1
|
Rocher EE, Luly KM, Tzeng SY, Sunshine JC, Green JJ. Efficient Polymeric Nanoparticle Gene Delivery Enabled Via Tri- and Tetrafunctional Branching. Biomacromolecules 2024; 25:7260-7273. [PMID: 39466232 DOI: 10.1021/acs.biomac.4c00954] [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: 10/29/2024]
Abstract
Poly(β-amino ester) (PBAE) nanoparticles (NPs) show great promise for nonviral gene delivery. Recent studies suggest branched PBAEs (BPBAEs) offer advantages over linear counterparts, but the effect of polymer structure has not been well investigated across many chemical constituents. Here, a library of BPBAEs was synthesized with tri- and tetrafunctional branching. These polymers self-assemble with DNA to form highly cationic, monodisperse NPs with notably small size (∼50 nm). Optimal transfection occurred with polymer structures that featured moderate PBAE branching, enabling complete DNA encapsulation, rapid NP uptake, and robust expression at low DNA doses and polymer amounts. Optimized NPs enabled efficient DNA delivery to diverse cell types in vitro while maintaining high cellular viability, demonstrating significant improvements over a well-performing linear PBAE counterpart. BPBAEs also facilitated efficient mRNA and siRNA delivery, highlighting the versatility of these structures and demonstrating the broad utility of BPBAE NPs as vectors for nucleic acid delivery.
Collapse
|
2
|
Cong X, Zhang Z, Li H, Yang YG, Zhang Y, Sun T. Nanocarriers for targeted drug delivery in the vascular system: focus on endothelium. J Nanobiotechnology 2024; 22:620. [PMID: 39396002 PMCID: PMC11470712 DOI: 10.1186/s12951-024-02892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
Endothelial cells (ECs) are pivotal in maintaining vascular health, regulating hemodynamics, and modulating inflammatory responses. Nanocarriers hold transformative potential for precise drug delivery within the vascular system, particularly targeting ECs for therapeutic purposes. However, the complex interactions between vascular ECs and nanocarriers present significant challenges for the development and clinical translation of nanotherapeutics. This review assesses recent advancements and key strategies in employing nanocarriers for drug delivery to vascular ECs. It suggested that through precise physicochemical design and surface modifications, nanocarriers can enhance targeting specificity and improve drug internalization efficiency in ECs. Additionally, we elaborated on the applications of nanocarriers specifically designed for targeting ECs in the treatment of cardiovascular diseases, cancer metastasis, and inflammatory disorders. Despite these advancements, safety concerns, the complexity of in vivo processes, and the challenge of achieving subcellular drug delivery remain significant obstacles to the effective targeting of ECs with nanocarriers. A comprehensive understanding of endothelial cell biology and its interaction with nanocarriers is crucial for realizing the full potential of targeted drug delivery systems.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - Zebin Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, 130012, Jilin, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China.
| |
Collapse
|
3
|
Chen B, Ren Q, Jiang P, Wu Q, Shuai Q, Yan Y. Combinatorial Synthesis of Alkyl Chain-Capped Poly(β-Amino Ester)s for Effective siRNA Delivery. Macromol Biosci 2024; 24:e2400168. [PMID: 39052313 DOI: 10.1002/mabi.202400168] [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: 04/08/2024] [Revised: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Poly (β-amino ester) (PBAE) is a class of biodegradable polymers containing ester bonds in their main chain, extensively investigated as cationic polymer carriers for siRNA. Most current PBAE carriers rely on termination with hydrophilic or charged amines. In this study, a polymer platform consisting of 168 PBAE polymers with hydrophobic alkyl chain terminals is constructed through sequential aza-Michael addition. A large number of effective carriers are identified through in vitro screening of the PBAE platform for siLuc delivery to HeLa-Luc cells. Specifically, PA8-C6 and PA8-C8 achieve remarkable gene knockdown efficacies of up to 80% with low cytotoxicity. Certain materials from the PA2 and PA5 series demonstrate potent siRNA delivery capabilities associated with elevated cytotoxicity. The pKa value of PBAE is predominantly determined by the hydrophilic amine side chains rather than the end-capping groups. A pKa range of ≈6.2-6.5 may contribute to the excellent delivery capability for PA8 series carriers. The co-formulation of PBAE carriers with helper lipids leads to the reduced size and surface charges of the polyplex NPs with siRNA, consequently decreasing the cytotoxicity and enhancing siRNA delivery efficacy. These findings hold significant implications for the development of novel degradable polymer carriers for siRNA delivery.
