1
|
Yadav K, Ebenezer Gnanakani SP, Kumar Sahu K, Sucheta, Dubey A, Minz S, Raza W, Pradhan M. Unleashing the potential of natural protein based nanoparticles for the delivery of therapeutic nucleic Acid: A comprehensive review. Int J Pharm 2025; 669:125049. [PMID: 39674384 DOI: 10.1016/j.ijpharm.2024.125049] [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: 05/14/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Nucleic acid-based therapeutics represent a revolutionary approach in treating genetic disorders, offering unprecedented potential for addressing pathologies at their molecular level. However, effective cellular delivery remains a critical challenge hindering their clinical implementation. While existing delivery systems, including viral vectors and lipid nanoparticles, have shown utility, they face limitations in immunogenicity, cargo capacity, and manufacturing complexity. Natural protein-based nanoparticles, derived from proteins such as albumin, ferritin, and elastin, have emerged as promising alternative delivery systems. These carriers offer distinct advantages including reduced immunogenicity, enhanced biocompatibility, and optimal biodegradation profiles. Their engineerable nature enables precise control over particle size, surface charge, and ligand conjugation, facilitating selective cellular targeting and improved pharmacokinetics. Recent technological advances have expanded the application of protein nanoparticles across various nucleic acid modalities, including mRNA, siRNA, and plasmid DNA. Extensive research has characterized these systems through rigorous in vitro and in vivo studies, advancing our understanding of their biological behavior and clinical potential. Advanced engineering methodologies have further enhanced their optimization for specific therapeutic applications. This review examines the development and potential of protein-based nanoparticles in nucleic acid delivery, highlighting their advantages and addressing current challenges. By analyzing recent advances and clinical progress, we underscore their significant potential to enhance the safety, specificity, and efficacy of nucleic acid therapeutics, potentially revolutionizing the treatment of genetic disorders.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kurud Road, Kohka, Bhilai 490024, Chhattisgarh, India
| | - S Princely Ebenezer Gnanakani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Limda, Waghodia, Vadodara, Gujarat 391760, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru 575018, Karnataka, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Wasim Raza
- Central Laboratory Facility, Chhattisgarh Council of Science and Technology, Vigyan Bhawan, Raipur, Chhattisgarh, India
| | | |
Collapse
|
2
|
Wang W, Tai S, Tao J, Yang L, Cheng X, Zhou J. Innovative hydrogel-based therapies for ischemia-reperfusion injury: bridging the gap between pathophysiology and treatment. Mater Today Bio 2024; 29:101295. [PMID: 39493810 PMCID: PMC11528235 DOI: 10.1016/j.mtbio.2024.101295] [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: 07/01/2024] [Revised: 09/21/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) commonly occurs in clinical settings, particularly in medical practices such as organ transplantation, cardiopulmonary resuscitation, and recovery from acute trauma, posing substantial challenges in clinical therapies. Current systemic therapies for IRI are limited by poor drug targeting, short efficacy, and significant side effects. Owing to their exceptional biocompatibility, biodegradability, excellent mechanical properties, targeting capabilities, controlled release potential, and properties mimicking the extracellular matrix (ECM), hydrogels not only serve as superior platforms for therapeutic substance delivery and retention, but also facilitate bioenvironment cultivation and cell recruitment, demonstrating significant potential in IRI treatment. This review explores the pathological processes of IRI and discusses the roles and therapeutic outcomes of various hydrogel systems. By categorizing hydrogel systems into depots delivering therapeutic agents, scaffolds encapsulating mesenchymal stem cells (MSCs), and ECM-mimicking hydrogels, this article emphasizes the selection of polymers and therapeutic substances, and details special crosslinking mechanisms and physicochemical properties, as well as summarizes the application of hydrogel systems for IRI treatment. Furthermore, it evaluates the limitations of current hydrogel treatments and suggests directions for future clinical applications.
Collapse
Affiliation(s)
- Weibo Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Supeng Tai
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Lexing Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Xi Cheng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
3
|
Pal P, Sharma M, Gupta SK, Potdar MB, Belgamwar AV. miRNA-124 loaded extracellular vesicles encapsulated within hydrogel matrices for combating chemotherapy-induced neurodegeneration. Biochem Biophys Res Commun 2024; 734:150778. [PMID: 39368371 DOI: 10.1016/j.bbrc.2024.150778] [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: 06/16/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Chemotherapy-induced neurodegeneration represents a significant challenge in cancer survivorship, manifesting in cognitive impairments that severely affect patients' quality of life. Emerging neuroregenerative therapies offer promise in mitigating these adverse effects, with miRNA-124 playing a pivotal role due to its critical functions in neural differentiation, neurogenesis, and neuroprotection. This review article delves into the innovative approach of using miRNA-124-loaded extracellular vesicles (EVs) encapsulated within hydrogel matrices as a targeted strategy for combating chemotherapy-induced neurodegeneration. We explore the biological underpinnings of miR-124 in neuroregeneration, detailing its mechanisms of action and therapeutic potential. The article further examines the roles and advantages of EVs as natural delivery systems for miRNAs and the application of hydrogel matrices in creating a sustained release environment conducive to neural tissue regeneration. By integrating these advanced materials and biological agents, we highlight a synergistic therapeutic strategy that leverages the bioactive properties of miR-124, the targeting capabilities of EVs, and the supportive framework of hydrogels. Preclinical studies and potential pathways to clinical translation are discussed, alongside the challenges, ethical considerations, and future directions in the field. This comprehensive review underscores the transformative potential of miR-124-loaded EVs in hydrogel matrices, offering insights into their development as a novel and integrative approach for addressing the complexities of chemotherapy-induced neurodegeneration.
Collapse
Affiliation(s)
- Pankaj Pal
- IIMT College of Pharmacy, IIMT Group of Colleges, Greater Noida, Uttar Pradesh, India.
| | - Monika Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| | - Sukesh Kumar Gupta
- Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA; KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India
| | - Mrugendra B Potdar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, Maharashtra, India
| | - Aarti V Belgamwar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, Maharashtra, India
| |
Collapse
|
4
|
Bains S, Giudicessi JR, Odening KE, Ackerman MJ. State of Gene Therapy for Monogenic Cardiovascular Diseases. Mayo Clin Proc 2024; 99:610-629. [PMID: 38569811 DOI: 10.1016/j.mayocp.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024]
Abstract
Over the past 2 decades, significant efforts have been made to advance gene therapy into clinical practice. Although successful examples exist in other fields, gene therapy for the treatment of monogenic cardiovascular diseases lags behind. In this review, we (1) highlight a brief history of gene therapy, (2) distinguish between gene silencing, gene replacement, and gene editing technologies, (3) discuss vector modalities used in the field with a special focus on adeno-associated viruses, (4) provide examples of gene therapy approaches in cardiomyopathies, channelopathies, and familial hypercholesterolemia, and (5) present current challenges and limitations in the gene therapy field.
