approaches, including the use of proteins, genes, stem/progenitor cells, and various combinations, have been employed to promote angiogenesis. While clinical trials for these approaches are ongoing, microvesicles and exosomes have recently been investigated as a cell-free approach to stimulate angiogenesis and may circumvent limitations of using viable cells. This review summarizes the approaches to accomplish therapeutic angiogenesis for IHD by highlighting the advances and challenges that addresses the applicability of a potential pro-angiogenic medicine.
Collapse
Affiliation(s)
- Takerra Johnson
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Lina Zhao
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Gygeria Manuel
- Department of Biochemistry, Spelman College, 350 Spelman Lane, Atlanta, GA, 30314, USA
| | - Herman Taylor
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Dong Liu
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| |
Collapse
|
24
|
Zhang Y, Xu Y, Wang L, Chen Y, Tian R, Jiao J, Xie H, Yang L, Gao F. Quantitative assessment of salvaged myocardial zone and intramyocardial hemorrhage using non-contrast faster T2 mapping in a rat model by 7T MRI. Exp Ther Med 2017; 14:3425-3432. [PMID: 29042929 PMCID: PMC5639411 DOI: 10.3892/etm.2017.4967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
The aim of this study was to determine the myocardial area at risk (AAR), infarction-core size (IS) and the salvaged myocardial zone (SMZ), and to evaluate the imaging and histological characteristics of intramyocardial hemorrhage (IMH) after myocardial infarction using non-contrast T2 mapping on 7T magnetic resonance imaging (MRI). Twenty Sprague Dawley (SD) rats were randomly divided into the sham and model groups (n=10 in each). In the model group, myocardial infarction models were established by left anterior descending branch ligation. After 24 h, all animals were imaged on a 7.0 Tesla system with cine spiral imaging, T2 mapping with late gadolinium enhancement (LGE). The rats were then sacrificed for measurement of the IS and AAR using 2,3,5-triphenylterazolium chloride (TTC) and hematoxylin and eosin (H&E) staining. T2 mapping revealed that the AAR in the model group was significantly higher than that in the sham group. No remarkable T2 value was noted in the entire heart of the sham group. LGE and TTC staining demonstrated similar IS. T2 mapping and H&E staining revealed a similar AAR as well. T2 mapping characterized the IMH as a phenomenon resulting from the area of hypointensity in the hyperintensity involving the infarct-core zone and corresponding T2 value 928.6±1.52 msec with IMH vs. 35.8±2.61 msec without IMH; n=3 with 18 slices; P=0.032). In conclusion, non-contrast T2 mapping was a reliable approach to quantitatively evaluate the SMZ and IMH.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China.,Department of Radiology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Yini Xu
- The Key Laboratory of Optional Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University Town, Guiyang, Guizhou 550025, P.R. China
| | - Lei Wang
- Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yushu Chen
- Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ruiqing Tian
- Department of Oncology, The First People's Hospital of Guiyang, Guiyang, Guizhou 550002, P.R. China
| | - Jun Jiao
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Hong Xie
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Li Yang
- Department of Radiology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Fabao Gao
- Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
25
|
Guo M, Shi JH, Wang PL, Shi DZ. Angiogenic Growth Factors for Coronary Artery Disease: Current Status and Prospects. J Cardiovasc Pharmacol Ther 2017; 23:130-141. [PMID: 29025278 DOI: 10.1177/1074248417735399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ming Guo
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun-He Shi
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Pei-Li Wang
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Da-Zhuo Shi
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
26
|
Zhu H, Li X, Yuan M, Wan W, Hu M, Wang X, Jiang X. Intramyocardial delivery of bFGF with a biodegradable and thermosensitive hydrogel improves angiogenesis and cardio-protection in infarcted myocardium. Exp Ther Med 2017; 14:3609-3615. [PMID: 29042955 PMCID: PMC5639332 DOI: 10.3892/etm.2017.5015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 03/17/2017] [Indexed: 11/30/2022] Open
Abstract
Basic fibroblast growth factor (bFGF), a known angiogenic factor, may provide a potential strategy for the treatment of myocardial infarction (MI), but it is limited by a relatively short half-life. Dex-PCL-HEMA/PNIPAAm hydrogel provides a reservoir for the controlled release of growth factors. The aim of the current study was to evaluate the effects of bFGF incorporated into a Dex-PCL-HEMA/PNIPAAm hydrogel on angiogenesis and cardiac health in a rat model of acute MI, induced by coronary artery ligation. Phosphate-buffered solution (PBS group), Dex-PCL-HEMA/PNIPAAm hydrogel (Gel group), bFGF in phosphate-buffered solution (bFGF group) or bFGF in hydrogel (Gel + bFGF group) was injected into a peri-infarcted area of cardiac tissue immediately following MI. On day 30 post-surgery, cardiac function was assessed by echocardiography, apoptosis index by terminal deoxynucleotidyl transferase dUTP nick-end labeling assessment and vascular development by immunohistochemical staining. The findings demonstrated that injection of bFGF along with hydrogel induced angiogenesis, reduced collagen content, MI area and cell apoptosis and improved cardiac function compared with the injection of either bFGF or hydrogel alone. bFGF incorporated with Dex-PCL-HEMA/PNIPAAm hydrogel injection induces angiogenesis, attenuates cardiac remodeling and improves cardiac function following MI.