Collapse
Affiliation(s)
- Baiqiu Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Pingge Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qiong Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qi Shuai
- College of Pharmaceutical Sciences and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
4
|
Dastgerdi NK, Dastgerdi NK, Bayraktutan H, Costabile G, Atyabi F, Dinarvand R, Longobardi G, Alexander C, Conte C. Enhancing siRNA cancer therapy: Multifaceted strategies with lipid and polymer-based carrier systems. Int J Pharm 2024; 663:124545. [PMID: 39098747 DOI: 10.1016/j.ijpharm.2024.124545] [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: 01/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Cancers are increasing in prevalence and many challenges remain for their treatment, such as chemoresistance and toxicity. In this context, siRNA-based therapeutics have many potential advantages for cancer therapies as a result of their ability to reduce or prevent expression of specific cancer-related genes. However, the direct delivery of naked siRNA is hindered by issues like enzymatic degradation, insufficient cellular uptake, and poor pharmacokinetics. Hence, the discovery of a safe and efficient delivery vehicle is essential. This review explores various lipid and polymer-based delivery systems for siRNA in cancer treatment. Both polymers and lipids have garnered considerable attention as carriers for siRNA delivery. While all of these systems protect siRNA and enhance transfection efficacy, each exhibits its unique strengths. Lipid-based delivery systems, for instance, demonstrate high entrapment efficacy and utilize cost-effective materials. Conversely, polymeric-based delivery systems offer advantages through chemical modifications. Nonetheless, certain drawbacks still limit their usage. To address these limitations, combining different materials in formulations (lipid, polymer, or targeting agent) could enhance pharmaceutical properties, boost transfection efficacy, and reduce side effects. Furthermore, co-delivery of siRNA with other therapeutic agents presents a promising strategy to overcome cancer resistance. Lipid-based delivery systems have been demonstrated to encapsulate many therapeutic agents and with high efficiency, but most are limited in terms of the functionalities they display. In contrast, polymeric-based delivery systems can be chemically modified by a wide variety of routes to include multiple components, such as release or targeting elements, from the same materials backbone. Accordingly, by incorporating multiple materials such as lipids, polymers, and/or targeting agents in RNA formulations it is possible to improve the pharmaceutical properties and therapeutic efficacy while reducing side effects. This review focuses on strategies to improve siRNA cancer treatments and discusses future prospects in this important field.
Collapse
Affiliation(s)
- Nazgol Karimi Dastgerdi
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Karimi Dastgerdi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran.
| | | | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy.
| |
Collapse
|
5
|
Gil-Cabrerizo P, Simon-Yarza T, Garbayo E, Blanco-Prieto MJ. Navigating the landscape of RNA delivery systems in cardiovascular disease therapeutics. Adv Drug Deliv Rev 2024; 208:115302. [PMID: 38574952 DOI: 10.1016/j.addr.2024.115302] [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: 11/28/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Cardiovascular diseases (CVDs) stand as the leading cause of death worldwide, posing a significant global health challenge. Consequently, the development of innovative therapeutic strategies to enhance CVDs treatment is imperative. RNA-based therapies, encompassing non-coding RNAs, mRNA, aptamers, and CRISPR/Cas9 technology, have emerged as promising tools for addressing CVDs. However, inherent challenges associated with RNA, such as poor cellular uptake, susceptibility to RNase degradation, and capture by the reticuloendothelial system, underscore the necessity of combining these therapies with effective drug delivery systems. Various non-viral delivery systems, including extracellular vesicles, lipid-based carriers, polymeric and inorganic nanoparticles, as well as hydrogels, have shown promise in enhancing the efficacy of RNA therapeutics. In this review, we offer an overview of the most relevant RNA-based therapeutic strategies explored for addressing CVDs and emphasize the pivotal role of delivery systems in augmenting their effectiveness. Additionally, we discuss the current status of these therapies and the challenges that hinder their clinical translation.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France