Collapse
Affiliation(s)
- Sahej Bains
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN; Department of Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN
| | - John R Giudicessi
- Department of Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine (Division of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo Clinic, Rochester, MN
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Michael J Ackerman
- Department of Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine (Division of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo Clinic, Rochester, MN; Department of Pediatric and Adolescent Medicine (Division of Pediatric Cardiology), Mayo Clinic, Rochester, MN.
| |
Collapse
|
5
|
Zhang Z, Huang C, Guan S, Wang L, Yin H, Yin J, Liu J, Wu J. Hybrid gelatin-ascorbyl phosphate scaffolds accelerate diabetic wound healing via ROS scavenging, angiogenesis and collagen remodeling. BIOMATERIALS ADVANCES 2024; 158:213779. [PMID: 38277902 DOI: 10.1016/j.bioadv.2024.213779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/26/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Skin wound healing, particularly diabetic wound healing, is challenging in clinical management. Impaired wound healing is associated with persistent oxidative stress, altered inflammatory responses, unsatisfactory angiogenesis and epithelialization. Magnesium ascorbyl phosphate (MAP), which is an ascorbic acid derivative and active ingredient in cosmetics, has been reported to scavenge reactive oxygen species (ROS), and is considered a potential therapeutic agent for diabetic wounds. Herein, we report a hybrid gelatin-MAP scaffolds that can reduces oxidative stress damage, enhances angiogenesis and collagen remodeling to accelerate diabetic wound repair. Preliminary insights based on network pharmacology indicate that MAP may accelerate wound repair through multiple biological pathways, including extracellular matrix remodeling and anti-apoptosis. In vitro studies showed that the hybrid hydrogel scaffold had suitable mechanical properties, excellent biocompatibility and bioactivity. Further animal experiments demonstrated that the hydrogel accelerated full-thickness wound repair in diabetic mice (repair rate MAP vs Control=91.791±3.306 % vs 62.962±6.758 %) through antioxidant, neuroangiogenesis, collagen remodeling, and up-regulated the expression of the related factors COL-1, CD31, VEGF, and CGRP. Overall, we developed a bioactive hybrid hydrogel encapsulating MAP that synergistically promotes diabetic wound repair through multiple biological effects. This potentially integrated therapeutic scaffold may enrich future surgical approaches for treating diabetic wounds.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chunlin Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Shiyao Guan
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
| | - Liying Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Hanxiao Yin
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Junqiang Yin
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China; Division of Life Science, The Hong Kong University of Science and Technology, 999077, Hong Kong.
| |
Collapse
|
6
|
Browne S, Petit N, Quondamatteo F. Functionalised biomaterials as synthetic extracellular matrices to promote vascularisation and healing of diabetic wounds. Cell Tissue Res 2024; 395:133-145. [PMID: 38051351 DOI: 10.1007/s00441-023-03849-4] [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: 03/08/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023]
Abstract
Diabetic foot ulcers (DFU) are a type of chronic wound that constitute one of the most serious and debilitating complications associated with diabetes. The lack of clinically efficacious treatments to treat these recalcitrant wounds can lead to amputations for those worst affected. Biomaterial-based approaches offer great hope in this regard, as they provide a template for cell infiltration and tissue repair. However, there is an additional need to treat the underlying pathophysiology of DFUs, in particular insufficient vascularization of the wound which significantly hampers healing. Thus, the addition of pro-angiogenic moieties to biomaterials is a promising strategy to promote the healing of DFUs and other chronic wounds. In this review, we discuss the potential of biomaterials as treatments for DFU and the approaches that can be taken to functionalise these biomaterials such that they promote vascularisation and wound healing in pre-clinical models.
Collapse
Affiliation(s)
- Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland.
- CÚRAM, Centre for Research in Medical Devices, University of Galway, H91 W2TY, Galway, Ireland.
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland.
| | - Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland
| | - Fabio Quondamatteo
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland.
| |
Collapse
|
7
|
Kohar R, Ghosh M, Sawale JA, Singh A, Rangra NK, Bhatia R. Insights into Translational and Biomedical Applications of Hydrogels as Versatile Drug Delivery Systems. AAPS PharmSciTech 2024; 25:17. [PMID: 38253917 DOI: 10.1208/s12249-024-02731-y] [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: 07/26/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Hydrogels are a network of crosslinked polymers which can hold a huge amount of water in their matrix. These might be soft, flexible, and porous resembling living tissues. The incorporation of different biocompatible materials and nanostructures into the hydrogels has led to emergence of multifunctional hydrogels with advanced properties. There are broad applications of hydrogels such as tissue culture, drug delivery, tissue engineering, implantation, water purification, and dressings. Besides these, it can be utilized in the field of medical surgery, in biosensors, targeted drug delivery, and drug release. Similarly, hyaluronic acid hydrogels have vast applications in biomedicines such as cell delivery, drug delivery, molecule delivery, micropatterning in cellular biology for tissue engineering, diagnosis and screening of diseases, tissue repair and stem cell microencapsulation in case of inflammation, angiogenesis, and other biological developmental processes. The properties like swellability, de-swellability, biodegradability, biocompatibility, and inert nature of the hydrogels in contact with body fluids, blood, and tissues make its tremendous application in the field of modern biomedicines nowadays. Various modifications in hydrogel formulations have widened their therapeutic applicability. These include 3D printing, conjugation, thiolation, multiple anchoring, and reduction. Various hydrogel formulations are also capable of dual drug delivery, dental surgery, medicinal implants, bone diseases, and gene and stem cells delivery. The presented review summarizes the unique properties of hydrogels along with their methods of preparation and significant biomedical applications as well as different types of commercial products available in the market and the regulatory guidance.