Collapse
Affiliation(s)
- Hongling Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mingjie Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Miaoyang Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoding Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
27
|
MacArthur JW, Steele AN, Goldstone AB, Cohen JE, Hiesinger W, Woo YJ. Injectable Bioengineered Hydrogel Therapy in the Treatment of Ischemic Cardiomyopathy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2017; 19:30. [PMID: 28337717 DOI: 10.1007/s11936-017-0530-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OPINION STATEMENT Over the past two decades, the field of cardiovascular medicine has seen the rapid development of multiple different modalities for the treatment of ischemic myocardial disease. Most research efforts have focused on strategies aimed at coronary revascularization, with significant technological advances made in percutaneous coronary interventions as well as coronary artery bypass graft surgery. However, recent research efforts have shifted towards ways to address the downstream effects of myocardial infarction on both cellular and molecular levels. To this end, the broad application of injectable hydrogel therapy after myocardial infarction has stimulated tremendous interest. In this article, we will review what hydrogels are, how they can be bioengineered in unique ways to optimize therapeutic potential, and how they can be used as part of a treatment strategy after myocardial infarction.
Collapse
Affiliation(s)
- John W MacArthur
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Jeffrey E Cohen
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Falk Cardiovascular Research Bldg, 2nd Floor, 300 Pasteur Drive, Stanford, CA, 94305-5407, USA.
| |
Collapse
|
28
|
Maslov M, Foianini S, Lovich M. Delivery of drugs, growth factors, genes and stem cells via intrapericardial, epicardial and intramyocardial routes for sustained local targeted therapy of myocardial disease. Expert Opin Drug Deliv 2017; 14:1227-1239. [PMID: 28276968 DOI: 10.1080/17425247.2017.1292249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Local myocardial delivery (LMD) of therapeutic agents is a promising strategy that aims to treat various myocardial pathologies. It is designed to deliver agents directly to the myocardium and minimize their extracardiac concentrations and side effects. LMD aims to enhance outcomes of existing therapies by broadening their therapeutic window and to utilize new agents that could not be otherwise be implemented systemically. Areas covered: This article provides a historical overview of six decades LMD evolution in terms of the approaches, including intrapericardial, epicardial, and intramyocardial delivery, and the wide array of classes of agents used to treat myocardial pathologies. We examines delivery of pharmaceutical compounds, targeted gene transfection and cell implantation techniques to produce therapeutic effects locally. We outline therapeutic indications, successes and failures as well as technical approaches for LMD. Expert opinion: While LMD is more complicated than conventional oral or intravenous administration, given recent advances in interventional cardiology, it is safe and may provide better therapeutic outcomes. LMD is complex as many factors impact pharmacokinetics and biologic result. The choice between routes of LMD is largely driven not only by the myocardial pathology but also by the nature and physicochemical properties of the therapeutic agents.
Collapse
Affiliation(s)
- Mikhail Maslov
- a Department of Anesthesiology, Pain Medicine and Critical Care , Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston , MA , USA
| | - Stephan Foianini
- a Department of Anesthesiology, Pain Medicine and Critical Care , Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston , MA , USA
| | - Mark Lovich
- a Department of Anesthesiology, Pain Medicine and Critical Care , Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
29
|
Chen G, Xu C, Cen M. RETRACTED ARTICLE: TIEG1 suppression enhances the therapeutic efficacy of human adipose-derived mesenchymal stem cells in myocardial infarct repair. Heart Vessels 2016; 31:2080. [PMID: 27480878 DOI: 10.1007/s00380-016-0878-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 07/22/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Guofan Chen
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Changfu Xu
- Department of Cardiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Mingqiu Cen
- Department of Cardiology, Xixi Hospital of Hangzhou, No. 2, Hengfu Road, Hangzhou, 310023, Zhejiang, China.
