1
|
Zhang B, Wang C, Guo M, Zhu F, Yu Z, Zhang W, Li W, Zhang Y, Tian W. Circadian Rhythm-Dependent Therapy by Composite Targeted Polyphenol Nanoparticles for Myocardial Ischemia-Reperfusion Injury. ACS NANO 2024; 18:28154-28169. [PMID: 39373010 DOI: 10.1021/acsnano.4c07690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Myocardial ischemia-reperfusion (IR) injury is a severe rhythmic disease with a high prevalence in the early morning. IR injury has a significant circadian rhythm in reactive oxygen species (ROS) and inflammation levels. The development of rhythmic drugs has become a priority in myocardial IR injury. In this study, resveratrol (RES) and proanthocyanidins (OPC) were utilized to design nanoparticles (NPs), with hyaluronic acid (HA) as the core, grafted with MMP-targeting peptides to improve delivery to injured myocardial regions (HA-RES-OPC-MMP NPs). NPs significantly scavenged ROS, attenuated inflammation, and activated the rhythm gene. Notably, the difference in therapeutic effects on myocardial IR injury in mice at Zeitgeber time (ZT)1 and ZT13 confirms that NPs are rhythm-dependent drugs. At ZT13, echocardiographic and MRI confirm that IR injury in mice was not as severe as at ZT1, yet NPs were also less effective in treatment. Further, Per1/2 knockout mice confirmed the rhythm-dependent treatment of myocardial IR injury by NPs. Molecular studies have shown that rhythmic characteristics of inflammation and Sirt1 transcript levels are the main reasons for the different rhythmic therapeutic effects of NPs. Circadian rhythm-dependent treatment of HA-RES-OPC-MMP NPs has excellent potential for more precise treatment of myocardial IR injury in the future.
Collapse
Affiliation(s)
- Bosong Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Cao Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Mingyue Guo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Fuxing Zhu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Zhenqiang Yu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Wenxiang Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Wenyu Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yijian Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Weiming Tian
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
2
|
Carvalho T, Bártolo R, Correia A, Vilela C, Wang S, Santos HA, Freire CSR. Implantable Patch of Oxidized Nanofibrillated Cellulose and Lysozyme Amyloid Nanofibrils for the Regeneration of Infarcted Myocardium Tissue and Local Delivery of RNA-Loaded Nanoparticles. Macromol Rapid Commun 2024; 45:e2400129. [PMID: 38778746 DOI: 10.1002/marc.202400129] [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: 03/05/2024] [Revised: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Biopolymeric implantable patches are popular scaffolds for myocardial regeneration applications. Besides being biocompatible, they can be tailored to have required properties and functionalities for this application. Recently, fibrillar biobased nanostructures prove to be valuable in the development of functional biomaterials for tissue regeneration applications. Here, periodate-oxidized nanofibrillated cellulose (OxNFC) is blended with lysozyme amyloid nanofibrils (LNFs) to prepare a self-crosslinkable patch for myocardial implantation. The OxNFC:LNFs patch shows superior wet mechanical properties (60 MPa for Young's modulus and 1.5 MPa for tensile stress at tensile strength), antioxidant activity (70% scavenging activity under 24 h), and bioresorbability ratio (80% under 91 days), when compared to the patches composed solely of NFC or OxNFC. These improvements are achieved while preserving the morphology, required thermal stability for sterilization, and biocompatibility toward rat cardiomyoblast cells. Additionally, both OxNFC and OxNFC:LNFs patches reveal the ability to act as efficient vehicles to deliver spermine modified acetalated dextran nanoparticles, loaded with small interfering RNA, with 80% of delivery after 5 days. This study highlights the value of simply blending OxNFC and LNFs, synergistically combining their key properties and functionalities, resulting in a biopolymeric patch that comprises valuable characteristics for myocardial regeneration applications.
Collapse
Affiliation(s)
- Tiago Carvalho
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, Aveiro, 3810-193, Portugal
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Raquel Bártolo
- Department of Biomaterials and Biomedical Technology, PRECISION - Personalized medicine Research Institute, University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Carla Vilela
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, Aveiro, 3810-193, Portugal
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomaterials and Biomedical Technology, PRECISION - Personalized medicine Research Institute, University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Carmen S R Freire
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
3
|
Chabronova A, Holmes TL, Hoang DM, Denning C, James V, Smith JGW, Peffers MJ. SnoRNAs in cardiovascular development, function, and disease. Trends Mol Med 2024; 30:562-578. [PMID: 38523014 DOI: 10.1016/j.molmed.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
Small nucleolar RNAs (snoRNAs) are emerging as important regulators of cardiovascular (patho)biology. Several roles of snoRNAs have recently been identified in heart development and congenital heart diseases, as well as their dynamic regulation in hypertrophic and dilated cardiomyopathies, coronary heart disease (CHD), myocardial infarction (MI), cardiac fibrosis, and heart failure. Furthermore, reports of changes in vesicular snoRNA expression and altered levels of circulating snoRNAs in response to cardiac stress suggest that snoRNAs also function in cardiac signaling and intercellular communication. In this review, we summarize and discuss key findings and outline the clinical potential of snoRNAs considering current challenges and gaps in the field of cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Alzbeta Chabronova
- Department of Musculoskeletal Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK.
| | - Terri L Holmes
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Duc M Hoang
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Chris Denning
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Victoria James
- School of Veterinary Medicine and Science, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - James G W Smith
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Mandy J Peffers
- Department of Musculoskeletal Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| |
Collapse
|
4
|
Tripathi S, Rani K, Raj VS, Ambasta RK. Drug repurposing: A multi targetted approach to treat cardiac disease from existing classical drugs to modern drug discovery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:151-192. [PMID: 38942536 DOI: 10.1016/bs.pmbts.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) are characterized by abnormalities in the heart, blood vessels, and blood flow. CVDs comprise a diverse set of health issues. There are several types of CVDs like stroke, endothelial dysfunction, thrombosis, atherosclerosis, plaque instability and heart failure. Identification of a new drug for heart disease takes longer duration and its safety efficacy test takes even longer duration of research and approval. This chapter explores drug repurposing, nano-therapy, and plant-based treatments for managing CVDs from existing drugs which saves time and safety issues with testing new drugs. Existing drugs like statins, ACE inhibitor, warfarin, beta blockers, aspirin and metformin have been found to be useful in treating cardiac disease. For better drug delivery, nano therapy is opening new avenues for cardiac research by targeting interleukin (IL), TNF and other proteins by proteome interactome analysis. Nanoparticles enable precise delivery to atherosclerotic plaques, inflammation areas, and damaged cardiac tissues. Advancements in nano therapeutic agents, such as drug-eluting stents and drug-loaded nanoparticles are transforming CVDs management. Plant-based treatments, containing phytochemicals from Botanical sources, have potential cardiovascular benefits. These phytochemicals can mitigate risk factors associated with CVDs. The integration of these strategies opens new avenues for personalized, effective, and minimally invasive cardiovascular care. Altogether, traditional drugs, phytochemicals along with nanoparticles can revolutionize the future cardiac health care by identifying their signaling pathway, mechanism and interactome analysis.
Collapse
Affiliation(s)
- Shyam Tripathi
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - Kusum Rani
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - V Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India.
| | - Rashmi K Ambasta
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India.
| |
Collapse
|
5
|
Jing J, Fang S, Li Y, Liu W, Wang C, Lan Y, Wang Y, Yang C. An enhanced cardio-protective effect of nanoparticles loaded with active components from Polygonum orientale L. against isoproterenol-induced myocardial ischemia in rats. Int J Pharm 2024; 655:124047. [PMID: 38531434 DOI: 10.1016/j.ijpharm.2024.124047] [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: 11/07/2023] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
In this study, nanoparticles loaded with active components from Polygonum orientale L. (PO), a traditional Chinese herb known for its anti-myocardial ischemic properties, were investigated for cardio-protective properties. Specifically, OVQ-Nanoparticles (OVQ-NPs) with Orientin (Ori), Vitexin (Vit), and Quercetin (Que) was obtained by double emulsion-solvent evaporation method. The OVQ-NPs exhibited a spherical shape, with a uniform size distribution of 136.77 ± 3.88 nm and a stable ζ-potential of -13.40 ± 2.24 mV. Notably, these nanoparticles exhibited a favorable sustained-release characteristic, resulting in an extended circulation time within the living organism. Consequently, the administration of these nanoparticles resulted in significant improvements in electrocardiograms and heart mass index of myocardial ischemic rats induced by isoproterenol, as well as decreased serum levels of CK, LDH, and AST. Furthermore, the results of histopathological examination, such as H&E staining and TUNEL staining, confirmed a reduced level of cardiac tissue pathology and apoptosis. Moreover, the quantification of biochemical indicators (SOD, MDA, GSH, NO, TNF-α, and IL-6) demonstrated that OVQ-NPs effectively mitigated myocardial ischemia by regulating oxidative stress and inflammatory pathways. In conclusion, OVQ-NPs demonstrate promising therapeutic potential as an intervention for myocardial ischemia, providing a new perspective on traditional Chinese medicine treatment in this area.
Collapse
Affiliation(s)
- Jincheng Jing
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; School of Pharmacy, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Shumei Fang
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; School of Pharmacy, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Yueting Li
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Wenting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; School of Pharmacy, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Cong Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; School of Pharmacy, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Yanyu Lan
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Yonglin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, Guizhou, China.
| | - Chang Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants/Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, Guizhou, China; Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, Guizhou, China.
| |
Collapse
|
6
|
Jumppanen M, Kinnunen SM, Zore M, Välimäki MJ, Talman V, Gennäs GBA, Ruskoaho HJ, Yli-Kauhaluoma J. Affinity chromatography reveals direct binding of the GATA4-NKX2-5 interaction inhibitor (3i-1000) with GATA4. Sci Rep 2024; 14:8938. [PMID: 38637629 PMCID: PMC11026519 DOI: 10.1038/s41598-024-59418-4] [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: 10/17/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Heart failure is a serious medical condition with a poor prognosis. Current treatments can only help manage the symptoms and slow the progression of heart failure. However, there is currently no cure to prevent and reverse cardiac remodeling. Transcription factors are in a central role in various cellular processes, and in the heart, GATA4 and NKX2-5 transcription factors mediate hypertrophic responses and remodeling. We have identified compounds that modulate the synergistic interaction of GATA4 and NKX2-5 and shown that the most promising compound (1, 3i-1000) is cardioprotective in vitro and in vivo. However, direct evidence of its binding site and mechanism of action has not been available. Due to the disordered nature of transcription factors, classical target engagement approaches cannot be utilized. Here, we synthesized a small-molecule ligand-binding pulldown probe of compound 1 to utilize affinity chromatography alongside CETSA, AlphaScreen, and molecular modeling to study ligand binding. These results provide the first evidence of direct physical binding of compound 1 selectively to GATA4. While developing drugs that target transcription factors presents challenges, advances in technologies and knowledge of intrinsically disordered proteins enable the identification of small molecules that can selectively target transcription factors.