Collapse
Affiliation(s)
- Ramesh Kohar
- Department of Pharmaceutical Analysis & Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Maitrayee Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Jyotiram A Sawale
- Department of Pharmacognosy, Krishna Institute of Pharmacy, Krishna Vishwa Vidyapeeth (Deemed to Be University), Karad, 415539, Maharashtra, India
| | - Amandeep Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Naresh Kumar Rangra
- Department of Pharmaceutical Analysis & Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Rohit Bhatia
- Department of Pharmaceutical Analysis & Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
8
|
Nottelet B, Buwalda S, van Nostrum CF, Zhao X, Deng C, Zhong Z, Cheah E, Svirskis D, Trayford C, van Rijt S, Ménard-Moyon C, Kumar R, Kehr NS, de Barros NR, Khademhosseini A, Kim HJ, Vermonden T. Roadmap on multifunctional materials for drug delivery. JPHYS MATERIALS 2024; 7:012502. [PMID: 38144214 PMCID: PMC10734278 DOI: 10.1088/2515-7639/ad05e8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/29/2023] [Accepted: 10/23/2023] [Indexed: 12/26/2023]
Abstract
This Roadmap on drug delivery aims to cover some of the most recent advances in the field of materials for drug delivery systems (DDSs) and emphasizes the role that multifunctional materials play in advancing the performance of modern DDSs in the context of the most current challenges presented. The Roadmap is comprised of multiple sections, each of which introduces the status of the field, the current and future challenges faced, and a perspective of the required advances necessary for biomaterial science to tackle these challenges. It is our hope that this collective vision will contribute to the initiation of conversation and collaboration across all areas of multifunctional materials for DDSs. We stress that this article is not meant to be a fully comprehensive review but rather an up-to-date snapshot of different areas of research, with a minimal number of references that focus upon the very latest research developments.
Collapse
Affiliation(s)
- Benjamin Nottelet
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
- Department of Pharmacy, Nîmes University Hospital, Univ Montpellier, 30900 Nimes, France
| | - Sytze Buwalda
- MINES Paris, PSL University, Center for Materials Forming, 06904 Sophia Antipolis, France
| | | | - Xiaofei Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People’s Republic of China
| | - Chao Deng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People’s Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People’s Republic of China
| | - Ernest Cheah
- School of Pharmacy, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Darren Svirskis
- School of Pharmacy, University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Chloe Trayford
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Sabine van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Cécilia Ménard-Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, 67000 Strasbourg, France
| | - Ravi Kumar
- Physikalisches Institute and Center of Soft Nanoscience, University of Münster, Münster, Germany
| | - Nermin Seda Kehr
- Physikalisches Institute and Center of Soft Nanoscience, University of Münster, Münster, Germany
- Department of Chemistry, Izmir Institute of Technology, Izmir, Turkey
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90274, United States of America
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90274, United States of America
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90274, United States of America
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Tina Vermonden
- Utrecht Institute for Pharmaceutical Sciences, Utrecht,The Netherlands
| |
Collapse
|
9
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Yadav K, Sahu KK, Sucheta, Gnanakani SPE, Sure P, Vijayalakshmi R, Sundar VD, Sharma V, Antil R, Jha M, Minz S, Bagchi A, Pradhan M. Biomedical applications of nanomaterials in the advancement of nucleic acid therapy: Mechanistic challenges, delivery strategies, and therapeutic applications. Int J Biol Macromol 2023; 241:124582. [PMID: 37116843 DOI: 10.1016/j.ijbiomac.2023.124582] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
In the past few decades, substantial advancement has been made in nucleic acid (NA)-based therapies. Promising treatments include mRNA, siRNA, miRNA, and anti-sense DNA for treating various clinical disorders by modifying the expression of DNA or RNA. However, their effectiveness is limited due to their concentrated negative charge, instability, large size, and host barriers, which make widespread application difficult. The effective delivery of these medicines requires safe vectors that are efficient & selective while having non-pathogenic qualities; thus, nanomaterials have become an attractive option with promising possibilities despite some potential setbacks. Nanomaterials possess ideal characteristics, allowing them to be tuned into functional bio-entity capable of targeted delivery. In this review, current breakthroughs in the non-viral strategy of delivering NAs are discussed with the goal of overcoming challenges that would otherwise be experienced by therapeutics. It offers insight into a wide variety of existing NA-based therapeutic modalities and techniques. In addition to this, it provides a rationale for the use of non-viral vectors and a variety of nanomaterials to accomplish efficient gene therapy. Further, it discusses the potential for biomedical application of nanomaterials-based gene therapy in various conditions, such as cancer therapy, tissue engineering, neurological disorders, and infections.
Collapse
Affiliation(s)
- Krishna Yadav
- Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | | | - Pavani Sure
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Sciences, Hyderabad, Telangana, India
| | - R Vijayalakshmi
- Department of Pharmaceutical Analysis, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - V D Sundar
- Department of Pharmaceutical Technology, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - Versha Sharma
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Ruchita Antil
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, England, United Kingdom of Great Britain and Northern Ireland
| | - Megha Jha
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, M.P., 484887, India
| | - Anindya Bagchi
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road La Jolla, CA 92037, USA
| | | |
Collapse
|
11
|
Fei Z, Gupta N, Li M, Xiao P, Hu X. Toward highly effective loading of DNA in hydrogels for high-density and long-term information storage. SCIENCE ADVANCES 2023; 9:eadg9933. [PMID: 37163589 PMCID: PMC10171811 DOI: 10.1126/sciadv.adg9933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Digital information, when converted into a DNA sequence, provides dense, stable, energy-efficient, and sustainable data storage. The most stable method for encapsulating DNA has been in an inorganic matrix of silica, iron oxide, or both, but are limited by low DNA uptake and complex recovery techniques. This study investigated a rationally designed thermally responsive functionally graded (TRFG) hydrogel as a simple and cost-effective method for storing DNA. The TRFG hydrogel shows high DNA uptake, long-term protection, and reusability due to nondestructive DNA extraction. The high loading capacity was achieved by directly absorbing DNA from the solution, which is then retained because of its interaction with a hyperbranched cationic polymer loaded into a negatively charged hydrogel matrix used as a support and because of its thermoresponsive nature, which allows DNA concentration within the hydrogel through multiple swelling/deswelling cycles. We were able to achieve a high DNA data density of 7.0 × 109 gigabytes per gram using a hydrogel-based system.