| |
Collapse
|
30
|
Spath NB, Mills NL, Cruden NL. Novel cardioprotective and regenerative therapies in acute myocardial infarction: a review of recent and ongoing clinical trials. Future Cardiol 2016; 12:655-672. [PMID: 27791385 PMCID: PMC5985502 DOI: 10.2217/fca-2016-0044] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022] Open
Abstract
Following the original large-scale randomized trials of aspirin and β-blockade, there have been a number of major advances in pharmacological and mechanical treatments for acute myocardial infarction. Despite this progress, myocardial infarction remains a major global cause of mortality and morbidity, driving a quest for novel treatments in this area. As the understanding of mitochondrial dynamics and the pathophysiology of reperfusion injury has evolved, the last three decades have seen advances in ischemic conditioning, pharmacological and metabolic cardioprotection, as well as biological and stem-cell therapies. The aim of this review is to provide a synopsis of adjunctive cardioprotective and regenerative therapies currently undergoing or entering early clinical trials in the treatment of patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Nicholas B Spath
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nicholas L Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
31
|
Zhang W, Huo Y, Wang X, Jia Y, Su L, Wang C, Li Y, Yang Y, Liu Y. Decellularized ovine arteries as biomatrix scaffold support endothelial of mesenchymal stem cells. Heart Vessels 2016; 31:1874-1881. [DOI: 10.1007/s00380-016-0834-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/08/2016] [Indexed: 12/20/2022]
|
32
|
Increased mortality and aggravation of heart failure in liver X receptor-α knockout mice after myocardial infarction. Heart Vessels 2016; 31:1370-9. [PMID: 26753692 DOI: 10.1007/s00380-015-0781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/02/2015] [Indexed: 12/23/2022]
Abstract
Liver X receptors, LXRα (NR1H3) and LXRβ (NR1H2), are best known as nuclear oxysterol receptors and physiological master regulators of lipid and cholesterol metabolism. LXRα play a protective role in acute myocardial ischemia/reperfusion (MI/R) injury, but its role in myocardial infarction (MI) is unknown. The present study was undertaken to determine the effect of LXRα knockout on survival and development of chronic heart failure after MI. Wild-type (WT) and LXRα(-/-) mice were subjected to MI followed by serial echocardiographic and histological assessments. Greater myocyte apoptosis and inflammation within the infarcted zones were found in LXRα(-/-) group at 3 days after MI. At 4 weeks post-MI, LXRα(-/-) MI murine hearts demonstrated significantly increased infarct size, reduced ejection fraction (LXRα(-/-) 29.4 % versus WT 34.4 %), aggravated left ventricular (LV) chamber dilation, enhanced fibrosis and reduced angiogenesis. In addition, LXRα(-/-) mice had increased mortality compared with WT mice. LXRα deficiency increases mortality, aggravates pathological injury and LV remodeling induced by MI. Drugs specifically targeting LXRα may be promising in the treatment of MI.
Collapse
|
33
|
Physical inactivity increases endostatin and osteopontin in patients with coronary artery disease. Heart Vessels 2015; 31:1603-8. [PMID: 26661073 DOI: 10.1007/s00380-015-0778-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022]
Abstract
The balance between the angiostatic factor endostatin (ES) and angiogenic factor osteopontin (OPN) is essential in physiological and pathological angiogenesis. Several circumstances might influence this equilibrium and are of distinct interest when investigating mechanisms in coronary artery disease (CAD). The present explorative cross-sectional study was to investigate the influence of physical inactivity on ES and OPN levels in 181 male and 71 female patients with angiographycally verified CAD. Anamnestic and laboratory data were collected; ES was measured in serum and OPN in plasma by ELISA. Univariate analysis of variance was used to test for the influence of physical activity on ES and OPN levels and age, BMI, sex and diabetes status were included as covariates. ES and OPN intercorrelated significantly (r = 0.42; p < 0.001). ES and OPN decreased significantly in response to increasing activity level of patients suffering CAD (F = 5.5; p < 0.001 and F = 3.6; p < 0.01 resp.). This study is the first to show a linear decrease in ES and OPN levels in CAD patients depending on the degree of physical activity undergone. Lower levels of ES and OPN in physically active patients might be a sign of increased angiogenesis and decreased inflammation and calcifying activity and therefore contribute to the understanding of the damaging effect of physical inactivity in cardiovascular disease.
Collapse
|