| | - Elisa Garbayo
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| | - María J Blanco-Prieto
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| |
Collapse
|
6
|
Ooi YJ, Huang C, Lau K, Chew SY, Park JG, Chan-Park MB. Nontoxic, Biodegradable Hyperbranched Poly(β-amino ester)s for Efficient siRNA Delivery and Gene Silencing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:14093-14112. [PMID: 38449351 DOI: 10.1021/acsami.3c10620] [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: 03/08/2024]
Abstract
RNA interference (RNAi)-mediated gene silencing is a promising therapeutic approach to treat various diseases, but safe and efficient delivery remains a major challenge to its clinical application. Non-viral gene vectors, such as poly(β-amino esters) (pBAEs), have emerged as a potential candidate due to their biodegradability, low toxicity profile, ease of synthesis, and high gene transfection efficiency for both DNA and siRNA delivery. However, achieving significant gene silencing using pBAEs often requires a large amount of polymer carrier (with polymer/siRNA weight ratio >100) or high siRNA dose (>100 nM), which might potentially exacerbate toxicity concerns during delivery. To overcome these barriers, we designed and optimized a series of hyperbranched pBAEs capable of efficiently condensing siRNA and achieving excellent silencing efficiency at a lower polymer/siRNA weight ratio (w/w) and siRNA dose. Through modulation of monomer combinations and branching density, we identified the top-performing hyperbranched pBAEs, named as h(A2B3)-1, which possess good siRNA condensation ability, low cytotoxicity, and high cellular uptake efficiency. Compared with Lipofectamine 2000, h(A2B3)-1 achieved lower cytotoxicity and higher siRNA silencing efficiency in HeLa cells at a polymer/siRNA weight ratio of 30 and 30 nM siRNA dose. Notably, h(A2B3)-1 enhanced the gene uptake in primary neural cells and effectively silenced the target gene in hard-to-transfect primary cortical neurons and oligodendrocyte progenitor cells, with gene knockdown efficiencies of 34.8 and 53.4% respectively. By incorporating a bioreducible disulfide compartment into the polymer backbone, the cytocompatibility of the h(A2B3)-1 was greatly enhanced while maintaining their good transfection efficiency. Together, the low cytotoxicity and high siRNA transfection efficiency of hyperbranched h(A2B3)-1 in this study demonstrated their great potential as a non-viral gene vector for efficient siRNA delivery and RNAi-mediated gene silencing. This provides valuable insight into the future development of safe and efficient non-viral siRNA delivery systems as well as their translation into clinical applications.
Collapse
Affiliation(s)
- Ying Jie Ooi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Chongquan Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Neuroscience@NTU, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore 637459, Singapore
| | - Kieran Lau
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jong Gu Park
- Welgene Inc, 693, Namcheon-ro, Namcheon-myeon, Gyeongsan-si, Gyeongsangbuk-do 38695, Republic of Korea
| | - Mary B Chan-Park
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| |
Collapse
|
7
|
Ali Zaidi SS, Fatima F, Ali Zaidi SA, Zhou D, Deng W, Liu S. Engineering siRNA therapeutics: challenges and strategies. J Nanobiotechnology 2023; 21:381. [PMID: 37848888 PMCID: PMC10583313 DOI: 10.1186/s12951-023-02147-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
Small interfering RNA (siRNA) is a potential method of gene silencing to target specific genes. Although the U.S. Food and Drug Administration (FDA) has approved multiple siRNA-based therapeutics, many biological barriers limit their use for treating diseases. Such limitations include challenges concerning systemic or local administration, short half-life, rapid clearance rates, nonspecific binding, cell membrane penetration inability, ineffective endosomal escape, pH sensitivity, endonuclease degradation, immunological responses, and intracellular trafficking. To overcome these barriers, various strategies have been developed to stabilize siRNA, ensuring their delivery to the target site. Chemical modifications implemented with nucleotides or the phosphate backbone can reduce off-target binding and immune stimulation. Encapsulation or formulation can protect siRNA from endonuclease degradation and enhance cellular uptake while promoting endosomal escape. Additionally, various techniques such as viral vectors, aptamers, cell-penetrating peptides, liposomes, and polymers have been developed for delivering siRNA, greatly improving their bioavailability and therapeutic potential.