Collapse
Affiliation(s)
- Zhongjie Fei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nupur Gupta
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, Singapore 637141, Singapore
| | - Mengjie Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Pengfeng Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiao Hu
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, Singapore 637141, Singapore
| |
Collapse
|
12
|
Lin X, Tsao CT, Kyomoto M, Zhang M. Injectable Natural Polymer Hydrogels for Treatment of Knee Osteoarthritis. Adv Healthc Mater 2022; 11:e2101479. [PMID: 34535978 DOI: 10.1002/adhm.202101479] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/29/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is a serious chronic and degenerative disease that increasingly occurs in the aged population. Its current clinical treatments are limited to symptom relief and cannot regenerate cartilage. Although a better understanding of OA pathophysiology has been facilitating the development of novel therapeutic regimen, delivery of therapeutics to target sites with minimal invasiveness, high retention, and minimal side effects remains a challenge. Biocompatible hydrogels have been recognized to be highly promising for controlled delivery and release of therapeutics and biologics for tissue repair. In this review, the current approaches and the challenges in OA treatment, and unique properties of injectable natural polymer hydrogels as delivery system to overcome the challenges are presented. The common methods for fabrication of injectable polysaccharide-based hydrogels and the effects of their composition and properties on the OA treatment are detailed. The strategies of the use of hydrogels for loading and release cargos are also covered. Finally, recent efforts on the development of injectable polysaccharide-based hydrogels for OA treatment are highlighted, and their current limitations are discussed.
Collapse
Affiliation(s)
- Xiaojie Lin
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| | - Ching Ting Tsao
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| | - Masayuki Kyomoto
- Medical R&D Center Corporate R&D Group KYOCERA Corporation 800 Ichimiyake, Yasu Shiga 520‐2362 Japan
| | - Miqin Zhang
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| |
Collapse
|
13
|
Meng F, Wang J, Yeo Y. Nucleic acid and oligonucleotide delivery for activating innate immunity in cancer immunotherapy. J Control Release 2022; 345:586-600. [PMID: 35351528 PMCID: PMC9133138 DOI: 10.1016/j.jconrel.2022.03.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/17/2022]
Abstract
A group of nucleic acids and oligonucleotides play various roles in the innate immune system. They can stimulate pattern recognition receptors to activate innate immune cells, encode immunostimulatory proteins or peptides, or silence specific genes to block negative regulators of immune cells. Given the limitations of current cancer immunotherapy, there has been increasing interest in harnessing innate immune responses by nucleic acids and oligonucleotides. The poor biopharmaceutical properties of nucleic acids and oligonucleotides make it critical to use carriers that can protect them in circulation, retain them in the tumor microenvironment, and bring them to intracellular targets. Therefore, various gene carriers have been repurposed to deliver nucleic acids and oligonucleotides for cancer immunotherapy and improve their safety and activity. Here, we review recent studies that employed carriers to enhance the functions of nucleic acids and oligonucleotides and overall immune responses to cancer, and discuss remaining challenges and future opportunities in the development of nucleic acid-based immunotherapeutics.
Collapse
Affiliation(s)
- Fanfei Meng
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Jianping Wang
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr., West Lafayette, IN 47907, USA.
| |
Collapse
|
14
|
Apartsin E, Venyaminova A, Majoral JP, Caminade AM. Dendriplex-Impregnated Hydrogels With Programmed Release Rate. Front Chem 2022; 9:780608. [PMID: 35071182 PMCID: PMC8766751 DOI: 10.3389/fchem.2021.780608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022] Open
Abstract
Hydrogels are biocompatible matrices for local delivery of nucleic acids; however, functional dopants are required to provide efficient delivery into cells. In particular, dendrimers, known as robust nucleic acid carriers, can be used as dopants. Herein, we report the first example of impregnating neutral hydrogels with siRNA-dendrimer complexes. The surface chemistry of dendrimers allows adjusting the release rate of siRNA-containing complexes. This methodology can bring new materials for biomedical applications.
Collapse
Affiliation(s)
- Evgeny Apartsin
- Laboratoire de Chimie de Coordination, CNRS, Toulouse, France.,LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | - Alya Venyaminova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination, CNRS, Toulouse, France.,LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination, CNRS, Toulouse, France.,LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| |
Collapse
|
15
|
Duran-Mota JA, Yani JQ, Almquist BD, Borrós S, Oliva N. Polyplex-Loaded Hydrogels for Local Gene Delivery to Human Dermal Fibroblasts. ACS Biomater Sci Eng 2021; 7:4347-4361. [PMID: 34081451 DOI: 10.1021/acsbiomaterials.1c00159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Impaired cutaneous healing leading to chronic wounds affects between 2 and 6% of the total population in most developed countries and it places a substantial burden on healthcare budgets. Current treatments involving antibiotic dressings and mechanical debridement are often not effective, causing severe pain, emotional distress, and social isolation in patients for years or even decades, ultimately resulting in limb amputation. Alternatively, gene therapy (such as mRNA therapies) has emerged as a viable option to promote wound healing through modulation of gene expression. However, protecting the genetic cargo from degradation and efficient transfection into primary cells remain significant challenges in the push to clinical translation. Another limiting aspect of current therapies is the lack of sustained release of drugs to match the therapeutic window. Herein, we have developed an injectable, biodegradable and cytocompatible hydrogel-based wound dressing that delivers poly(β-amino ester)s (pBAEs) nanoparticles in a sustained manner over a range of therapeutic windows. We also demonstrate that pBAE nanoparticles, successfully used in previous in vivo studies, protect the mRNA load and efficiently transfect human dermal fibroblasts upon sustained release from the hydrogel wound dressing. This prototype wound dressing technology can enable the development of novel gene therapies for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Jose Antonio Duran-Mota
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, Barcelona 08017, Spain.,Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Júlia Quintanas Yani
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, Barcelona 08017, Spain.,Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Benjamin D Almquist
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Salvador Borrós
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, Barcelona 08017, Spain
| | - Nuria Oliva
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
16
|
Abstract
For therapeutic materials to be successfully delivered to the heart, several barriers need to be overcome, including the anatomical challenges of access, the mechanical force of the blood flow, the endothelial barrier, the cellular barrier and the immune response. Various vectors and delivery methods have been proposed to improve the cardiac-specific uptake of materials to modify gene expression. Viral and non-viral vectors are widely used to deliver genetic materials, but each has its respective advantages and shortcomings. Adeno-associated viruses have emerged as one of the best tools for heart-targeted gene delivery. In addition, extracellular vesicles, including exosomes, which are secreted by most cell types, have gained popularity for drug delivery to several organs, including the heart. Accumulating evidence suggests that extracellular vesicles can carry and transfer functional proteins and genetic materials into target cells and might be an attractive option for heart-targeted delivery. Extracellular vesicles or artificial carriers of non-viral and viral vectors can be bioengineered with immune-evasive and cardiotropic properties. In this Review, we discuss the latest strategies for targeting and delivering therapeutic materials to the heart and how the knowledge of different vectors and delivery methods could successfully translate cardiac gene therapy into the clinical setting.