Collapse
Affiliation(s)
- Mikael Jumppanen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Matej Zore
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Gustav Boije Af Gennäs
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Heikki J Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland.
| |
Collapse
|
7
|
Chen Y, Zhang J. A bibliography of smart nanomaterials biological application in myocardial infarction research. Medicine (Baltimore) 2024; 103:e37672. [PMID: 38579096 PMCID: PMC10994481 DOI: 10.1097/md.0000000000037672] [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] [Received: 08/30/2023] [Accepted: 02/29/2024] [Indexed: 04/07/2024] Open
Abstract
Myocardial infarction has been considered the top cause of mortality globally. Numerous studies investigated the biological application of smart nanomaterials in myocardial infarction. Our study aimed to provide an overview of this area through bibliography research. Literature related to the biological application of nanomaterials was retrieved from the web of science core collection database. Bibliography analysis was performed using Microsoft Excel, VOSviewer, Citespace, and the R package "bibliometrix." A total of 1226 publications were included. The USA, China, and India carried out the most of studies. Harvard University is the most productive institution. Matthias Nahrendorf ranked first in article volume and also owned the highest impact. Keyword burst analysis indicated the frontiers and hotspots to be gold nanoparticles and iron oxide nanoparticles. This bibliography analysis provides a comprehensive overview of uncovered current research trends and emerging hotspots of nanomaterials' biological application in myocardial infarction, thus inspiring further investigations.
Collapse
Affiliation(s)
- Yi Chen
- Department of Emergency Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Institute of Disaster Medicine, Sichuan University, Nursing Key Laboratory of Sichuan Province, Chengdu, China
| | - Jianna Zhang
- Department of Emergency Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Institute of Disaster Medicine, Sichuan University, Nursing Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
8
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [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: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
9
|
Hu N, Sun M, Lv N, Gao Y, Fu X, Xing D, Guo X, Zhai S, Zhang R. ROS-Suppression Nanoplatform Combined Activation of STAT3/Bcl-2 Pathway for Preventing Myocardial Infarction in Mice. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12188-12201. [PMID: 38288981 DOI: 10.1021/acsami.3c16735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2024]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide. The most effective way to treat myocardial infarction is to rescue ischemic cardiomyocytes. After an ischemic event, the overproduction of reactive oxygen species (ROS) is a key driver of myocardial injury. The produced ROS affects mitochondrial function and induces apoptosis in cardiomyocytes. This was accomplished by constructing platelet-membrane-encapsulated ROS-responsive drug-releasing nanoparticles (PMN@NIC-MalNPs) to deliver malonate and niclosamide (NIC). The results revealed that PMN@NIC-MalNPs degraded and released malonate and niclosamide in a high-level ROS microenvironment, effectively reducing the oxidative stress and apoptosis rate. By enhancing basal mitochondrial oxygen consumption rate (OCR), adenosine triphosphate (ATP) production, and spare respiratory capacity (SRC) in vitro, reduced the oxidative stress levels and restored mitochondrial function. In vivo studies revealed that the PMN@NIC-MalNPs improved cardiac dysfunction, inhibited succinate dehydrogenase (SDH) activity, increased ATP production, and reduced the myocardial infarct size in myocardial infarction model mice. Further, transcriptome analysis and Western blot revealed that PMN@NIC-MalNPs prevented apoptosis by activating the expressions of the signal transducer and activator of transcription 3 (STAT3) and Bcl-2, and inhibiting the expression of Bax. Thus, this study provides a novel therapeutic solution for treating myocardial infarction and predicting the viability of an antioxidant and antiapoptotic therapeutic solution in the treatment of myocardial injury.
Collapse
Affiliation(s)
- Nan Hu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
| | - Meng Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
- Cardiology Department, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030032, China
| | - Nan Lv
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030032, China
| | - Yangyang Gao
- The First Clinical Medical College, Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Xiaohong Fu
- The First Clinical Medical College, Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Dayi Xing
- The First Clinical Medical College, Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Xiang Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
| | - Shaodong Zhai
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
10
|
Wen H, Martínez MG, Happonen E, Qian J, Vallejo VG, Mendazona HJ, Jokivarsi K, Scaravilli M, Latonen L, Llop J, Lehto VP, Xu W. A PEG-assisted membrane coating to prepare biomimetic mesoporous silicon for PET/CT imaging of triple-negative breast cancer. Int J Pharm 2024; 652:123764. [PMID: 38176479 DOI: 10.1016/j.ijpharm.2023.123764] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 11/27/2023] [Accepted: 12/31/2023] [Indexed: 01/06/2024]
Abstract
Triple-negative breast cancer (TNBC) diagnosis remains challenging without expressing critical receptors. Cancer cell membrane (CCm) coating has been extensively studied for targeted cancer diagnostics due to attractive features such as good biocompatibility and homotypic tumor-targeting. However, the present study found that widely used CCm coating approaches, such as extrusion, were not applicable for functionalizing irregularly shaped nanoparticles (NPs), such as porous silicon (PSi). To tackle this challenge, we proposed a novel approach that employs polyethylene glycol (PEG)-assisted membrane coating, wherein PEG and CCm are respectively functionalized on PSi NPs through chemical conjugation and physical absorption. Meanwhile, the PSi NPs were grafted with the bisphosphonate (BP) molecules for radiolabeling. Thanks to the good chelating ability of BP and homotypic tumor targeting of cancer CCm coating, a novel PSi-based contrast agent (CCm-PEG-89Zr-BP-PSi) was developed for targeted positron emission tomography (PET)/computed tomography (CT) imaging of TNBC. The novel imaging agent showed good radiochemical purity (∼99 %) and stability (∼95 % in PBS and ∼99 % in cell medium after 48 h). Furthermore, the CCm-PEG-89Zr-BP-PSi NPs had efficient homotypic targeting ability in vitro and in vivo for TNBC. These findings demonstrate a versatile biomimetic coating method to prepare novel NPs for tumor-targeted diagnosis.
Collapse
Affiliation(s)
- Huang Wen
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland
| | - María Gómez Martínez
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain
| | - Emilia Happonen
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland
| | - Jing Qian
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland
| | - Vanessa Gómez Vallejo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain
| | - Helena Jorge Mendazona
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain
| | - Kimmo Jokivarsi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Mauro Scaravilli
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | - Leena Latonen
- School of Medicine, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland
| | - Jordi Llop
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain
| | - Vesa-Pekka Lehto
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland.
| | - Wujun Xu
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland.
| |
Collapse
|
11
|
Sun M, Hu N, Gao Y, Lv N, Fu X, Li Y, Zhai S, Zhang R. Platelet Membrane-Encapsulated Nanocomplexes Based on Profundity Scavenging ROS Strategy for Myocardial Infarction Therapy. Adv Healthc Mater 2024; 13:e2303101. [PMID: 38174837 DOI: 10.1002/adhm.202303101] [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: 09/15/2023] [Revised: 12/09/2023] [Indexed: 01/05/2024]
Abstract
Ischemia-induced myocardial injury has become a serious threat to human health, and its treatment remains a challenge. The occurrence of ischemic events leads to a burst release of reactive oxygen species (ROS), which triggers extensive oxidative damage and leads to dysfunctional autophagy, making it difficult for cells to maintain homeostasis. Antioxidants and modulation of autophagy have thus become promising strategies for the treatment of ischemic myocardial injury. This study proposes an antioxidant-activated autophagy therapeutic regimen based on combining melanin (Mel), an excellent antioxidant with metformin mimetic ploymetformin via electrostatic interactions, to obtain a nanocomplex (Met-Mel). The nanocomplex is finally encapsulated with platelet membranes (PMN) to construct a biomimetic nanoparticle (PMN@Met-Mel) capable of targeting injured myocardium. The prepared PMN@Met-Mel has good Mel loading capacity and optimal biosafety. It exhibits excellent antioxidant activity and autophagy activation, rapidly restoring mitochondrial function. Moreover, RNA sequencing (RNA-seq) analysis reveals that PMN@Met-Mel operates mechanistically by triggering the activation of the autophagy pathway. Subsequent in vivo experiments showcase promising cardioprotective effects of these nanoparticles. These discoveries present a newly devised nanoplatform with promising potential for the effective treatment of myocardial infarction.
Collapse
Affiliation(s)
- Meng Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Department of Cardiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Nan Hu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Yangyang Gao
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Nan Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaohong Fu
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Yafeng Li
- The Nephrology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Shaodong Zhai
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
12
|
Peng Y, Yang Z, Sun H, Li J, Lan X, Liu S. Nanomaterials in Medicine: Understanding Cellular Uptake, Localization, and Retention for Enhanced Disease Diagnosis and Therapy. Aging Dis 2024:AD.2024.0206-1. [PMID: 38421835 DOI: 10.14336/ad.2024.0206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Nanomaterials (NMs) have emerged as promising tools for disease diagnosis and therapy due to their unique physicochemical properties. To maximize the effectiveness and design of NMs-based medical applications, it is essential to comprehend the complex mechanisms of cellular uptake, subcellular localization, and cellular retention. This review illuminates the various pathways that NMs take to get from the extracellular environment to certain intracellular compartments by investigating the various mechanisms that underlie their interaction with cells. The cellular uptake of NMs involves complex interactions with cell membranes, encompassing endocytosis, phagocytosis, and other active transport mechanisms. Unique uptake patterns across cell types highlight the necessity for customized NMs designs. After internalization, NMs move through a variety of intracellular routes that affect where they are located subcellularly. Understanding these pathways is pivotal for enhancing the targeted delivery of therapeutic agents and imaging probes. Furthermore, the cellular retention of NMs plays a critical role in sustained therapeutic efficacy and long-term imaging capabilities. Factors influencing cellular retention include nanoparticle size, surface chemistry, and the cellular microenvironment. Strategies for prolonging cellular retention are discussed, including surface modifications and encapsulation techniques. In conclusion, a comprehensive understanding of the mechanisms governing cellular uptake, subcellular localization, and cellular retention of NMs is essential for advancing their application in disease diagnosis and therapy. This review provides insights into the intricate interplay between NMs and biological systems, offering a foundation for the rational design of next-generation nanomedicines.
Collapse
Affiliation(s)
- Yue Peng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhengshuang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Sun
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiuwan Lan
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
13
|
Arshad I, Kanwal A, Zafar I, Unar A, Mouada H, Razia IT, Arif S, Ahsan M, Kamal MA, Rashid S, Khan KA, Sharma R. Multifunctional role of nanoparticles for the diagnosis and therapeutics of cardiovascular diseases. ENVIRONMENTAL RESEARCH 2024; 242:117795. [PMID: 38043894 DOI: 10.1016/j.envres.2023.117795] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 10/26/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
The increasing burden of cardiovascular disease (CVD) remains responsible for morbidity and mortality worldwide; their effective diagnostic or treatment methods are of great interest to researchers. The use of NPs and nanocarriers in cardiology has drawn much interest. The present comprehensive review provides deep insights into the use of current and innovative approaches in CVD diagnostics to offer practical ways to utilize nanotechnological interventions and the critical elements in the CVD diagnosis, associated risk factors, and management strategies of patients with chronic CVDs. We proposed a decision tree-based solution by discussing the emerging applications of NPs for the higher number of rules to increase efficiency in treating CVDs. This review-based study explores the screening methods, tests, and toxicity to provide a unique way of creating a multi-parametric feature that includes cutting-edge techniques for identifying cardiovascular problems and their treatments. We discussed the benefits and drawbacks of various NPs in the context of cost, space, time and complexity that have been previously suggested in the literature for the diagnosis of CVDs risk factors. Also, we highlighted the advances in using NPs for targeted and improved drug delivery and discussed the evolution toward the nano-cardiovascular potential for medical science. Finally, we also examined the mixed-based diagnostic approaches crucial for treating cardiovascular disorders, broad applications and the potential future applications of nanotechnology in medical sciences.