Collapse
Affiliation(s)
- Syed Saqib Ali Zaidi
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Faria Fatima
- College of Medical Technology, Ziauddin University, Karachi, 74700, Pakistan
| | - Syed Aqib Ali Zaidi
- Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, Chongqing Diabetic Foot Medical Research Center, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China.
| | - Shuai Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Jin Y, Adams F, Nguyen A, Sturm S, Carnerio S, Müller-Caspary K, Merkel OM. Synthesis and application of spermine-based amphiphilic poly(β-amino ester)s for siRNA delivery. NANOSCALE ADVANCES 2023; 5:5256-5262. [PMID: 37767040 PMCID: PMC10521211 DOI: 10.1039/d3na00272a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023]
Abstract
Small interfering RNA (siRNA) can trigger RNA interference (RNAi) to therapeutically silence disease-related genes in human cells. The approval of siRNA therapeutics by the FDA in recent years generated a new hope in novel and efficient siRNA therapeutics. However, their therapeutic application is still limited by the lack of safe and efficient transfection vehicles. In this study, we successfully synthesized a novel amphiphilic poly(β-amino ester) based on the polyamine spermine, hydrophobic decylamine and 1,4-butanediol diacrylate, which was characterized by 1H NMR spectroscopy and size exclusion chromatography (SEC, Mn = 6000 Da). The polymer encapsulated siRNA quantitatively from N/P 5 on as assessed by fluorescence intercalation while maintaining optimal polyplex sizes and zeta potentials. Biocompatibility and cellular delivery efficacy were also higher than those of the commonly used cationic, hyperbranched polymer polyethylenimine (PEI, 25 kDa). Optimized formulations mediated around 90% gene silencing in enhanced green fluorescence protein expressing H1299 cells (H1299-eGFP) as determined by flow cytometry. These results suggest that spermine-based, amphiphilic poly(β-amino ester)s are very promising candidates for efficient siRNA delivery.
Collapse
Affiliation(s)
- Yao Jin
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Friederike Adams
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Anny Nguyen
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Sebastian Sturm
- Department of Chemistry and Centre for NanoScience, Ludwig-Maximilians-University Munich Butenandtstr. 11 81377 Munich Germany
| | - Simone Carnerio
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Knut Müller-Caspary
- Department of Chemistry and Centre for NanoScience, Ludwig-Maximilians-University Munich Butenandtstr. 11 81377 Munich Germany
| | - Olivia M Merkel
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| |
Collapse
|
9
|
Recent advances in nanomedicines for imaging and therapy of myocardial ischemia-reperfusion injury. J Control Release 2023; 353:563-590. [PMID: 36496052 DOI: 10.1016/j.jconrel.2022.11.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Myocardial ischemia-reperfusion injury (IRI) is becoming a typical cardiovascular disease with increasing worldwide incidence. It is usually induced by the restoration of normal blood flow to the ischemic myocardium after a period of recanalization and directly leads to myocardial damage. Notably, the pathological mechanism of myocardial IRI is closely related to inflammation, oxidative stress, Ca2+ overload, and the opening of mitochondrial permeability transition pore channels. Therefore, monitoring of these changes and imaging lesions is a key to timely clinical diagnosis. Nanomedicines have shown great value in the diagnosis and treatment of myocardial IRI, with advantages including passive/active targeting, prolonged circulation, improved bioavailability, versatile carrier selection, and synergistic integration of different imaging and therapeutic agents in single particles with the same pharmaceutics. Because theranostic nanomedicines for myocardial IRI have advanced rapidly, we conduct an updated review on this topic. The special focus is on how to rationally design the nanomedicines to achieve optimal imaging and therapy. We hope this review would stimulate the interest of researchers with different backgrounds and expedite the development of nanomedicines for myocardial IRI.