Collapse
Affiliation(s)
- Susmita Sahoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Taro Kariya
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
17
|
|
18
|
Dutta K, Das R, Medeiros J, Thayumanavan S. Disulfide Bridging Strategies in Viral and Nonviral Platforms for Nucleic Acid Delivery. Biochemistry 2021; 60:966-990. [PMID: 33428850 PMCID: PMC8753971 DOI: 10.1021/acs.biochem.0c00860] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Self-assembled nanostructures that are sensitive to environmental stimuli are promising nanomaterials for drug delivery. In this class, disulfide-containing redox-sensitive strategies have gained enormous attention because of their wide applicability and simplicity of nanoparticle design. In the context of nucleic acid delivery, numerous disulfide-based materials have been designed by relying on covalent or noncovalent interactions. In this review, we highlight major advances in the design of disulfide-containing materials for nucleic acid encapsulation, including covalent nucleic acid conjugates, viral vectors or virus-like particles, dendrimers, peptides, polymers, lipids, hydrogels, inorganic nanoparticles, and nucleic acid nanostructures. Our discussion will focus on the context of the design of materials and their impact on addressing the current shortcomings in the intracellular delivery of nucleic acids.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
19
|
Lisitsyna E, Efimov A, Depresle C, Cauchois P, Vuorimaa-Laukkanen E, Laaksonen T, Durandin N. Deciphering Multiple Critical Parameters of Polymeric Self-Assembly by Fluorescence Spectroscopy of a Single Molecular Rotor BODIPY-C12. Macromolecules 2021. [DOI: 10.1021/acs.macromol.0c02167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ekaterina Lisitsyna
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
| | - Alexander Efimov
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
| | - Clémentine Depresle
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
- INSA Rouen Normandie, 685 Avenue de l’université, 76800 Saint-Etienne-du-Rouvray, France
| | - Pierre Cauchois
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
- Ecole Nationale Supérieure de Chimie de Lille, Avenue Mendeleiev, 59652 Villeneuve-d’Ascq, France
| | - Elina Vuorimaa-Laukkanen
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
| | - Timo Laaksonen
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Nikita Durandin
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33720 Tampere, Finland
| |
Collapse
|
20
|
Khan A, Alamry KA, Asiri AM. Multifunctional Biopolymers‐Based Composite Materials for Biomedical Applications: A Systematic Review. ChemistrySelect 2021; 6:154-176. [DOI: 10.1002/slct.202003978] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/26/2020] [Indexed: 01/06/2025]
Abstract
AbstractBiopolymers are considered as a favorable group of substances with a broad array of applications, of which biomedical field stands out. The interesting features of biopolymers such as low‐cost, non‐cytotoxicity, hydrophilicity, biodegradation and biocompatibility make them promising and excellent feedstock to be used in implantable devices. The bounteous reactive functional groups in the backbone structure of polysaccharides and its derivatives could be utilized to develop hydrogels, nano‐composite and 3D scaffolds with appealing structures and desired features, leading to promising research attention towards biomedical fields. The present review describes the foremost properties as well as potential of different biopolymers, and their composites for application in implantable biomedical systems. This work may introduce readers about the comprehension of state‐of‐the‐art advances, real present challenges along with the future anticipation of eco‐friendly and biomimetic techniques for the modification of biopolymeric materials to improve their biomedical applications.
Collapse
Affiliation(s)
- Ajahar Khan
- Faculty of Science Department of Chemistry King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Khalid A. Alamry
- Faculty of Science Department of Chemistry King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Abdullah M. Asiri
- Faculty of Science Department of Chemistry King Abdulaziz University Jeddah 21589 Saudi Arabia
- Centre of Excellence for Advanced Materials Research King Abdulaziz University Jeddah 21589 Saudi Arabia
| |
Collapse
|
21
|
Han S, Park Y, Kim H, Nam H, Ko O, Lee JB. Double Controlled Release of Therapeutic RNA Modules through Injectable DNA-RNA Hybrid Hydrogel. ACS APPLIED MATERIALS & INTERFACES 2020; 12:55554-55563. [PMID: 33259200 DOI: 10.1021/acsami.0c12506] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Advances in the DNA nanotechnology have enabled the fabrication of DNA-based hydrogels with precisely controlled structures and tunable mechanical and biological properties. Compared to DNA hydrogel, preparation of RNA-based hydrogel remains challenging due to the inherent instability of naked RNA. To overcome these limitations, we fabricated a DNA-RNA hybrid hydrogel via stepwise dual enzymatic polymerization. Multimeric short hairpin RNAs (shRNAs) were hybridized with functional DNA aptamers for targeting and mechanical properties of the hydrogel. The obtained DNA-RNA hybrid hydrogel was ultrasoft, robust, and injectable hence reconfigurable into any confined structures. As a model system, the hydrogel was able to mimic microtubule structures under physiological conditions and designed to release the functional small interfering RNA (siRNA)-aptamer complex (SAC) sequentially. In addition, we encoded restriction enzyme-responsive sites in DNA-RNA hybrid hydrogel to boost the release of SAC. This novel strategy provides an excellent platform for systematic RNA delivery through double-controlled release, SAC release from hydrogel, and subsequent release of siRNA from the SAC, which has promising potential in RNA therapy.
Collapse
Affiliation(s)
- Sangwoo Han
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Yongkuk Park
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Hyejin Kim
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Hyangsu Nam
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Ohsung Ko
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| | - Jong Bum Lee
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemungu, Seoul 02504, Republic of Korea
| |
Collapse
|
22
|
Shin DY, Park JU, Choi MH, Kim S, Kim HE, Jeong SH. Polydeoxyribonucleotide-delivering therapeutic hydrogel for diabetic wound healing. Sci Rep 2020; 10:16811. [PMID: 33033366 PMCID: PMC7546631 DOI: 10.1038/s41598-020-74004-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Patients with diabetes experience delayed wound healing because of the uncontrolled glucose level in their bloodstream, which leads to impaired function of white blood cells, poor circulation, decreased production and repair of new blood vessels. Treatment using polydeoxyribonucleotide (PDRN), which is a DNA extracted from the sperm cells of salmon, has been introduced to accelerate the healing process of diabetic wounds. To accelerate the wound-healing process, sustained delivery of PDRN is critical. In this study, taking advantage of the non-invasive gelation property of alginate, PDRN was loaded inside the hydrogel (Alg-PDRN). The release behavior of PDRN was altered by controlling the crosslinking density of the Alg hydrogel. The amount of PDRN was the greatest inside the hydrogel with the highest crosslinking density because of the decreased diffusion. However, there was an optimal degree of crosslinking for the effective release of PDRN. In vitro studies using human dermal fibroblasts and diabetes mellitus fibroblasts and an in ovo chorioallantoic membrane assay confirmed that the Alg-PDRN hydrogel effectively induced cell proliferation and expression of angiogenic growth factors and promoted new blood vessel formation. Its effectiveness for accelerated diabetic wound healing was also confirmed in an in-vivo animal experiment using a diabetic mouse model.