Collapse
Affiliation(s)
- Ihtesham Arshad
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, 56300, Pakistan.
| | - Ayesha Kanwal
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, 56300, Pakistan.
| | - Imran Zafar
- Department of Bioinformatics and Computational Biology, Virtual University, Punjab, 54700, Pakistan.
| | - Ahsanullah Unar
- Department of Precision Medicine, University of Campania 'L. Vanvitelli', Naples, Italy.
| | - Hanane Mouada
- Department of Process Engineering, Institute of science University Center of Tipaza, Tipaza, Algeria.
| | | | - Safina Arif
- Medical Lab Technology, University of Lahore, Lahore, 54590, Pakistan.
| | - Muhammad Ahsan
- Institute of Environmental and Agricultural Sciences, University of Okara, Okara, 56300, Pakistan.
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, China; King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Bangladesh; Enzymoics, 7 Peterlee place, Hebersham, NSW, 2770, Australia; Novel Global Community Educational Foundation, Australia.
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam BinAbdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia.
| | - Khalid Ali Khan
- Unit of Bee Research and Honey Production, Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha, 61413, Saudi Arabia; Applied College, King Khalid University, P. O. Box 9004, Abha, 61413, Saudi Arabia.
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
14
|
Leng J, Wang C, Liang Z, Qiu F, Zhang S, Yang Y. An updated review of YAP: A promising therapeutic target against cardiac aging? Int J Biol Macromol 2024; 254:127670. [PMID: 37913886 DOI: 10.1016/j.ijbiomac.2023.127670] [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: 07/11/2023] [Revised: 09/05/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
The transcriptional co-activator Yes-associated protein (YAP) functions as a downstream effector of the Hippo signaling pathway and plays a crucial role in cardiomyocyte survival. In its non-phosphorylated activated state, YAP binds to transcription factors, activating the transcription of downstream target genes. It also regulates cell proliferation and survival by selectively binding to enhancers and activating target genes. However, the upregulation of the Hippo pathway in human heart failure inhibits cardiac regeneration and disrupts astrogenesis, thus preventing the nuclear translocation of YAP. Existing literature indicates that the Hippo/YAP axis contributes to inflammation and fibrosis, potentially playing a role in the development of cardiac, vascular and renal injuries. Moreover, it is a key mediator of myofibroblast differentiation and fibrosis in the infarcted heart. Given these insights, can we harness YAP's regenerative potential in a targeted manner? In this review, we provide a detailed discussion of the Hippo signaling pathway and consolidate concepts for the development and intervention of cardiac anti-aging drugs to leverage YAP signaling as a pivotal target.
Collapse
Affiliation(s)
- Jingzhi Leng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China
| | - Chuanzhi Wang
- College of Sports Science, South China Normal University, Guangzhou, China
| | - Zhide Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Fanghui Qiu
- School of Physical Education, Qingdao University, China
| | - Shuangshuang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China.
| | - Yuan Yang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China.
| |
Collapse
|
15
|
Ebrahimnejad P, Mohammadi Z, Babaei A, Ahmadi M, Amirkhanloo S, Asare-Addo K, Nokhodchid A. Novel Strategies Using Sagacious Targeting for Site-Specific Drug Delivery in Breast Cancer Treatment: Clinical Potential and Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:35-84. [PMID: 37824418 DOI: 10.1615/critrevtherdrugcarriersyst.v41.i1.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
For more than a decade, researchers have been working to achieve new strategies and smart targeting drug delivery techniques and technologies to treat breast cancer (BC). Nanotechnology presents a hopeful strategy for targeted drug delivery into the building of new therapeutics using the properties of nanomaterials. Nanoparticles are of high regard in the field of diagnosis and the treatment of cancer. The use of these nanoparticles as an encouraging approach in the treatment of various cancers has drawn the interest of researchers in recent years. In order to achieve the maximum therapeutic effectiveness in the treatment of BC, combination therapy has also been adopted, leading to minimal side effects and thus an enhancement in the quality of life for patients. This review article compares, discusses and criticizes the approaches to treat BC using novel design strategies and smart targeting of site-specific drug delivery systems.
Collapse
Affiliation(s)
- Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Mohammadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Melika Ahmadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shervin Amirkhanloo
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kofi Asare-Addo
- Department of Pharmacy, University of Huddersfield, Huddersfield, UK
| | - Ali Nokhodchid
- Lupin Pharmaceutical Research Center, Coral Springs, Florida, USA; Pharmaceutics Research Lab, Arundel Building, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
16
|
Carvalho T, Bártolo R, Pedro SN, Valente BFA, Pinto RJB, Vilela C, Shahbazi MA, Santos HA, Freire CSR. Injectable Nanocomposite Hydrogels of Gelatin-Hyaluronic Acid Reinforced with Hybrid Lysozyme Nanofibrils-Gold Nanoparticles for the Regeneration of Damaged Myocardium. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37200222 DOI: 10.1021/acsami.3c03874] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Biopolymeric injectable hydrogels are promising biomaterials for myocardial regeneration applications. Besides being biocompatible, they adjust themselves, perfectly fitting the surrounding tissue. However, due to their nature, biopolymeric hydrogels usually lack desirable functionalities, such as antioxidant activity and electrical conductivity, and in some cases, mechanical performance. Protein nanofibrils (NFs), such as lysozyme nanofibrils (LNFs), are proteic nanostructures with excellent mechanical performance and antioxidant activity, which can work as nanotemplates to produce metallic nanoparticles. Here, gold nanoparticles (AuNPs) were synthesized in situ in the presence of LNFs, and the obtained hybrid AuNPs@LNFs were incorporated into gelatin-hyaluronic acid (HA) hydrogels for myocardial regeneration applications. The resulting nanocomposite hydrogels showed improved rheological properties, mechanical resilience, antioxidant activity, and electrical conductivity, especially for the hydrogels containing AuNPs@LNFs. The swelling and bioresorbability ratios of these hydrogels are favorably adjusted at lower pH levels, which correspond to the ones in inflamed tissues. These improvements were observed while maintaining important properties, namely, injectability, biocompatibility, and the ability to release a model drug. Additionally, the presence of AuNPs allowed the hydrogels to be monitorable through computer tomography. This work demonstrates that LNFs and AuNPs@LNFs are excellent functional nanostructures to formulate injectable biopolymeric nanocomposite hydrogels for myocardial regeneration applications.
Collapse
Affiliation(s)
- Tiago Carvalho
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Raquel Bártolo
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sónia N Pedro
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Bruno F A Valente
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Ricardo J B Pinto
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Carla Vilela
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Carmen S R Freire
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
17
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Ji X, Meng Y, Wang Q, Tong T, Liu Z, Lin J, Li B, Wei Y, You X, Lei Y, Song M, Wang L, Guo Y, Qiu Y, Chen Z, Mai B, Xie S, Wu J, Cao N. Cysteine-Based Redox-Responsive Nanoparticles for Fibroblast-Targeted Drug Delivery in the Treatment of Myocardial Infarction. ACS NANO 2023; 17:5421-5434. [PMID: 36929948 DOI: 10.1021/acsnano.2c10042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Upon myocardial infarction (MI), activated cardiac fibroblasts (CFs) begin to remodel the myocardium, leading to cardiac fibrosis and even heart failure. No therapeutic approaches are currently available to prevent the development of MI-induced pathological fibrosis. Most pharmacological trials fail from poor local drug activity and side effects caused by systemic toxicity, largely due to the lack of a heart-targeted drug delivery system that is selective for activated CFs. Here, we developed a reduced glutathione (GSH)-responsive nanoparticle platform capable of targeted delivering of drugs to activated CFs within the infarct area of a post-MI heart. Compared with systemic drug administration, CF-targeted delivery of PF543, a sphingosine kinase 1 inhibitor identified in a high-throughput antifibrotic drug screening, had higher therapeutic efficacy and lower systemic toxicity in a MI mouse model. Our results provide a CF-targeted strategy to deliver therapeutic agents for pharmacological intervention of cardiac fibrosis.
Collapse
Affiliation(s)
- Xiaoqian Ji
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Yabin Meng
- School of Biomedical Engineering, Sun Yat-Sen University, Guangdong 510006, China
| | - Qiyuan Wang
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Tong Tong
- School of Biomedical Engineering, Sun Yat-Sen University, Guangdong 510006, China
| | - Zhun Liu
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Jianqing Lin
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Bin Li
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Yan Wei
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Xinru You
- School of Biomedical Engineering, Sun Yat-Sen University, Guangdong 510006, China
| | - Yushan Lei
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Mingyuan Song
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Liying Wang
- School of Biomedical Engineering, Sun Yat-Sen University, Guangdong 510006, China
| | - Yijie Guo
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Yuexiang Qiu
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Zhongyan Chen
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| | - Bifang Mai
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangdong 510120, China
| | - Shuanglun Xie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangdong 510120, China
| | - Jun Wu
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong 510120, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, SAR 999077, China
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangdong 511400, China
| | - Nan Cao
- Zhongshan School of Medicine and the Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong 518107, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangdong 510080, China
| |
Collapse
|
19
|
Saeed S, Ud Din SR, Khan SU, Gul R, Kiani FA, Wahab A, Zhong M. Nanoparticle: A Promising Player in Nanomedicine and its Theranostic Applications for the Treatment of Cardiovascular Diseases. Curr Probl Cardiol 2023; 48:101599. [PMID: 36681209 DOI: 10.1016/j.cpcardiol.2023.101599] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death around the world, a trend that will progressively grow over the next decade. Recently, with the advancement of nanotechnology, innovative nanoparticles (NPs) have been efficiently utilized in disease diagnosis and theranostic applications. In this review, we highlighted the benchmark summary of the recently synthesized NPs that are handy for imaging, diagnosis, and treatment of CVDs. NPs are the carrier of drug-delivery payloads actively reaching more areas of the heart and arteries, allowing them novel therapeutic agents for CVDs. Herein, due to the limited availability of literature, we only focused on NPs mechanism in the cardiovascular system and various treatment-based approaches that opens a new window for future research and versatile approach in the field of medical and clinical applications. Moreover, current challenges and limitations for the detection of CVDs has also discussed.
Collapse
Affiliation(s)
- Sumbul Saeed
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, P.R China.
| | - Shahid Ullah Khan
- Women Medical and Dental College, Khyber Medical University, Khyber Pakhtunkhwa, Pakistan
| | - Rukhsana Gul
- Department of Chemistry, Kohat University of Science and Technology, Khyber Pakhtunkhwa, Pakistan
| | - Faisal Ayub Kiani
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Bahauddin Zakariyah University, Multan, 60800, Pakistan.