Collapse
|
10
|
Liu GW, Guzman EB, Menon N, Langer RS. Lipid Nanoparticles for Nucleic Acid Delivery to Endothelial Cells. Pharm Res 2023; 40:3-25. [PMID: 36735106 PMCID: PMC9897626 DOI: 10.1007/s11095-023-03471-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Endothelial cells play critical roles in circulatory homeostasis and are also the gateway to the major organs of the body. Dysfunction, injury, and gene expression profiles of these cells can cause, or are caused by, prevalent chronic diseases such as diabetes, cardiovascular disease, and cancer. Modulation of gene expression within endothelial cells could therefore be therapeutically strategic in treating longstanding disease challenges. Lipid nanoparticles (LNP) have emerged as potent, scalable, and tunable carrier systems for delivering nucleic acids, making them attractive vehicles for gene delivery to endothelial cells. Here, we discuss the functions of endothelial cells and highlight some receptors that are upregulated during health and disease. Examples and applications of DNA, mRNA, circRNA, saRNA, siRNA, shRNA, miRNA, and ASO delivery to endothelial cells and their targets are reviewed, as well as LNP composition and morphology, formulation strategies, target proteins, and biomechanical factors that modulate endothelial cell targeting. Finally, we discuss FDA-approved LNPs as well as LNPs that have been tested in clinical trials and their challenges, and provide some perspectives as to how to surmount those challenges.
Collapse
Affiliation(s)
- Gary W Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Edward B Guzman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nandita Menon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Strand Therapeutics, MA, 02215, Boston, USA
| | - Robert S Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
11
|
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
|
12
|
Hou M, Wu X, Zhao Z, Deng Q, Chen Y, Yin L. Endothelial cell-targeting, ROS-ultrasensitive drug/siRNA co-delivery nanocomplexes mitigate early-stage neutrophil recruitment for the anti-inflammatory treatment of myocardial ischemia reperfusion injury. Acta Biomater 2022; 143:344-355. [PMID: 35189380 DOI: 10.1016/j.actbio.2022.02.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/30/2022] [Accepted: 02/15/2022] [Indexed: 12/13/2022]
Abstract
Neutrophils serve as a key contributor to the pathophysiology of myocardial ischemia reperfusion injury (MIRI), because the unregulated activation and infiltration of neutrophils lead to overwhelming inflammation in the myocardium to cause tissue damage. Herein, endothelial cell-targeting and reactive oxygen species (ROS)-ultrasensitive nanocomplexes (NCs) were developed to mediate efficient co-delivery of VCAM-1 siRNA (siVCAM-1) and dexamethasone (DXM), which cooperatively inhibited neutrophil recruitment by impeding neutrophil migration and adhesion. RPPT was first synthesized via crosslinking of PEI 600 with ditellurium followed by modification with PEG and the endothelial cell-targeting peptide cRGD. RPPT was allowed to envelope the DXM-loaded PLGA nanoparticles and condense the siVCAM-1. After systemic administration in rats experiencing MIRI, the cRGD-modified NCs efficiently targeted and entered the inflamed endothelial cells, wherein RPPT was sensitively degraded by over-produced ROS to trigger intracellular siVCAM-1 release and potentiate the VCAM-1 silencing efficiency. As a consequence of the complementary function of DXM and siVCAM-1, the NCs notably mitigated neutrophil infiltration into ischemic myocardium, provoking potent anti-inflammatory efficacy to reduce MIRI and recover cardiac function. The present study offers an effective approach for the controlled co-delivery of siRNA and drug cargoes, and it also highlights the importance of multi-dimensional manipulation of neutrophils in anti-inflammatory treatment. STATEMENT OF SIGNIFICANCE: The unregulated activation and infiltration of neutrophils lead to overwhelming inflammation in the myocardium after myocardial ischemia reperfusion injury (MIRI). Here, endothelial cell-targeting and ROS-ultrasensitive nanocomplexes (NCs), comprised of PLGA NPs decorated with cRGD-poly(ethylene glycol) (PEG)-modified, ditellurium-crosslinked PEI (RPPT), were developed to mediate efficient co-delivery of VCAM-1 siRNA (siVCAM-1) and dexamethasone (DXM). DXM and siVCAM-1 with complementary functions inhibited both the migration and adhesion of neutrophils, efficiently interventing the neutrophil recruitment and interrupting the self-amplified inflammation cascade in the injured myocardium. The molecular design of RPPT renders an effective example for constructing polymeric materials with high ROS sensitivity, and it resolves the critical dilemma related to polycation-mediated siRNA delivery, such as siRNA encapsulation versus release, and transfection efficiency versus toxicity.