Collapse
Affiliation(s)
- Da Yong Shin
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji-Ung Park
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Min-Ha Choi
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Sukwha Kim
- Medical Big Data Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hyoun-Ee Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Advanced Institutes of Convergence Technology, Seoul National University, Gwanggyo, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Seol-Ha Jeong
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
23
|
Bholakant R, Qian H, Zhang J, Huang X, Huang D, Feijen J, Zhong Y, Chen W. Recent Advances of Polycationic siRNA Vectors for Cancer Therapy. Biomacromolecules 2020; 21:2966-2982. [DOI: 10.1021/acs.biomac.0c00438] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Raut Bholakant
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| | - Hongliang Qian
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| | - Junmei Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xin Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jan Feijen
- Department of Polymer Chemistry and Biomaterials, Faculty of Science and Technology, TECHMED Centre, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
24
|
Fliervoet LAL, Zhang H, van Groesen E, Fortuin K, Duin NJCB, Remaut K, Schiffelers RM, Hennink WE, Vermonden T. Local release of siRNA using polyplex-loaded thermosensitive hydrogels. NANOSCALE 2020; 12:10347-10360. [PMID: 32369076 DOI: 10.1039/d0nr03147j] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
One of the challenges for the clinical translation of RNA interference (RNAi)-based therapies concerns the deposition of therapeutically effective doses of the nucleic acids, like siRNA, at a local tissue level without severe off-target effects. To address this issue, hydrogels can be used as matrices for the local and sustained release of the siRNA cargo. In this study, the formation of polyplexes based on siRNA and poly(2-dimethylaminoethyl methacrylate) (PDMAEMA)-based polymers was investigated, followed by their loading in a thermosensitive hydrogel to promote local siRNA release. A multifunctional NPD triblock copolymer consisting of a thermosensitive poly(N-isopropylacrylamide) (PNIPAM, N), a hydrophilic poly(ethylene glycol) (PEG, P), and a cationic PDMAEMA (D) block was used to study the binding properties with siRNA taking the non-thermosensitive PD polymer as control. For both polymers, small polyplexes with sizes ranging from 10-20 nm were formed in aqueous solution (HBS buffer, 20 mM HEPES, 150 mM NaCl, pH 7.4) when prepared at a N/P charge ratio of 5 or higher. Formulating the siRNA into NPD or PD polyplexes before loading into the thermosensitive PNIPAM-PEG-PNIPAM hydrogel resulted in a more controlled and sustained release compared to free siRNA release from the hydrogel. The polyplexes were released for 128 hours in HBS, when changing the release medium twice a day, while free siRNA was completely released within 50 hours with already 40% being released after changing the release medium just once. The release of the polyplexes was dependent on the dissolution rate of the hydrogel matrix. Moreover, intact polyplexes were released from the hydrogels with a similar size as before loading, suggesting that the hydrogel material did not compromise the polyplex stability. Finally, it was shown that the released polyplexes were still biologically active and transfected FaDu cells, which was observed by siRNA-induced luciferase silencing in vitro. This study shows the development of an injectable thermosensitive hydrogel to promote local and sustained release of siRNA, which can potentially be used to deliver siRNA for various applications, such as the treatment of tumors.
Collapse
Affiliation(s)
- Lies A L Fliervoet
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, PO Box 80082, 3508 TB Utrecht, the Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Najafi M, Asadi H, van den Dikkenberg J, van Steenbergen MJ, Fens MHAM, Hennink WE, Vermonden T. Conversion of an Injectable MMP-Degradable Hydrogel into Core-Cross-Linked Micelles. Biomacromolecules 2020; 21:1739-1751. [PMID: 31945299 PMCID: PMC7218746 DOI: 10.1021/acs.biomac.9b01675] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/16/2020] [Indexed: 01/01/2023]
Abstract
In this study, a new type of injectable hydrogel called "HyMic" that can convert into core cross-linked (CCL) micelles upon exposure to matrix metalloproteinases (MMP's), was designed and developed for drug delivery applications. HyMic is composed of CCL micelles connected via an enzyme cleavable linker. To this end, two complementary ABA block copolymers with polyethylene glycol (PEG) as B block were synthesized using atom transfer radical polymerization (ATRP). The A blocks were composed of a random copolymer of N-isopropylacrylamide (NIPAM) and either N-(2-hydroxypropyl)methacrylamide-cysteine (HPMA-Cys) or N-(2-hydroxypropyl) methacrylamide-ethylthioglycolate succinic acid (HPMA-ETSA). Mixing the aqueous solutions of the obtained polymers and rising the temperature above the cloud point of the PNIPAM block resulted in the self-assembly of these polymers into flower-like micelles composed of a hydrophilic PEG shell and hydrophobic core. The micellar core was cross-linked by native chemical ligation between the cysteine (in HPMA-Cys) and thioester (in HPMA-ETSA) functionalities. A slight excess of thioester to cysteine groups (molar ratio 3:2) was used to allow further chemical reactions exploiting the unreacted thioester groups. The obtained micelles displayed a Z-average diameter of 80 ± 1 nm (PDI 0.1), and ζ-potential of -4.2 ± 0.4 mV and were linked using two types of pentablock copolymers of P(NIPAM-co-HPMA-Cys)-PEG-peptide-PEG-P(NIPAM-co-HPMA-Cys) (Pep-NC) to yield hydrogels. The pentablock copolymers were synthesized using a PEG-peptide-PEG ATRP macroinitiator and the peptide midblock (lysine-glycine-proline-glutamine-isoleucine-phenylalanine-glycine-glutamine-lysine (Lys-Gly-Pro-Gln-Gly-Ile-Phe-Gly-Gln-Lys)) consisted of either l- or d-amino acids (l-Pep-NC or d-Pep-NC), of which the l-amino acid sequence is a substrate for matrix metalloproteases 2 and 9 (MMPs 2 and 9). Upon mixing of the CCL micelles and the linker (l/d-Pep-NC), the cysteine functionalities of the l/d-Pep-NC reacted with remaining thioester moieties in the micellar core via native chemical ligation yielding a hydrogel within 160 min as demonstrated by rheological measurements. As anticipated, the gel cross-linked with l-Pep-NC was degraded in 7-45 days upon exposure to metalloproteases in a concentration-dependent manner, while the gel cross-linked with the d-Pep-NC remained intact even after 2 months. Dynamic light scattering analysis of the release medium revealed the presence of nanoparticles with a Z-average diameter of ∼120 nm (PDI < 0.3) and ζ-potential of ∼-3 mV, indicating release of core cross-linked micelles upon HyMic exposure to metalloproteases. An in vitro study demonstrated that the released CCL micelles were taken up by HeLa cells. Therefore, HyMic as an injectable and enzyme degradable hydrogel displaying controlled and on-demand release of CCL micelles has potential for intracellular drug delivery in tissues with upregulation of MMPs, for example, in cancer tissues.