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan.
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, P.R China.
| |
Collapse
|
20
|
Li D, Son Y, Jang M, Wang S, Zhu W. Nanoparticle Based Cardiac Specific Drug Delivery. BIOLOGY 2023; 12:biology12010082. [PMID: 36671774 PMCID: PMC9856055 DOI: 10.3390/biology12010082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Heart failure secondary to myocardial injuries is a leading cause of death worldwide. Recently, a growing number of novel therapies have emerged for injured myocardium repairment. However, delivering therapeutic agents specifically to the injured heart remains a significant challenge. Nanoparticles are the most commonly used vehicles for targeted drug delivery. Various nanoparticles have been synthesized to deliver drugs and other therapeutic molecules to the injured heart via passive or active targeting approaches, and their targeting specificity and therapeutic efficacies have been investigated. Here, we summarized nanoparticle-based, cardiac-specific drug delivery systems, their potency for treating heart diseases, and the mechanisms underlying these cardiac-targeting strategies. We also discussed the clinical studies that have employed nanoparticle-based cardiac-specific drug delivery.
Collapse
Affiliation(s)
- Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Department of Cardiology, Dongfang Hospital, The Second Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yura Son
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Correspondence: (S.W.); (W.Z.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Correspondence: (S.W.); (W.Z.)
| |
Collapse
|
21
|
Zhang DX, Tieu T, Esser L, Wojnilowicz M, Lee CH, Cifuentes-Rius A, Thissen H, Voelcker NH. Differential Surface Engineering Generates Core-Shell Porous Silicon Nanoparticles for Controlled and Targeted Delivery of an Anticancer Drug. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54539-54549. [PMID: 36469497 DOI: 10.1021/acsami.2c16370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
An approach to differentially modify the internal surface of porous silicon nanoparticles (pSiNPs) with hydrophobic dodecene and the external surface with antifouling poly-N-(2-hydroxypropyl) acrylamide (polyHPAm) as well as a cell-targeting peptide was developed. Specifically, to generate these core-shell pSiNPs, the interior surface of a porous silicon (pSi) film was hydrosilylated with 1-dodecene, followed by ultrasonication to create pSiNPs. The new external surfaces were modified by silanization with a polymerization initiator, and surface-initiated atom transfer radical polymerization was performed to introduce polyHPAm brushes. Afterward, a fraction of the polymer side chain hydroxyl groups was activated to conjugate cRGDfK─a peptide with a high affinity and selectivity for the ανβ3 integrin receptor that is overexpressed in prostate and melanoma cancers. Finally, camptothecin, a hydrophobic anti-cancer drug, was successfully loaded into the pores. This drug delivery system showed excellent colloidal stability in a cell culture medium, and the in vitro drug release kinetics could be fine-tuned by the combination of internal and external surface modifications. In vitro studies by confocal microscopy and flow cytometry revealed improved cellular association attributed to cRGDfK. Furthermore, the cell viability results showed that the drug-loaded and peptide-functionalized nanoparticles had enhanced cytotoxicity toward a C4-2B prostate carcinoma cell line in both 2D cell culture and a 3D spheroid model.
Collapse
Affiliation(s)
- De-Xiang Zhang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Terence Tieu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Marcin Wojnilowicz
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Chieh-Hua Lee
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
| | - Anna Cifuentes-Rius
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Helmut Thissen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| |
Collapse
|
22
|
Yue T, Xiong S, Zheng D, Wang Y, Long P, Yang J, Danzeng D, Gao H, Wen X, Li X, Hou J. Multifunctional biomaterial platforms for blocking the fibrosis process and promoting cellular restoring effects in myocardial fibrosis therapy. Front Bioeng Biotechnol 2022; 10:988683. [PMID: 36185428 PMCID: PMC9520723 DOI: 10.3389/fbioe.2022.988683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Myocardial fibrosis is the result of abnormal healing after acute and chronic myocardial damage and is a direct cause of heart failure and cardiac insufficiency. The clinical approach is to preserve cardiac function and inhibit fibrosis through surgery aimed at dredging blood vessels. However, this strategy does not adequately address the deterioration of fibrosis and cardiac function recovery. Therefore, numerous biomaterial platforms have been developed to address the above issues. In this review, we summarize the existing biomaterial delivery and restoring platforms, In addition, we also clarify the therapeutic strategies based on biomaterial platforms, including general strategies to block the fibrosis process and new strategies to promote cellular restoring effects. The development of structures with the ability to block further fibrosis progression as well as to promote cardiomyocytes viability should be the main research interests in myocardial fibrosis, and the reestablishment of structures necessary for normal cardiac function is central to the treatment of myocardial fibrosis. Finally, the future application of biomaterials for myocardial fibrosis is also highlighted.
Collapse
Affiliation(s)
- Tian Yue
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Shiqiang Xiong
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
| | - Dezhi Zheng
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Pan Long
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jiali Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Dunzhu Danzeng
- Department of Basic Medicine, Medical College, Tibet University, Lhasa, China
| | - Han Gao
- Department of Basic Medicine, Medical College, Tibet University, Lhasa, China
| | - Xudong Wen
- Department of Gastroenterology and Hepatology, Chengdu First People’s Hospital, Chengdu, China
- *Correspondence: Xudong Wen, ; Xin Li, ; Jun Hou,
| | - Xin Li
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- *Correspondence: Xudong Wen, ; Xin Li, ; Jun Hou,
| | - Jun Hou
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Cardiovascular Disease Research Institute of Chengdu, Chengdu, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
- *Correspondence: Xudong Wen, ; Xin Li, ; Jun Hou,
| |
Collapse
|
23
|
Iravani S, Varma RS. Advanced Drug Delivery Micro- and Nanosystems for Cardiovascular Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185843. [PMID: 36144581 PMCID: PMC9506137 DOI: 10.3390/molecules27185843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Advanced drug delivery micro- and nanosystems have been widely explored due to their appealing specificity/selectivity, biodegradability, biocompatibility, and low toxicity. They can be applied for the targeted delivery of pharmaceuticals, with the benefits of good biocompatibility/stability, non-immunogenicity, large surface area, high drug loading capacity, and low leakage of drugs. Cardiovascular diseases, as one of the primary mortalities cause worldwide with significant impacts on the quality of patients’ life, comprise a variety of heart and circulatory system pathologies, such as peripheral vascular diseases, myocardial infarction, heart failure, and coronary artery diseases. Designing novel micro- and nanosystems with suitable targeting properties and smart release behaviors can help circumvent crucial challenges of the tolerability, low stability, high toxicity, and possible side- and off-target effects of conventional drug delivery routes. To overcome different challenging issues, namely physiological barriers, low efficiency of drugs, and possible adverse side effects, various biomaterials-mediated drug delivery systems have been formulated with reduced toxicity, improved pharmacokinetics, high bioavailability, sustained release behavior, and enhanced therapeutic efficacy for targeted therapy of cardiovascular diseases. Despite the existing drug delivery systems encompassing a variety of biomaterials for treating cardiovascular diseases, the number of formulations currently approved for clinical use is limited due to the regulatory and experimental obstacles. Herein, the most recent advancements in drug delivery micro- and nanosystems designed from different biomaterials for the treatment of cardiovascular diseases are deliberated, with a focus on the important challenges and future perspectives.
Collapse
Affiliation(s)
- Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
- Correspondence: (S.I.); (R.S.V.)
| | - Rajender S. Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University in Olomouc, Šlechtitelů 27, Olomouc 78371, Czech Republic
- Correspondence: (S.I.); (R.S.V.)
| |
Collapse
|
24
|
Yang L, Peng J, Shi A, Wang X, Li J, Su Y, Yin K, Zhao L, Zhao Y. Myocardium-Targeted Micelle Nanomedicine That Salvages the Heart from Ischemia/Reperfusion Injury. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38562-38574. [PMID: 35973832 DOI: 10.1021/acsami.2c11117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cardioprotective medication is the common treatment to relieve myocardial ischemia/reperfusion (I/R) injury. However, limited by the low bioavailability of therapeutic drugs, the therapeutic outcome is barely satisfactory. Because the I/R injury can enhance the permeability of the vasculature and allow the extravasation of nanoparticles into the surrounding tissue, herein we formulate the cardiotonic drug olprinone (Olp) in cross-linked micelles as the nanomedicine to achieve myocardium-targeted delivery after systematic administration. As a result, the local concentration of Olp in the injured myocardium is raised by orders of magnitude with prolonged drug duration time. The treatment successfully preserves the pumping efficiency of the heart, alleviates ventricular remodeling, and thus stops the positive feedback loop for the deteriorated cardiac function. Consequently, the myocardium-targeted nanomedicine significantly salvages the heart from I/R injury before irreversible pathological changes take place.
Collapse
Affiliation(s)
- Liqiang Yang
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Juanjuan Peng
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Aiping Shi
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xueshen Wang
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Junyao Li
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yaoquan Su
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Kunpeng Yin
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Lingzhi Zhao
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
25
|
Lv Q, Ma B, Li W, Fu G, Wang X, Xiao Y. Nanomaterials-Mediated Therapeutics and Diagnosis Strategies for Myocardial Infarction. Front Chem 2022; 10:943009. [PMID: 35873037 PMCID: PMC9301085 DOI: 10.3389/fchem.2022.943009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
The alarming mortality and morbidity rate of myocardial infarction (MI) is becoming an important impetus in the development of early diagnosis and appropriate therapeutic approaches, which are critical for saving patients' lives and improving post-infarction prognosis. Despite several advances that have been made in the treatment of MI, current strategies are still far from satisfactory. Nanomaterials devote considerable contribution to tackling the drawbacks of conventional therapy of MI by improving the homeostasis in the cardiac microenvironment via targeting, immune modulation, and repairment. This review emphasizes the strategies of nanomaterials-based MI treatment, including cardiac targeting drug delivery, immune-modulation strategy, antioxidants and antiapoptosis strategy, nanomaterials-mediated stem cell therapy, and cardiac tissue engineering. Furthermore, nanomaterials-based diagnosis strategies for MI was presented in term of nanomaterials-based immunoassay and nano-enhanced cardiac imaging. Taken together, although nanomaterials-based strategies for the therapeutics and diagnosis of MI are both promising and challenging, such a strategy still explores the immense potential in the development of the next generation of MI treatment.