Collapse
|
13
|
Zhu J, Guo M, Cui Y, Meng Y, Ding J, Zeng W, Zhou W. Surface Coating of Pulmonary siRNA Delivery Vectors Enabling Mucus Penetration, Cell Targeting, and Intracellular Radical Scavenging for Enhanced Acute Lung Injury Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:5090-5100. [PMID: 35060376 DOI: 10.1021/acsami.1c23069] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pulmonary delivery of anti-inflammatory siRNA presents a promising approach for localized therapy of acute lung injury (ALI), while polycationic vectors can be easily trapped by the negatively charged airway mucin glycoproteins and arbitrarily internalized by epithelial cells with nontargetability for immunological clearance. Herein, we report a material, the dopamine (DA)-grafted hyaluronic acid (HA-DA), coating on an anti-TNF-α vector to address these limitations. HA-DA was simply synthesized and facilely coated on poly(β-amino ester) (BP)-based siRNA vectors via electrostatic attraction. The resulting HA-DA/BP/siRNA displayed significantly enhanced mucus penetration, attributable to the charge screen effect of HA-DA and the bioadhesive nature of the grafting DA. After transmucosal delivery, the nanosystem could target diseased macrophages via CD44-mediated internalization and rapidly escape from endo/lysosomes through the proton sponge effect, resulting in effective TNF-α regulation. Meanwhile, DA modification endowed the coating material with robust antioxidative capability to scavenge a broad spectrum of reactive oxygen/nitrogen species (RONS), which protected the lung tissue from oxidative damage and synergized with anti-TNF-α to inhibit a cytokine storm. As a result, a remarkable amelioration of ALI was achieved in a lipopolysaccharide (LPS)-stimulated mice model. This study provides a multifunctional coating material to facilitate pulmonary drug delivery for the treatment of lung diseases.
Collapse
Affiliation(s)
- Jiaojiao Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Man Guo
- Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Yanhui Cui
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yingcai Meng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Academician Workstation, Changsha Medical University, Changsha 410219, China
| |
Collapse
|
14
|
Liu X, Ding F, Guo Y, Jiang K, Fu Y, Zhu L, Li M, Zhu X, Zhang C. Complexing the Pre-assembled Brush-like siRNA with Poly(β-amino ester) for Efficient Gene Silencing. ACS APPLIED BIO MATERIALS 2022; 5:1857-1867. [PMID: 35107256 DOI: 10.1021/acsabm.1c01182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Small interfering RNA (siRNA) has been emerging as a highly selective and effective pharmaceutics for treating broad classes of diseases. However, the practical application of siRNA agent is often hampered by its poor crossing of the cellular membrane barrier and ineffective releasing from endosome to cytoplasm, leading to low gene silencing efficacy for clinical purposes. Thus far, cationic lipid and polymer-based vectors have been extensively explored for gene delivery. Yet condensing the rigid and highly negatively charged siRNA duplex to form a stable complex vehicle usually requires a large load of cationic carriers, prone to raising the toxicity issue for delivery. Herein, we develop a simple strategy that can efficiently condense the siRNAs into nanoparticle vehicles for target gene regulation. In this approach, we first employ a DNA-grafted polycaprolactone (DNA-g-PCL) brush as template to organize the small rigid siRNAs into a large brush-like structure (siRNA-brush) through nucleic acid hybridization. Then, the siRNA-brush assembly is condensed by an ionizable and biodegradable polymer (poly(β-amino ester), PBAE) under acidic buffer condition to form a stable nanoparticle for siRNA delivery. Compared to the free siRNAs with poor complexing capability with PBAE, the large brush-like siRNA assemblies with more complicated topological architecture significantly promotes their electrostatic interaction with PBAE, enabling the formation of complexed nanoparticles at low weight ratio of polymer to siRNA. Additionally, PBAE/siRNA-brush complexes exhibit good biocompatibility and stability under physiological condition, as well as enhanced cellular internalization. When equipped with functional siRNAs, the obtained delivery system demonstrates excellent downregulation of target genes both in vitro and in vivo, through which the progression of hypertrophic scars can be retarded with negligible adverse effects in an xenografted mouse model.