Collapse
Affiliation(s)
- Marzieh Najafi
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Hamed Asadi
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
- Polymer
Laboratory, Chemistry Department, School of Science, University of Tehran, Tehran, Iran
| | - Joep van den Dikkenberg
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Mies J. van Steenbergen
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Marcel H. A. M. Fens
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Science for Life, Faculty of Science, Utrecht
University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
26
|
Askari E, Seyfoori A, Amereh M, Gharaie SS, Ghazali HS, Ghazali ZS, Khunjush B, Akbari M. Stimuli-Responsive Hydrogels for Local Post-Surgical Drug Delivery. Gels 2020; 6:E14. [PMID: 32397180 PMCID: PMC7345431 DOI: 10.3390/gels6020014] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023] Open
Abstract
Currently, surgical operations, followed by systemic drug delivery, are the prevailing treatment modality for most diseases, including cancers and trauma-based injuries. Although effective to some extent, the side effects of surgery include inflammation, pain, a lower rate of tissue regeneration, disease recurrence, and the non-specific toxicity of chemotherapies, which remain significant clinical challenges. The localized delivery of therapeutics has recently emerged as an alternative to systemic therapy, which not only allows the delivery of higher doses of therapeutic agents to the surgical site, but also enables overcoming post-surgical complications, such as infections, inflammations, and pain. Due to the limitations of the current drug delivery systems, and an increasing clinical need for disease-specific drug release systems, hydrogels have attracted considerable interest, due to their unique properties, including a high capacity for drug loading, as well as a sustained release profile. Hydrogels can be used as local drug performance carriers as a means for diminishing the side effects of current systemic drug delivery methods and are suitable for the majority of surgery-based injuries. This work summarizes recent advances in hydrogel-based drug delivery systems (DDSs), including formulations such as implantable, injectable, and sprayable hydrogels, with a particular emphasis on stimuli-responsive materials. Moreover, clinical applications and future opportunities for this type of post-surgery treatment are also highlighted.
Collapse
Affiliation(s)
- Esfandyar Askari
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran P.O. Box 1517964311, Iran;
| | - Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (M.A.); (S.S.G.); (B.K.)
| | - Meitham Amereh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (M.A.); (S.S.G.); (B.K.)
| | - Sadaf Samimi Gharaie
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (M.A.); (S.S.G.); (B.K.)
| | - Hanieh Sadat Ghazali
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran P.O. Box 16846-13114, Iran;
| | - Zahra Sadat Ghazali
- Biomedical Engineering Department, Amirkabir University of Technology (AUT), Tehran P.O. Box 158754413, Iran;
| | - Bardia Khunjush
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (M.A.); (S.S.G.); (B.K.)
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (M.A.); (S.S.G.); (B.K.)
- Center for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
27
|
Shen T, Zhang Y, Zhou S, Lin S, Zhang XB, Zhu G. Nucleic Acid Immunotherapeutics for Cancer. ACS APPLIED BIO MATERIALS 2020; 3:2838-2849. [PMID: 33681722 DOI: 10.1021/acsabm.0c00101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The past decade has witnessed the blossom of two fields: nucleic acid therapeutics and cancer immunotherapy. Unlike traditional small molecule medicines or protein biologics, nucleic acid therapeutics have characteristic features such as storing genetic information, immunomodulation, and easy conformational recovery. Immunotherapy uses the patients' own immune system to treat cancer. A variety of strategies have been developed for cancer immunotherapy including immune checkpoint blockade, adoptive cell transfer therapy, therapeutic vaccines, and oncolytic virotherapy. Interestingly, nucleic acid therapeutics have emerged as a pivotal class of regimen for cancer immunotherapy. Examples of such nucleic acid immunotherapeutics include immunostimulatory DNA/RNA, mRNA/plasmids that can be translated into immunotherapeutic proteins/peptides, and genome-editing nucleic acids. Like many other therapeutic nucleic acids, nucleic acid immunotherapeutics often require chemical modifications to protect them from enzymatic degradation and need drug delivery systems for optimal delivery to target tissues and cells and subcellular locations. In this review, we attempted to summarize recent advancement in the interfacial field of nucleic acid immunotherapeutics for cancer treatment.