Collapse
Affiliation(s)
- Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Boxuan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wujiao Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Yun Xiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Qian J, Wen H, Tamarov K, Xu W, Lehto V. Recent Developments in Porous Silicon Nanovectors with Various Imaging Modalities in the Framework of Theranostics. ChemMedChem 2022; 17:e202200004. [PMID: 35212460 PMCID: PMC9314675 DOI: 10.1002/cmdc.202200004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/24/2022] [Indexed: 11/17/2022]
Abstract
The number of in vitro, ex vivo, and in vivo studies on porous silicon (PSi) nanoparticles for biomedical applications has increased extensively over the last decade. The focus of the reports has been on the carrier properties of PSi concerning the therapeutic aspect due to several beneficial nanovector characteristics including high payload capacity, biocompatibility, and versatile surface chemistry. Recently, increasing attention has been paid to the diagnostic aspects of PSi, which is typically attributed to the biotraceability of the nanovector. Also, PSi has been studied as a contrast agent. When both these aspects, therapy and diagnosis, are integrated into one nanovector, we can discuss a real nanotheranostics approach. Herein, we review the recent progress developing PSi for various imaging modalities, specifically focusing on optical imaging, magnetic resonance imaging, and nuclear medicine imaging. Furthermore, we summarized the knowledge gaps that must be covered before applying PSi in clinical imaging, highlighting future research trends.
Collapse
Affiliation(s)
- Jing Qian
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
- College of Chemistry and Chemical EngineeringLanzhou UniversityLanzhou730000China
| | - Huang Wen
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| | - Konstantin Tamarov
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| | - Wujun Xu
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| |
Collapse
|
27
|
George TA, Hsu CC, Meeson A, Lundy DJ. Nanocarrier-Based Targeted Therapies for Myocardial Infarction. Pharmaceutics 2022; 14:930. [PMID: 35631516 PMCID: PMC9143269 DOI: 10.3390/pharmaceutics14050930] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/30/2022] Open
Abstract
Myocardial infarction is a major cause of morbidity and mortality worldwide. Due to poor inherent regeneration of the adult mammalian myocardium and challenges with effective drug delivery, there has been little progress in regenerative therapies. Nanocarriers, including liposomes, nanoparticles, and exosomes, offer many potential advantages for the therapy of myocardial infarction, including improved delivery, retention, and prolonged activity of therapeutics. However, there are many challenges that have prevented the widespread clinical use of these technologies. This review aims to summarize significant principles and developments in the field, with a focus on nanocarriers using ligand-based or cell mimicry-based targeting. Lastly, a discussion of limitations and potential future direction is provided.
Collapse
Affiliation(s)
- Thomashire A. George
- International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
| | - Chuan-Chih Hsu
- Department of Cardiovascular Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Annette Meeson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK;
| | - David J. Lundy
- International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
28
|
Liu C, Fan Z, He D, Chen H, Zhang S, Guo S, Zheng B, Cen H, Zhao Y, Liu H, Wang L. Designer Functional Nanomedicine for Myocardial Repair by Regulating the Inflammatory Microenvironment. Pharmaceutics 2022; 14:pharmaceutics14040758. [PMID: 35456592 PMCID: PMC9025700 DOI: 10.3390/pharmaceutics14040758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction is a major global health problem, and the repair of damaged myocardium is still a major challenge. Myocardial injury triggers an inflammatory response: immune cells infiltrate into the myocardium while activating myofibroblasts and vascular endothelial cells, promoting tissue repair and scar formation. Fragments released by cardiomyocytes become endogenous “danger signals”, which are recognized by cardiac pattern recognition receptors, activate resident cardiac immune cells, release thrombin factors and inflammatory mediators, and trigger severe inflammatory responses. Inflammatory signaling plays an important role in the dilation and fibrosis remodeling of the infarcted heart, and is a key event driving the pathogenesis of post-infarct heart failure. At present, there is no effective way to reverse the inflammatory microenvironment in injured myocardium, so it is urgent to find new therapeutic and diagnostic strategies. Nanomedicine, the application of nanoparticles for the prevention, treatment, and imaging of disease, has produced a number of promising applications. This review discusses the treatment and challenges of myocardial injury and describes the advantages of functional nanoparticles in regulating the myocardial inflammatory microenvironment and overcoming side effects. In addition, the role of inflammatory signals in regulating the repair and remodeling of infarcted hearts is discussed, and specific therapeutic targets are identified to provide new therapeutic ideas for the treatment of myocardial injury.
Collapse
Affiliation(s)
- Chunping Liu
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhijin Fan
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China;
| | - Dongyue He
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Huiqi Chen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Shihui Zhang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Sien Guo
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Bojun Zheng
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Huan Cen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Yunxuan Zhao
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Hongxing Liu
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510230, China
- Correspondence: (H.L.); (L.W.)
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
- Correspondence: (H.L.); (L.W.)
| |
Collapse
|
29
|
Gonciar D, Mocan T, Agoston-Coldea L. Nanoparticles Targeting the Molecular Pathways of Heart Remodeling and Regeneration. Pharmaceutics 2022; 14:pharmaceutics14040711. [PMID: 35456545 PMCID: PMC9028351 DOI: 10.3390/pharmaceutics14040711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiovascular diseases are the main cause of death worldwide, a trend that will continue to grow over the next decade. The heart consists of a complex cellular network based mainly on cardiomyocytes, but also on endothelial cells, smooth muscle cells, fibroblasts, and pericytes, which closely communicate through paracrine factors and direct contact. These interactions serve as valuable targets in understanding the phenomenon of heart remodeling and regeneration. The advances in nanomedicine in the controlled delivery of active pharmacological agents are remarkable and may provide substantial contribution to the treatment of heart diseases. This review aims to summarize the main mechanisms involved in cardiac remodeling and regeneration and how they have been applied in nanomedicine.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| | - Teodora Mocan
- Physiology Department, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca 400162, Romania
- Correspondence:
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| |
Collapse
|
30
|
Chopra H, Bibi S, Mishra AK, Tirth V, Yerramsetty SV, Murali SV, Ahmad SU, Mohanta YK, Attia MS, Algahtani A, Islam F, Hayee A, Islam S, Baig AA, Emran TB. Nanomaterials: A Promising Therapeutic Approach for Cardiovascular Diseases. JOURNAL OF NANOMATERIALS 2022; 2022:1-25. [DOI: 10.1155/2022/4155729] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Cardiovascular diseases (CVDs) are a primary cause of death globally. A few classic and hybrid treatments exist to treat CVDs. However, they lack in both safety and effectiveness. Thus, innovative nanomaterials for disease diagnosis and treatment are urgently required. The tiny size of nanomaterials allows them to reach more areas of the heart and arteries, making them ideal for CVDs. Atherosclerosis causes arterial stenosis and reduced blood flow. The most common treatment is medication and surgery to stabilize the disease. Nanotechnologies are crucial in treating vascular disease. Nanomaterials may be able to deliver medications to lesion sites after being infused into the circulation. Newer point-of-care devices have also been considered together with nanomaterials. For example, this study will look at the use of nanomaterials in imaging, diagnosing, and treating CVDs.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Shabana Bibi
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, 650091 Yunnan, China
- The International Joint Research Center for Sustainable Utilization of Cordyceps Bioresources in China and Southeast Asia, Yunnan University, Kunming, 650091 Yunnan, China
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, Republic of Korea
| | - Vineet Tirth
- Mechanical Engineering Department, College of Engineering, King Khalid University, Abha, 61421 Asir, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Guraiger, Abha, 61413 Asir, P.O. Box No. 9004, Saudi Arabia
| | - Sree Vandana Yerramsetty
- Department of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613402, India
| | - Sree Varshini Murali
- Department of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613402, India
| | - Syed Umair Ahmad
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Yugal Kishore Mohanta
- Department of Applied Biology, University of Science and Technology Meghalaya, Ri-Bhoi 793101, India
| | - Mohamed S. Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Ali Algahtani
- Mechanical Engineering Department, College of Engineering, King Khalid University, Abha, 61421 Asir, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Guraiger, Abha, 61413 Asir, P.O. Box No. 9004, Saudi Arabia
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Abdul Hayee
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Saiful Islam
- Civil Engineering Department, College of Engineering, King Khalid University, Abha, 61421 Asir, Saudi Arabia
| | - Atif Amin Baig
- Unit of Biochemistry, Faculty of Medicine, Universiti Sultan Zainal Abidin, Malaysia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|
31
|
Wang J, Zhang S, Di L. Acute myocardial infarction therapy: in vitro and in vivo evaluation of atrial natriuretic peptide and triphenylphosphonium dual ligands modified, baicalin-loaded nanoparticulate system. Drug Deliv 2021; 28:2198-2204. [PMID: 34662253 PMCID: PMC8525923 DOI: 10.1080/10717544.2021.1989086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Myocardial infarction (MI) is one of the most common ischemic heart diseases. It is very essential to explore new types of cardioprotective drugs delivery systems in this area. Objective The aim of the present study was to investigate the protective effect of baicalin (BA) and puerarin (PU) against acute MI rat models. BA and PU co-loaded nanoparticulate system were developed to improve bioavailability of the drugs, to prolong retention time in vivo and to enhance the protective effect. Methods In the present study, ANP and TPP contained ligands were synthesized. ANP/TPP-BN-LPNs were prepared and its physico-chemical properties were evaluated. The MI therapy efficiency of ANP/TPP-BN-LPNs was assessed in rats after intravenous injection. Single ligand contained LPNs, no ligand contained LPNs, and BN solution formulations were also prepared and used for the comparison. Results ANP/TPP-BN-LPNs were uniform and spheroidal particles. The size of ANP/TPP-BN-LPNs was 98.5 ± 2.9 nm, with a zeta potential of –19.5 ± 1.9 mV. The dual ligands modified LPNs exhibited significantly improved therapeutic efficiency compared with the single ligand modified LPNs and other systems. In vivo infarct therapy studies in rats proved that ANP/TPP-BN-LPNs were a promising system for efficient delivery of cardiovascular drugs for the treatment of cardiovascular diseases. Conclusions ANP/TPP-BN-LPNs could be used as a long-circulating and heart-targeting drug delivery system.
Collapse
Affiliation(s)
- Jie Wang
- Intervention Center, Linyi People's Hospital Beicheng New District Hospital, Linyi, PR China
| | - Shouwen Zhang
- Cardiology Pacing and Electrophysiology Ward, Linyi People's Hospital Beicheng New District Hospital, Linyi, PR China
| | - Lizhe Di
- Oral Cavity Clinic, Linyi People's Hospital, Linyi, PR China
| |
Collapse
|
32
|
Pan Q, Xu J, Wen CJ, Xiong YY, Gong ZT, Yang YJ. Nanoparticles: Promising Tools for the Treatment and Prevention of Myocardial Infarction. Int J Nanomedicine 2021; 16:6719-6747. [PMID: 34621124 PMCID: PMC8491866 DOI: 10.2147/ijn.s328723] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
Despite several recent advances, current therapy and prevention strategies for myocardial infarction are far from satisfactory, owing to limitations in their applicability and treatment effects. Nanoparticles (NPs) enable the targeted and stable delivery of therapeutic compounds, enhance tissue engineering processes, and regulate the behaviour of transplants such as stem cells. Thus, NPs may be more effective than other mechanisms, and may minimize potential adverse effects. This review provides evidence for the view that function-oriented systems are more practical than traditional material-based systems; it also summarizes the latest advances in NP-based strategies for the treatment and prevention of myocardial infarction.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Cen-Jin Wen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
33
|
Guo J, Yang Z, Wang X, Xu Y, Lu Y, Qin Z, Zhang L, Xu J, Wang W, Zhang J, Tang J. Advances in Nanomaterials for Injured Heart Repair. Front Bioeng Biotechnol 2021; 9:686684. [PMID: 34513807 PMCID: PMC8424111 DOI: 10.3389/fbioe.2021.686684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the leading causes of mortality worldwide. Because of the limited regenerative capacity of adult myocardium to compensate for the loss of heart tissue after ischemic infarction, scientists have been exploring the possible mechanisms involved in the pathological process of ASCVD and searching for alternative means to regenerate infarcted cardiac tissue. Although numerous studies have pursued innovative solutions for reversing the pathological process of ASCVD and improving the effectiveness of delivering therapeutics, the translation of those advances into downstream clinical applications remains unsatisfactory because of poor safety and low efficacy. Recently, nanomaterials (NMs) have emerged as a promising new strategy to strengthen both the efficacy and safety of ASCVD therapy. Thus, a comprehensive review of NMs used in ASCVD treatment will be useful. This paper presents an overview of the pathophysiological mechanisms of ASCVD and the multifunctional mechanisms of NM-based therapy, including antioxidative, anti-inflammation and antiapoptosis mechanisms. The technological improvements of NM delivery are summarized and the clinical transformations concerning the use of NMs to treat ASCVD are examined. Finally, this paper discusses the challenges and future perspectives of NMs in cardiac regeneration to provide insightful information for health professionals on the latest advancements in nanotechnologies for ASCVD treatment.