Collapse
Affiliation(s)
- Xinlong Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fei Ding
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuanyuan Guo
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kai Jiang
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yucheng Fu
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lijuan Zhu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Ming Li
- Department of Dermatology, Institute of Dermatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
15
|
Wang Y, Hou M, Duan S, Zhao Z, Wu X, Chen Y, Yin L. Macrophage-targeting gene silencing orchestrates myocardial microenvironment remodeling toward the anti-inflammatory treatment of ischemia-reperfusion (IR) injury. Bioact Mater 2022; 17:320-333. [PMID: 35386446 PMCID: PMC8965030 DOI: 10.1016/j.bioactmat.2022.01.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/02/2022] [Accepted: 01/18/2022] [Indexed: 01/03/2023] Open
Abstract
Ischemia-reperfusion (IR) injury represents a major cause of myocardial dysfunction after infarction and thrombolytic therapy, and it is closely related to the free radical explosion and overwhelming inflammatory responses. Herein, macrophage-targeting nanocomplexes (NCs) are developed to mediate efficient co-delivery of siRNA against MOF (siMOF) and microRNA-21 (miR21) into myocardial macrophages, cooperatively orchestrating the myocardial microenvironment against IR injury. Bioreducible, branched poly(β-amino ester) (BPAE-SS) is designed to co-condense siMOF and miR21 into NCs in a multivalency-reinforced approach, and they are surface-decorated with carboxylated mannan (Man-COOH) to shield the positive surface charges and enhance the serum stability. The final MBSsm NCs are efficiently internalized by myocardial macrophages after systemic administration, wherein BPAE-SS is degraded into small segments by intracellular glutathione to promote the siMOF/miR21 release, finally provoking efficient gene silencing. Thus, cardiomyocyte protection and macrophage modulation are realized via the combined effects of ROS scavenging, inflammation inhibition, and autophagy attenuation, which ameliorates the myocardial microenvironment and restores the cardiac function via positive cellular crosstalk. This study renders promising solutions to address the multiple systemic barriers against in vivo nucleic acid delivery, and it also offers new options for IR injury by manipulating multiple reciprocal bio-reactions. Macrophage-targeting and reduction-dissociable NCs mediate efficient siMOF/miR21 co-delivery. siMOF and miR21 cooperatively inhibit ROS production, inflammation, and autophagy. siMOF and miR21 orchestrate microenvironment remodeling via cellular cross-talk. NCs mediate efficient treatment of myocardial ischemia reperfusion injury.
Collapse
|
16
|
Vasconcelos-Cardoso M, Girao H. A new weapon in the armamentarium to tackle inflammation associated with myocardial infarction. Rev Port Cardiol 2022; 41:207-208. [DOI: 10.1016/j.repc.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
17
|
Yan Y, Liu XY, Lu A, Wang XY, Jiang LX, Wang JC. Non-viral vectors for RNA delivery. J Control Release 2022; 342:241-279. [PMID: 35016918 PMCID: PMC8743282 DOI: 10.1016/j.jconrel.2022.01.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/13/2022]
Abstract
RNA-based therapy is a promising and potential strategy for disease treatment by introducing exogenous nucleic acids such as messenger RNA (mRNA), small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides (ASO) to modulate gene expression in specific cells. It is exciting that mRNA encoding the spike protein of COVID-19 (coronavirus disease 2019) delivered by lipid nanoparticles (LNPs) exhibits the efficient protection of lungs infection against the virus. In this review, we introduce the biological barriers to RNA delivery in vivo and discuss recent advances in non-viral delivery systems, such as lipid-based nanoparticles, polymeric nanoparticles, N-acetylgalactosamine (GalNAc)-siRNA conjugate, and biomimetic nanovectors, which can protect RNAs against degradation by ribonucleases, accumulate in specific tissue, facilitate cell internalization, and allow for the controlled release of the encapsulated therapeutics.
Collapse
Affiliation(s)
- Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiao-Yu Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiang-Yu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Lin-Xia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China..