Collapse
Affiliation(s)
- Tingting Shen
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha 410082, China; Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences-School of Pharmacy; Massey Cancer Center; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Yu Zhang
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences-School of Pharmacy; Massey Cancer Center; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23298, United States; Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Shurong Zhou
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences-School of Pharmacy; Massey Cancer Center; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Shuibin Lin
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Bing Zhang
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha 410082, China
| | - Guizhi Zhu
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences-School of Pharmacy; Massey Cancer Center; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
28
|
Fliervoet LA, Lisitsyna ES, Durandin NA, Kotsis I, Maas-Bakker RFM, Yliperttula M, Hennink WE, Vuorimaa-Laukkanen E, Vermonden T. Structure and Dynamics of Thermosensitive pDNA Polyplexes Studied by Time-Resolved Fluorescence Spectroscopy. Biomacromolecules 2020; 21:73-88. [PMID: 31500418 PMCID: PMC6961130 DOI: 10.1021/acs.biomac.9b00896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/29/2019] [Indexed: 12/15/2022]
Abstract
Combining multiple stimuli-responsive functionalities into the polymer design is an attractive approach to improve nucleic acid delivery. However, more in-depth fundamental understanding how the multiple functionalities in the polymer structures are influencing polyplex formation and stability is essential for the rational development of such delivery systems. Therefore, in this study the structure and dynamics of thermosensitive polyplexes were investigated by tracking the behavior of labeled plasmid DNA (pDNA) and polymer with time-resolved fluorescence spectroscopy using fluorescence resonance energy transfer (FRET). The successful synthesis of a heterofunctional poly(ethylene glycol) (PEG) macroinitiator containing both an atom transfer radical polymerization (ATRP) and reversible addition-fragmentation chain-transfer (RAFT) initiator is reported. The use of this novel PEG macroinitiator allows for the controlled polymerization of cationic and thermosensitive linear triblock copolymers and labeling of the chain-end with a fluorescent dye by maleimide-thiol chemistry. The polymers consisted of a thermosensitive poly(N-isopropylacrylamide) (PNIPAM, N), hydrophilic PEG (P), and cationic poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA, D) block, further referred to as NPD. Polymer block D chain-ends were labeled with Cy3, while pDNA was labeled with FITC. The thermosensitive NPD polymers were used to prepare pDNA polyplexes, and the effect of the N/P charge ratio, temperature, and composition of the triblock copolymer on the polyplex properties were investigated, taking nonthermosensitive PD polymers as the control. FRET was observed both at 4 and 37 °C, indicating that the introduction of the thermosensitive PNIPAM block did not compromise the polyplex structure even above the polymer's cloud point. Furthermore, FRET results showed that the NPD- and PD-based polyplexes have a less dense core compared to polyplexes based on cationic homopolymers (such as PEI) as reported before. The polyplexes showed to have a dynamic character meaning that the polymer chains can exchange between the polyplex core and shell. Mobility of the polymers allow their uniform redistribution within the polyplex and this feature has been reported to be favorable in the context of pDNA release and subsequent improved transfection efficiency, compared to nondynamic formulations.
Collapse
Affiliation(s)
- Lies A.
L. Fliervoet
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Ekaterina S. Lisitsyna
- Chemistry
and Advanced Materials, Faculty of Engineering and Natural Sciences, Tampere University, FI-33014 Tampere, Finland
| | - Nikita A. Durandin
- Chemistry
and Advanced Materials, Faculty of Engineering and Natural Sciences, Tampere University, FI-33014 Tampere, Finland
| | - Ilias Kotsis
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Roel F. M. Maas-Bakker
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Marjo Yliperttula
- Division
of Pharmaceutical Biosciences and Drug Research Program, University of Helsinki, P.O. Box 56 (Viikinkaari 5E), 00014 Helsinki, Finland
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Elina Vuorimaa-Laukkanen
- Chemistry
and Advanced Materials, Faculty of Engineering and Natural Sciences, Tampere University, FI-33014 Tampere, Finland
| | - Tina Vermonden
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
29
|
Lei K, Li Z, Zhu D, Sun C, Sun Y, Yang C, Zheng Z, Wang X. Polysaccharide-based recoverable double-network hydrogel with high strength and self-healing properties. J Mater Chem B 2020; 8:794-802. [PMID: 31904754 DOI: 10.1039/c9tb01679a] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Polysaccharide-based hydrogels (PSBHs) have received significant attention for numerous bio-applications due to their biocompatibility and non-immunogenic performance. However, the construction of PSBH with superior mechanical properties by a simple method is rarely adequately researched. This study focuses on the construction of a novel PSBH with superior mechanical and recoverable properties by integrating the synergistic and complementary interactions of covalent bond-associated oxidized sodium alginate (SA-CHO) gel and hydrogen bond-associated agarose (Aga) gel. With the synergy and complementarity of the SA-CHO and Aga networks, the hydrogel exhibited 17 and 15 times (20 and 9 times) greater compressive stress and modulus, respectively, compared with the SA-CHO gel (Aga gel). The hydrogel also displayed excellent fatigue resistance, recurrent shapeability, acid resistance and recovery ability, as well as self-healing ability. This study provides a unique perspective for enhancing the mechanical properties of PSBH through the synergy and complementarity of different kinds of polysaccharides without sacrificing the functionality of the PSBH.
Collapse
Affiliation(s)
- Kun Lei
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhao Li
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Dandan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Chengyuan Sun
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yunlong Sun
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Chongchong Yang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhen Zheng
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xinling Wang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
30
|
Lin J, Jo SB, Kim TH, Kim HW, Chew SY. RNA interference in glial cells for nerve injury treatment. J Tissue Eng 2020; 11:2041731420939224. [PMID: 32670539 PMCID: PMC7338726 DOI: 10.1177/2041731420939224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/13/2020] [Indexed: 12/12/2022] Open
Abstract
Drivers of RNA interference are potent for manipulating gene and protein levels, which enable the restoration of dysregulated mRNA expression that is commonly associated with injuries and diseases. This review summarizes the potential of targeting neuroglial cells, using RNA interference, to treat nerve injuries sustained in the central nervous system. In addition, the various methods of delivering these RNA interference effectors will be discussed.
Collapse
Affiliation(s)
- Junquan Lin
- School of Chemical and Biomedical
Engineering, Nanyang Technological University, Singapore
| | - Seung Bin Jo
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
| | - Tae-Hyun Kim
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science
& BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook
University, Cheonan, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration
Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science
& BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook
University, Cheonan, Republic of Korea
- UCL Eastman-Korea Dental Medicine
Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Sing Yian Chew
- School of Chemical and Biomedical
Engineering, Nanyang Technological University, Singapore
- Lee Kong Chian School of Medicine,
Nanyang Technological University, Singapore
| |
Collapse
|
31
|
|
32
|
Darge HF, Andrgie AT, Tsai HC, Lai JY. Polysaccharide and polypeptide based injectable thermo-sensitive hydrogels for local biomedical applications. Int J Biol Macromol 2019; 133:545-563. [DOI: 10.1016/j.ijbiomac.2019.04.131] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 01/19/2023]
|
33
|
Fliervoet LAL, van Nostrum CF, Hennink WE, Vermonden T. Balancing hydrophobic and electrostatic interactions in thermosensitive polyplexes for nucleic acid delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.1088/2399-7532/ab12ee] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|