Collapse
Affiliation(s)
- Jiacheng Guo
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhenzhen Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Wang
- Department of Medical Record Management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanyan Xu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Yongzheng Lu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhen Qin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Li Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Jing Xu
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
34
|
Liu C, Chen L, Ma Y, Hu K, Wu P, Pan L, Chen H, Li L, Hu H, Zhang J. Pulmonary circulation-mediated heart targeting for the prevention of heart failure by inhalation of intrinsically bioactive nanoparticles. Am J Cancer Res 2021; 11:8550-8569. [PMID: 34373758 PMCID: PMC8343995 DOI: 10.7150/thno.61875] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/08/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure is a serious clinical and public health problem. Currently there is an unmet demand for effective therapies for heart failure. Herein we reported noninvasive inhalation delivery of nanotherapies to prevent heart failure. Methods: A reactive oxygen species (ROS)-scavenging material (TPCD) was synthesized, which was processed into antioxidative and anti-inflammatory nanoparticles (i.e., TPCD NP). By decoration with a mitochondrial-targeting moiety, a multilevel targeting nanotherapy TTPCD NP was engineered. Pulmonary accumulation of inhaled TPCD NP and underlying mechanisms were examined in mice. In vivo efficacies of nanotherapies were evaluated in mice with doxorubicin (DOX)-induced cardiomyopathy. Further, an antioxidative, anti-inflammatory, and pro-resolving nanotherapy (i.e., ATTPCD NP) was developed, by packaging a peptide Ac2-26. In vitro and in vivo efficacies of ATTPCD NP were also evaluated. Results: TPCD NP alleviated DOX-induced oxidative stress and cell injury by internalization in cardiomyocytes and scavenging overproduced ROS. Inhaled TPCD NP can accumulate in the heart of mice by transport across the lung epithelial and endothelial barriers. Correspondingly, inhaled TPCD NP effectively inhibited DOX-induced heart failure in mice. TTPCD NP showed considerably enhanced heart targeting capability, cellular uptake efficiency, and mitochondrial localization capacity, thereby potentiating therapeutic effects. Notably, TPCD NP can serve as bioactive and ROS-responsive nanovehicles to achieve combination therapy with Ac2-26, affording further enhanced efficacies. Importantly, inhaled TPCD NP displayed good safety at a dose 5-fold higher than the efficacious dose. Conclusions: Inhalation delivery of nanoparticles is an effective, safe, and noninvasive strategy for targeted treatment of heart diseases. TPCD NP-based nanotherapies are promising drugs for heart failure and other acute/chronic heart diseases associated with oxidative stress.
Collapse
|
35
|
Cheng R, Wang S, Moslova K, Mäkilä E, Salonen J, Li J, Hirvonen J, Xia B, Santos HA. Quantitative Analysis of Porous Silicon Nanoparticles Functionalization by 1H NMR. ACS Biomater Sci Eng 2021; 8:4132-4139. [PMID: 34292713 PMCID: PMC9554871 DOI: 10.1021/acsbiomaterials.1c00440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Porous silicon (PSi)
nanoparticles have been applied in various
fields, such as catalysis, imaging, and biomedical applications, because
of their large specific surface area, easily modifiable surface chemistry,
biocompatibility, and biodegradability. For biomedical applications,
it is important to precisely control the surface modification of PSi-based
materials and quantify the functionalization density, which determines
the nanoparticle’s behavior in the biological system. Therefore,
we propose here an optimized solution to quantify the functionalization
groups on PSi, based on the nuclear magnetic resonance (NMR) method
by combining the hydrolysis with standard 1H NMR experiments.
We optimized the hydrolysis conditions to degrade the PSi, providing
mobility to the molecules for NMR detection. The NMR parameters were
also optimized by relaxation delay and the number of scans to provide
reliable NMR spectra. With an internal standard, we quantitatively
analyzed the surficial amine groups and their sequential modification
of polyethylene glycol. Our investigation provides a reliable, fast,
and straightforward method in quantitative analysis of the surficial
modification characterization of PSi requiring a small amount of sample.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Karina Moslova
- Department of Chemistry, Faculty of Science, University of Helsinki, Helsinki FI-00014, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku FI-20014, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku FI-20014, Finland
| | - Jiachen Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.,College of Science Key Laboratory of Forest Genetics & Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Bing Xia
- College of Science Key Laboratory of Forest Genetics & Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.,Helsinki Insititute of Life Science, HiLIFE, University of Helsinki, Helsinki FI-00014, Finland
| |
Collapse
|
36
|
Albumin Nanoparticle Formulation for Heart-Targeted Drug Delivery: In Vivo Assessment of Congestive Heart Failure. Pharmaceuticals (Basel) 2021; 14:ph14070697. [PMID: 34358122 PMCID: PMC8308836 DOI: 10.3390/ph14070697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 01/14/2023] Open
Abstract
Congestive heart failure is a fatal cardiovascular disease resulting in tissue necrosis and loss of cardiac contractile function. Inotropic drugs such as milrinone are commonly used to improve the myocardial contractility and heart function. However, milrinone is associated with severe side effects and lower circulation time. In this article, a novel protein nanoparticle formulation for heart-targeted delivery of milrinone has been designed and tested. The formulation was prepared using albumin protein conjugated with the targeting ligand, angiotensin II peptide to form nanoparticles following the ethanol desolvation method. The formulation was characterized for size, charge, and morphology and tested in a rat model of congestive heart failure to study pharmacokinetics, biodistribution, and efficacy. The overall cardiac output parameters were evaluated comparing the formulation with the control non-targeted drug, milrinone lactate. This formulation exhibited improved pharmacokinetics with a mean retention time of 123.7 min, half-life of 101.3 min, and clearance rate of 0.24 L/(kg*h). The targeted formulation also significantly improved ejection fraction and fractional shortening parameters thus improving cardiac function. This study demonstrates a new approach in delivering inotropic drugs such as milrinone for superior treatment of congestive heart failure.
Collapse
|
37
|
Qi Y, Li J, Nie Q, Gao M, Yang Q, Li Z, Li Q, Han S, Ding J, Li Y, Zhang J. Polyphenol-assisted facile assembly of bioactive nanoparticles for targeted therapy of heart diseases. Biomaterials 2021; 275:120952. [PMID: 34147720 DOI: 10.1016/j.biomaterials.2021.120952] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022]
Abstract
It remains a great challenge for targeted therapy of heart diseases. To achieve desirable heart targeting, we developed a polyphenol-assisted nanoprecipitation/self-assembly approach for facile engineering of functional nanoparticles. Three different materials were employed as representative carriers, while gallic acid, catechin, epigallocatechin gallate, and tannic acid (TA) served as typical polyphenols with varied numbers of phenolic hydroxyl groups. By optimizing different parameters, such as polyphenol types and the weight ratio of carrier materials and polyphenols, well-defined nanoparticles with excellent physicochemical properties can be easily prepared. Regardless of various carrier materials, TA-derived nanoparticles showed potent reactive oxygen species-scavenging activity, especially nanoparticles produced from a cyclodextrin-derived bioactive material (TPCD). By internalization into cardiomyocytes, TPCD/TA nanoparticles (defined as TPTN) effectively protected cells from hypoxic-ischemic injury. After intravenous injection, TPTN considerably accumulated in the injured heart in two murine models of ventricular fibrillation cardiac arrest in rats and myocardial hypertrophy in mice. Correspondingly, intravenously delivered TPTN afforded excellent therapeutic effects in both heart diseases. Preliminary experiments also revealed good safety of TPTN. These results substantiated that TPTN is a promising nanotherapy for targeted treatment of heart diseases, while polyphenol-assisted self-assembly is a facile but robust strategy to develop heart-targeting delivery systems.
Collapse
Affiliation(s)
- Yuantong Qi
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jingru Li
- Department of Biomedical Engineering and Medical Imaging, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qiang Nie
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mingjie Gao
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qinghua Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zimeng Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qi Li
- Department of Biomedical Engineering and Medical Imaging, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Songling Han
- State Key Lab of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Ding
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongqin Li
- Department of Biomedical Engineering and Medical Imaging, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; State Key Lab of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
38
|
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
|
39
|
Wang S, Fontana F, Shahbazi MA, Santos HA. Acetalated dextran based nano- and microparticles: synthesis, fabrication, and therapeutic applications. Chem Commun (Camb) 2021; 57:4212-4229. [PMID: 33913978 DOI: 10.1039/d1cc00811k] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acetalated dextran (Ac-DEX) is a pH-responsive dextran derivative polymer. Prepared by a simple acetalation reaction, Ac-DEX has tunable acid-triggered release profile. Despite its relatively short research history, Ac-DEX has shown great potential in various therapeutic applications. Furthermore, the recent functionalization of Ac-DEX makes versatile derivatives with additional properties. Herein, we summarize the cutting-edge development of Ac-DEX and related polymers. Specifically, we focus on the chemical synthesis, nano- and micro-particle fabrication techniques, the controlled-release mechanisms, and the rational design Ac-DEX-based of drug delivery systems in various biomedical applications. Finally, we briefly discuss the challenges and future perspectives in the field.