| |
Collapse
|
18
|
Hyun J, Eom J, Song J, Seo I, Um SH, Park KM, Bhang SH. Poly(amino ester)-Based Polymers for Gene and Drug Delivery Systems and Further Application toward Cell Culture System. Macromol Biosci 2021; 21:e2100106. [PMID: 34117832 DOI: 10.1002/mabi.202100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/20/2021] [Indexed: 11/10/2022]
Abstract
Various synthetic polymers based on poly(amino ester) (PAE) are suggested as candidates for gene and drug delivery owing to their pH-responsiveness, which contributes to efficient delivery performance. PAE-based pH-responsive polymers are more biodegradable and hydrophilic than other types of pH-responsive polymers. The functionality of PAE-based polymers can be reinforced by using different chemical modifications to improve the efficiency of gene and drug delivery. Additionally, PAE-based polymers are used in many ways in the biomedical field, such as in transdermal delivery and stem cell culture systems. Here, the recent novel PAE-based polymers designed for gene and drug delivery systems along with their further applications toward adult stem cell culture systems are reviewed. The synthetic tactics are contemplated and pros and cons of each type of polymer are analyzed, and detailed examples of the different types are analyzed.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jiin Eom
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihun Song
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Inwoo Seo
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyung Min Park
- Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
19
|
Tejedor S, Dolz‐Pérez I, Decker CG, Hernándiz A, Diez JL, Álvarez R, Castellano D, García NA, Ontoria‐Oviedo I, Nebot VJ, González‐King H, Igual B, Sepúlveda P, Vicent MJ. Polymer Conjugation of Docosahexaenoic Acid Potentiates Cardioprotective Therapy in Preclinical Models of Myocardial Ischemia/Reperfusion Injury. Adv Healthc Mater 2021; 10:e2002121. [PMID: 33720548 DOI: 10.1002/adhm.202002121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/16/2021] [Indexed: 01/16/2023]
Abstract
While coronary angioplasty represents an effective treatment option following acute myocardial infarction, the reperfusion of the occluded coronary artery can prompt ischemia-reperfusion (I/R) injury that significantly impacts patient outcomes. As ω-3 polyunsaturated fatty acids (PUFAs) have proven, yet limited cardioprotective abilities, an optimized polymer-conjugation approach is reported that improves PUFAs bioavailability to enhance cardioprotection and recovery in animal models of I/R-induced injury. Poly-l-glutamic acid (PGA) conjugation improves the solubility and stability of di-docosahexaenoic acid (diDHA) under physiological conditions and protects rat neonatal ventricular myocytes from I/R injury by reducing apoptosis, attenuating autophagy, inhibiting reactive oxygen species generation, and restoring mitochondrial membrane potential. Enhanced protective abilities are associated with optimized diDHA loading and evidence is provided for the inherent cardioprotective potential of PGA itself. Pretreatment with PGA-diDHA before reperfusion in a small animal I/R model provides for cardioprotection and limits area at risk (AAR). Furthermore, the preliminary findings suggest that PGA-diDHA administration in a swine I/R model may provide cardioprotection, limit edema and decrease AAR. Overall, the evaluation of PGA-diDHA in relevant preclinical models provides evidence for the potential of polymer-conjugated PUFAs in the mitigation of I/R injury associated with coronary angioplasty.
Collapse
Affiliation(s)
- Sandra Tejedor
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Irene Dolz‐Pérez
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Caitlin G. Decker
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Amparo Hernándiz
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Jose L. Diez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Raquel Álvarez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Delia Castellano
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Nahuel A. García
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Imelda Ontoria‐Oviedo
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Vicent J. Nebot
- Polypeptide Therapeutic Solutions S.L. Av. Benjamin Franklin 19, Paterna Valencia 46980 Spain
| | - Hernán González‐King
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Begoña Igual
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| |
Collapse
|
20
|
Wang X, Su B, Gao B, Zhou J, Ren XK, Guo J, Xia S, Zhang W, Feng Y. Cascaded bio-responsive delivery of eNOS gene and ZNF580 gene to collaboratively treat hindlimb ischemia via pro-angiogenesis and anti-inflammation. Biomater Sci 2020; 8:6545-6560. [DOI: 10.1039/d0bm01573c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cascaded, bio-responsively delivered eNOS gene and ZNF580 gene overcome transfection bottlenecks and collaboratively exert anti-ischemic function via promoting angiogenesis and alleviating inflammation.
Collapse
Affiliation(s)
- Xiaoyu Wang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
| | - Bin Su
- Department of Clinical Research
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- P. R. China
| | - Bin Gao
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
| | - Jiaying Zhou
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Xiang-kui Ren
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Jintang Guo
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine
- Affiliated Hospital
- Logistics University of People's Armed Police Force
- Tianjin 300162
- P. R. China
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology
- Logistics University of People's Armed Police Force
- Tianjin 300162
- P. R. China
| | - Yakai Feng
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| |
Collapse
|