Collapse
Affiliation(s)
- Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran and Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
40
|
Välimäki MJ, Leigh RS, Kinnunen SM, March AR, de Sande AH, Kinnunen M, Varjosalo M, Heinäniemi M, Kaynak BL, Ruskoaho H. GATA-targeted compounds modulate cardiac subtype cell differentiation in dual reporter stem cell line. Stem Cell Res Ther 2021; 12:190. [PMID: 33736688 PMCID: PMC7977156 DOI: 10.1186/s13287-021-02259-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background Pharmacological modulation of cell fate decisions and developmental gene regulatory networks holds promise for the treatment of heart failure. Compounds that target tissue-specific transcription factors could overcome non-specific effects of small molecules and lead to the regeneration of heart muscle following myocardial infarction. Due to cellular heterogeneity in the heart, the activation of gene programs representing specific atrial and ventricular cardiomyocyte subtypes would be highly desirable. Chemical compounds that modulate atrial and ventricular cell fate could be used to improve subtype-specific differentiation of endogenous or exogenously delivered progenitor cells in order to promote cardiac regeneration. Methods Transcription factor GATA4-targeted compounds that have previously shown in vivo efficacy in cardiac injury models were tested for stage-specific activation of atrial and ventricular reporter genes in differentiating pluripotent stem cells using a dual reporter assay. Chemically induced gene expression changes were characterized by qRT-PCR, global run-on sequencing (GRO-seq) and immunoblotting, and the network of cooperative proteins of GATA4 and NKX2-5 were further explored by the examination of the GATA4 and NKX2-5 interactome by BioID. Reporter gene assays were conducted to examine combinatorial effects of GATA-targeted compounds and bromodomain and extraterminal domain (BET) inhibition on chamber-specific gene expression. Results GATA4-targeted compounds 3i-1000 and 3i-1103 were identified as differential modulators of atrial and ventricular gene expression. More detailed structure-function analysis revealed a distinct subclass of GATA4/NKX2-5 inhibitory compounds with an acetyl lysine-like domain that contributed to ventricular cells (%Myl2-eGFP+). Additionally, BioID analysis indicated broad interaction between GATA4 and BET family of proteins, such as BRD4. This indicated the involvement of epigenetic modulators in the regulation of GATA-dependent transcription. In this line, reporter gene assays with combinatorial treatment of 3i-1000 and the BET bromodomain inhibitor (+)-JQ1 demonstrated the cooperative role of GATA4 and BRD4 in the modulation of chamber-specific cardiac gene expression. Conclusions Collectively, these results indicate the potential for therapeutic alteration of cell fate decisions and pathological gene regulatory networks by GATA4-targeted compounds modulating chamber-specific transcriptional programs in multipotent cardiac progenitor cells and cardiomyocytes. The compound scaffolds described within this study could be used to develop regenerative strategies for myocardial regeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02259-z.
Collapse
Affiliation(s)
- Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Robert S Leigh
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Alexander R March
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Ana Hernández de Sande
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Matias Kinnunen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bogac L Kaynak
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| |
Collapse
|
41
|
Nanostructured Polymeric, Liposomal and Other Materials to Control the Drug Delivery for Cardiovascular Diseases. Pharmaceutics 2020; 12:pharmaceutics12121160. [PMID: 33260547 PMCID: PMC7760553 DOI: 10.3390/pharmaceutics12121160] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, taking an estimated 17.9 million lives each year, representing one third of global mortality. As existing therapies still have limited success, due to the inability to control the biodistribution of the currently approved drugs, the quality of life of these patients is modest. The advent of nanomedicine has brought new insights in innovative treatment strategies. For this reason, several novel nanotechnologies have been developed for both targeted and prolonged delivery of therapeutics to the cardiovascular system tο minimize side effects. In this regard, nanoparticles made of natural and/or synthetic nanomaterials, like liposomes, polymers or inorganic materials, are emerging alternatives for the encapsulation of already approved drugs to control their delivery in a targeted way. Therefore, nanomedicine has attracted the attention of the scientific community as a potential platform to deliver therapeutics to the injured heart. In this review, we discuss the current types of biomaterials that have been investigated as potential therapeutic interventions for CVDs as they open up a host of possibilities for more targeted and effective therapies, as well as minimally invasive treatments.
Collapse
|
42
|
Rahikkala A, Fontana F, Bauleth-Ramos T, Correia A, Kemell M, Seitsonen J, Mäkilä E, Sarmento B, Salonen J, Ruokolainen J, Hirvonen J, Santos HA. Hybrid red blood cell membrane coated porous silicon nanoparticles functionalized with cancer antigen induce depletion of T cells. RSC Adv 2020; 10:35198-35205. [PMID: 35515680 PMCID: PMC9056825 DOI: 10.1039/d0ra05900e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Erythrocyte-based drug delivery systems have been investigated for their biocompatibility, long circulation time, and capability to transport cargo all around the body, thus presenting enormous potential in medical applications. In this study, we investigated hybrid nanoparticles consisting of nano-sized autologous or allogeneic red blood cell (RBC) membranes encapsulating porous silicon nanoparticles (PSi NPs). These NPs were functionalized with a model cancer antigen TRP2, which was either expressed on the surface of the RBCs by a cell membrane-mimicking block copolymer polydimethylsiloxane-b-poly-2-methyl-2-oxazoline, or attached on the PSi NPs, thus hidden within the encapsulation. When in the presence of peripheral blood immune cells, these NPs resulted in apoptotic cell death of T cells, where the NPs having TRP2 within the encapsulation led to a stronger T cell deletion. The deletion of the T cells did not change the relative proportion of CD4+ and cytotoxic CD8+ T cells. Overall, this work shows the combination of nano-sized RBCs, PSi, and antigenic peptides may have use in the treatment of autoimmune diseases. We report a study on the effect of red blood cell membrane based cancer antigen-functionalized nanoparticles on peripheral blood T cells. These nanoparticles induce apoptosis of T cells and they may have use in treating autoimmune diseases.![]()
Collapse
Affiliation(s)
- Antti Rahikkala
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki FI-00014 Helsinki Finland
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki FI-00014 Helsinki Finland
| | - Tomás Bauleth-Ramos
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto Rua Alfredo Allen, 208 4200-135 Porto Portugal.,Instituto de Engenharia Biomédica (INEB), University of Porto Rua Alfredo Allen, 208 4200-135 Porto Portugal.,Instituto Ciências Biomédicas Abel Salazar (ICBAS), University of Porto Rua Jorge Viterbo 228 4150-180 Porto Portugal
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki FI-00014 Helsinki Finland
| | - Marianna Kemell
- Department of Chemistry, University of Helsinki FI-00014 Helsinki Finland
| | - Jani Seitsonen
- Nanomicroscopy Center, Aalto University FI-02150 Espoo Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics, University of Turku FI-20014 Turku Finland
| | - Bruno Sarmento
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde 4585-116 Gandra Portugal
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics, University of Turku FI-20014 Turku Finland
| | | | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki FI-00014 Helsinki Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki FI-00014 Helsinki Finland .,Helsinki Institute of Life Science (HiLIFE), University of Helsinki FI-00014 Helsinki Finland
| |
Collapse
|
43
|
Li R, Ng TS, Garlin MA, Weissleder R, Miller MA. Understanding the in vivo Fate of Advanced Materials by Imaging. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910369. [PMID: 38545084 PMCID: PMC10972611 DOI: 10.1002/adfm.201910369] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/09/2020] [Indexed: 11/13/2024]
Abstract
Engineered materials are ubiquitous in biomedical applications ranging from systemic drug delivery systems to orthopedic implants, and their actions unfold across multiple time- and length-scales. The efficacy and safety of biologics, nanomaterials, and macroscopic implants are all dictated by the same general principles of pharmacology as apply to small molecule drugs, comprising how the body affects materials (pharmacokinetics, PK) and conversely how materials affect the body (pharmacodynamics, PD). Imaging technologies play an increasingly insightful role in monitoring both of these processes, often simultaneously: translational macroscopic imaging modalities such as MRI and PET/CT offer whole-body quantitation of biodistribution and structural or molecular response, while ex vivo approaches and optical imaging via in vivo (intravital) microscopy reveal behaviors at subcellular resolution. In this review, the authors survey developments in imaging the in situ behavior of systemically and locally administered materials, with a particular focus on using microscopy to understand transport, target engagement, and downstream host responses at a single-cell level. The themes of microenvironmental influence, controlled drug release, on-target molecular action, and immune response, especially as mediated by macrophages and other myeloid cells are examined. Finally, the future directions of how new imaging technologies may propel efficient clinical translation of next-generation therapeutics and medical devices are proposed.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Michelle A. Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
- Department of Systems Biology, Harvard Medical School
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
44
|
Zhang DX, Esser L, Vasani RB, Thissen H, Voelcker NH. Porous silicon nanomaterials: recent advances in surface engineering for controlled drug-delivery applications. Nanomedicine (Lond) 2020; 14:3213-3230. [PMID: 31855121 DOI: 10.2217/nnm-2019-0167] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Porous silicon (pSi) nanomaterials are increasingly attractive for biomedical applications due to their promising properties such as simple and feasible fabrication procedures, tunable morphology, versatile surface modification routes, biocompatibility and biodegradability. This review focuses on recent advances in surface modification of pSi for controlled drug delivery applications. A range of functionalization strategies and fabrication methods for pSi-polymer hybrids are summarized. Surface engineering solutions such as stimuli-responsive polymer grafting, stealth coatings and active targeting modifications are highlighted as examples to demonstrate what can be achieved. Finally, the current status of engineered pSi nanomaterials for in vivo applications is reviewed and future prospects and challenges in drug-delivery applications are discussed.
Collapse
Affiliation(s)
- De-Xiang Zhang
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.,Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Lars Esser
- Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Roshan B Vasani
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Helmut Thissen
- Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.,Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia
| |
Collapse
|
45
|
Mateos S, Lifante J, Li C, Ximendes EC, Muñoz-Ortiz T, Yao J, de la Fuente-Fernández M, García Villalón ÁL, Granado M, Zabala Gutierrez I, Rubio-Retama J, Jaque D, Ortgies DH, Fernández N. Instantaneous In Vivo Imaging of Acute Myocardial Infarct by NIR-II Luminescent Nanodots. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907171. [PMID: 32548926 DOI: 10.1002/smll.201907171] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 04/28/2020] [Indexed: 06/11/2023]
Abstract
Fast and precise localization of ischemic tissues in the myocardium after an acute infarct is required by clinicians as the first step toward accurate and efficient treatment. Nowadays, diagnosis of a heart attack at early times is based on biochemical blood analysis (detection of cardiac enzymes) or by ultrasound-assisted imaging. Alternative approaches are investigated to overcome the limitations of these classical techniques (time-consuming procedures or low spatial resolution). As occurs in many other fields of biomedicine, cardiological preclinical imaging can also benefit from the fast development of nanotechnology. Indeed, bio-functionalized near-infrared-emitting nanoparticles are herein used for in vivo imaging of the heart after an acute myocardial infarct. Taking advantage of the superior acquisition speed of near-infrared fluorescence imaging, and of the efficient selective targeting of the near-infrared-emitting nanoparticles, in vivo images of the infarcted heart are obtained only a few minutes after the acute infarction event. This work opens an avenue toward cost-effective, fast, and accurate in vivo imaging of the ischemic myocardium after an acute infarct.
Collapse
Affiliation(s)
- Sergio Mateos
- Departamento de Fisiología - Facultad de Medicina, Fluorescence Imaging Group, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid, 28029, Spain
| | - José Lifante
- Departamento de Fisiología - Facultad de Medicina, Fluorescence Imaging Group, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid, 28029, Spain
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid, 28034, Spain
| | - Chunyan Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230036, China
| | - Erving C Ximendes
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid, 28034, Spain
- Departamento de Física de Materiales - Facultad de Ciencias, Fluorescence Imaging Group, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente 7, Madrid, 28049, Spain
| | - Tamara Muñoz-Ortiz
- Departamento de Física de Materiales - Facultad de Ciencias, Fluorescence Imaging Group, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente 7, Madrid, 28049, Spain
| | - Jingke Yao
- Departamento de Física de Materiales - Facultad de Ciencias, Fluorescence Imaging Group, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente 7, Madrid, 28049, Spain
| | - María de la Fuente-Fernández
- Departamento de Fisiología - Facultad de Medicina, Fluorescence Imaging Group, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid, 28029, Spain
| | - Ángel Luis García Villalón
- Departamento de Fisiología - Facultad de Medicina, Fluorescence Imaging Group, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid, 28029, Spain
| | - Miriam Granado
- Departamento de Fisiología - Facultad de Medicina, Fluorescence Imaging Group, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid, 28029, Spain
| | - Irene Zabala Gutierrez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Plaza de Ramón y Cajal, s/n, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Jorge Rubio-Retama
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid, 28034, Spain
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Plaza de Ramón y Cajal, s/n, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Daniel Jaque
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid, 28034, Spain
- Departamento de Física de Materiales - Facultad de Ciencias, Fluorescence Imaging Group, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente 7, Madrid, 28049, Spain
| | - Dirk H Ortgies
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid, 28034, Spain
- Departamento de Física de Materiales - Facultad de Ciencias, Fluorescence Imaging Group, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente 7, Madrid, 28049, Spain
| | - Nuria Fernández
- Departamento de Fisiología - Facultad de Medicina, Fluorescence Imaging Group, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid, 28029, Spain
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid, 28034, Spain
| |
Collapse
|
46
|
Fan C, Joshi J, Li F, Xu B, Khan M, Yang J, Zhu W. Nanoparticle-Mediated Drug Delivery for Treatment of Ischemic Heart Disease. Front Bioeng Biotechnol 2020; 8:687. [PMID: 32671049 PMCID: PMC7326780 DOI: 10.3389/fbioe.2020.00687] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/02/2020] [Indexed: 12/26/2022] Open
Abstract
The regenerative capacity of an adult cardiac tissue is insufficient to repair the massive loss of heart tissue, particularly cardiomyocytes (CMs), following ischemia or other catastrophic myocardial injuries. The delivery methods of therapeutics agents, such as small molecules, growth factors, exosomes, cells, and engineered tissues have significantly advanced in medical science. Furthermore, with the controlled release characteristics, nanoparticle (NP) systems carrying drugs are promising in enhancing the cardioprotective potential of drugs in patients with cardiac ischemic events. NPs can provide sustained exposure precisely to the infarcted heart via direct intramyocardial injection or intravenous injection with active targets. In this review, we present the recent advances and challenges of different types of NPs loaded with agents for the repair of myocardial infarcted heart tissue.
Collapse
Affiliation(s)
- Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jyotsna Joshi
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States
| | - Fan Li
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States
| | - Bing Xu
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States
| | - Mahmood Khan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
47
|
Pala R, Anju VT, Dyavaiah M, Busi S, Nauli SM. Nanoparticle-Mediated Drug Delivery for the Treatment of Cardiovascular Diseases. Int J Nanomedicine 2020; 15:3741-3769. [PMID: 32547026 PMCID: PMC7266400 DOI: 10.2147/ijn.s250872] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the foremost causes of high morbidity and mortality globally. Preventive, diagnostic, and treatment measures available for CVDs are not very useful, which demands promising alternative methods. Nanoscience and nanotechnology open a new window in the area of CVDs with an opportunity to achieve effective treatment, better prognosis, and less adverse effects on non-target tissues. The application of nanoparticles and nanocarriers in the area of cardiology has gathered much attention due to the properties such as passive and active targeting to the cardiac tissues, improved target specificity, and sensitivity. It has reported that more than 50% of CVDs can be treated effectively through the use of nanotechnology. The main goal of this review is to explore the recent advancements in nanoparticle-based cardiovascular drug carriers. This review also summarizes the difficulties associated with the conventional treatment modalities in comparison to the nanomedicine for CVDs.
Collapse
Affiliation(s)
- Rajasekharreddy Pala
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA92618, USA
- Department of Medicine, University of California Irvine, Irvine, CA92868, USA
| | - V T Anju
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Madhu Dyavaiah
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA92618, USA
- Department of Medicine, University of California Irvine, Irvine, CA92868, USA
| |
Collapse
|
48
|
GATA4-targeted compound exhibits cardioprotective actions against doxorubicin-induced toxicity in vitro and in vivo: establishment of a chronic cardiotoxicity model using human iPSC-derived cardiomyocytes. Arch Toxicol 2020; 94:2113-2130. [PMID: 32185414 PMCID: PMC7303099 DOI: 10.1007/s00204-020-02711-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Doxorubicin is a widely used anticancer drug that causes dose-related cardiotoxicity. The exact mechanisms of doxorubicin toxicity are still unclear, partly because most in vitro studies have evaluated the effects of short-term high-dose doxorubicin treatments. Here, we developed an in vitro model of long-term low-dose administration of doxorubicin utilizing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Moreover, given that current strategies for prevention and management of doxorubicin-induced cardiotoxicity fail to prevent cancer patients developing heart failure, we also investigated whether the GATA4-targeted compound 3i-1000 has cardioprotective potential against doxorubicin toxicity both in vitro and in vivo. The final doxorubicin concentration used in the chronic toxicity model in vitro was chosen based on cell viability data evaluation. Exposure to doxorubicin at the concentrations of 1–3 µM markedly reduced (60%) hiPSC-CM viability already within 48 h, while a 14-day treatment with 100 nM doxorubicin concentration induced only a modest 26% reduction in hiPCS-CM viability. Doxorubicin treatment also decreased DNA content in hiPSC-CMs. Interestingly, the compound 3i-1000 attenuated doxorubicin-induced increase in pro-B-type natriuretic peptide (proBNP) expression and caspase-3/7 activation in hiPSC-CMs. Moreover, treatment with 3i-1000 for 2 weeks (30 mg/kg/day, i.p.) inhibited doxorubicin cardiotoxicity by restoring left ventricular ejection fraction and fractional shortening in chronic in vivo rat model. In conclusion, the results demonstrate that long-term exposure of hiPSC-CMs can be utilized as an in vitro model of delayed doxorubicin-induced toxicity and provide in vitro and in vivo evidence that targeting GATA4 may be an effective strategy to counteract doxorubicin-induced cardiotoxicity.
Collapse
|
49
|
Zanjanizadeh Ezazi N, Ajdary R, Correia A, Mäkilä E, Salonen J, Kemell M, Hirvonen J, Rojas OJ, Ruskoaho HJ, Santos HA. Fabrication and Characterization of Drug-Loaded Conductive Poly(glycerol sebacate)/Nanoparticle-Based Composite Patch for Myocardial Infarction Applications. ACS APPLIED MATERIALS & INTERFACES 2020; 12:6899-6909. [PMID: 31967771 PMCID: PMC7450488 DOI: 10.1021/acsami.9b21066] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Heart tissue engineering is critical in the treatment of myocardial infarction, which may benefit from drug-releasing smart materials. In this study, we load a small molecule (3i-1000) in new biodegradable and conductive patches for application in infarcted myocardium. The composite patches consist of a biocompatible elastomer, poly(glycerol sebacate) (PGS), coupled with collagen type I, used to promote cell attachment. In addition, polypyrrole is incorporated because of its electrical conductivity and to induce cell signaling. Results from the in vitro experiments indicate a high density of cardiac myoblast cells attached on the patches, which stay viable for at least 1 month. The degradation of the patches does not show any cytotoxic effect, while 3i-1000 delivery induces cell proliferation. Conductive patches show high blood wettability and drug release, correlating with the rate of degradation of the PGS matrix. Together with the electrical conductivity and elongation characteristics, the developed biomaterial fits the mechanical, conductive, and biological demands required for cardiac treatment.
Collapse
Affiliation(s)
- Nazanin Zanjanizadeh Ezazi
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Rubina Ajdary
- Department of Bioproducts and Biosystems, School of Chemical
Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo, Finland
| | - Alexandra Correia
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Marianna Kemell
- Department of Chemistry, University of
Helsinki, FI-00014 Helsinki, Finland
| | - Jouni Hirvonen
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Orlando J. Rojas
- Department of Bioproducts and Biosystems, School of Chemical
Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo, Finland
- Departments of Chemical
& Biological Engineering, Chemistry, and Wood Science, The University of British Columbia, 2360 East Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hélder A. Santos
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
- E-mail: .
Tel: +358 2941 59661
| |
Collapse
|
50
|
Torrieri G, Fontana F, Figueiredo P, Liu Z, Ferreira MPA, Talman V, Martins JP, Fusciello M, Moslova K, Teesalu T, Cerullo V, Hirvonen J, Ruskoaho H, Balasubramanian V, Santos HA. Dual-peptide functionalized acetalated dextran-based nanoparticles for sequential targeting of macrophages during myocardial infarction. NANOSCALE 2020; 12:2350-2358. [PMID: 31930241 DOI: 10.1039/c9nr09934d] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The advent of nanomedicine has recently started to innovate the treatment of cardiovascular diseases, in particular myocardial infarction. Although current approaches are very promising, there is still an urgent need for advanced targeting strategies. In this work, the exploitation of macrophage recruitment is proposed as a novel and synergistic approach to improve the addressability of the infarcted myocardium achieved by current peptide-based heart targeting strategies. For this purpose, an acetalated dextran-based nanosystem is designed and successfully functionalized with two different peptides, atrial natriuretic peptide (ANP) and linTT1, which target, respectively, cardiac cells and macrophages associated with atherosclerotic plaques. The biocompatibility of the nanocarrier is screened on both macrophage cell lines and primary macrophages, showing high safety, in particular after functionalization of the nanoparticles' surface. Furthermore, the system shows higher association versus uptake ratio towards M2-like macrophages (approximately 2-fold and 6-fold increase in murine and human primary M2-like macrophages, respectively, compared to M1-like). Overall, the results demonstrate that the nanosystem has potential to exploit the "hitchhike" effect on M2-like macrophages and potentially improve, in a dual targeting strategy, the ability of the ANP peptide to target infarcted heart.
Collapse
Affiliation(s)
- Giulia Torrieri
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Patrícia Figueiredo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Mónica P A Ferreira
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Virpi Talman
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00140, Helsinki, Finland and National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - João P Martins
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Manlio Fusciello
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00140, Helsinki, Finland
| | - Karina Moslova
- Department of Chemistry, University of Helsinki, FI-00014, Helsinki, Finland
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Tartu, 50411, Estonia and Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - Vincenzo Cerullo
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00140, Helsinki, Finland and Helsinki Institute of Life Science, HiLIFE, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00140, Helsinki, Finland
| | - Vimalkumar Balasubramanian
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Helsinki Institute of Life Science, HiLIFE, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|