1
|
Baek H, Yang SW, Kim MK, Kim D, Lee C, Kim S, Lee Y, Park M, Hwang HS, Paik HJ, Kang YS. Activation of Immune Responses Through the RIG-I Pathway Using TRITC-Dextran Encapsulated Nanoparticles. Immune Netw 2024; 24:e44. [PMID: 39801741 PMCID: PMC11711124 DOI: 10.4110/in.2024.24.e44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Pathogen-associated molecular patterns (PAMPs) are highly conserved motifs originating from microorganisms that act as ligands for pattern recognition receptors (PRRs), which are crucial for defense against pathogens. Thus, PAMP-mimicking vaccines may induce potent immune activation and provide broad-spectrum protection against microbes. Dextran encapsulation can regulate the surface characteristics of nanoparticles (NPs) and induces their surface modification. To determine whether dextran-encapsulated NPs can be used to develop antiviral vaccines by mimicking viral PAMPs, we synthesized NPs in a cyclohexane inverse miniemulsion (Basic-NPs) and further encapsulated them with dextran or tetramethylrhodamine isothiocyanate (TRITC)-dextran (Dex-NPs or TDex-NPs). We hypothesized that these dextran encapsulated NPs could activate innate immunity through cell surface or cytosolic PRRs. In vitro and in vivo experiments were performed using RAW 264.7 and C57BL/6 mice to test different concentrations and routes of administration. Only TDex-NPs rapidly increased retinoic acid-inducible gene I (RIG-I) at 8 h and directly bound to it, producing 120-300 pg/ml of IFN-α via the ERK/NF-κB signaling pathway in both in vitro and in vivo models. The effect of TDex-NPs in mice was observed exclusively with footpad injections. Our findings suggest that TRITC-dextran encapsulated NPs exhibit surface properties for RIG-I binding, offering potential development as a novel antiviral and anticancer RIG-I agonist.
Collapse
Affiliation(s)
- Hayeon Baek
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, Seoul 05029, Korea
| | - Seung-Woo Yang
- Sanford Consortium for Regenerative Medicine, School of Medicine, University of California, San Diego, CA 92521, USA
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea
| | - Min-Kyung Kim
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, Seoul 05029, Korea
| | - Dongwoo Kim
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea
| | - Chaeyeon Lee
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea
| | - Seulki Kim
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Yunseok Lee
- Department of Animal Science and Technology, College of Sang-Huh Life Science, Konkuk University, Seoul 05029, Korea
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, Seoul 05029, Korea
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea
| | - Hyun-jong Paik
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, Seoul 05029, Korea
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
2
|
Liang L, Deng Y, Ao Z, Liao C, Tian J, Li C, Yu X. Recent progress in biomimetic nanomedicines based on versatile targeting strategy for atherosclerosis therapy. J Drug Target 2024; 32:606-623. [PMID: 38656224 DOI: 10.1080/1061186x.2024.2347353] [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: 01/30/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis (AS) is considered to be one of the major causes of cardiovascular disease. Its pathological microenvironment is characterised by increased production of reactive oxygen species, lipid oxides, and excessive inflammatory factors, which accumulate at the monolayer endothelial cells in the vascular wall to form AS plaques. Therefore, intervention in the pathological microenvironment would be beneficial in delaying AS. Researchers have designed biomimetic nanomedicines with excellent biocompatibility and the ability to avoid being cleared by the immune system through different therapeutic strategies to achieve better therapeutic effects for the characteristics of AS. Biomimetic nanomedicines can further enhance delivery efficiency and improve treatment efficacy due to their good biocompatibility and ability to evade clearance by the immune system. Biomimetic nanomedicines based on therapeutic strategies such as neutralising inflammatory factors, ROS scavengers, lipid clearance and integration of diagnosis and treatment are versatile approaches for effective treatment of AS. The review firstly summarises the targeting therapeutic strategy of biomimetic nanomedicine for AS in recent 5 years. Biomimetic nanomedicines using cell membranes, proteins, and extracellular vesicles as carriers have been developed for AS.
Collapse
Affiliation(s)
- Lijuan Liang
- Department of Pharmacy, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Changli Liao
- Science and Technology Department, Southwest Medical University, Luzhou, Sichuan, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Phillips SG, Lankone AR, O'Hagan SS, Ganji N, Fairbrother DH. Gas-Phase Functionalization of Phytoglycogen Nanoparticles and the Role of Reagent Structure in the Formation of Self-Limiting Hydrophobic Shells. Biomacromolecules 2024; 25:2902-2913. [PMID: 38593289 DOI: 10.1021/acs.biomac.4c00026] [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: 04/11/2024]
Abstract
A suite of acyl chloride structural isomers (C6H11OCl) was used to effect gas-phase esterification of starch-based phytoglycogen nanoparticles (PhG NPs). The surface degree of substitution (DS) was quantified using X-ray photoelectron spectroscopy, while the overall DS was quantified using 1H NMR spectroscopy. Gas-phase modification initiates at the NP surface, with the extent of surface and overall esterification determined by both the reaction time and the steric footprint of the acyl chloride reagent. The less sterically hindered acyl chlorides diffuse fully into the NP interior, while the branched isomers are restricted to the near-surface region and form self-limiting hydrophobic shells, with shell thicknesses decreasing with increasing steric footprint. These differences in substitution were also reflected in the solubility of the NPs, with water solubility systematically decreasing with increasing DS. The ability to separately control both the surface and overall degree of functionalization and thereby form thin hydrophobic shells has significant implications for the development of polysaccharide-based biopolymers as nanocarrier delivery systems.
Collapse
Affiliation(s)
- Savannah G Phillips
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Alyssa R Lankone
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | | | - Nasim Ganji
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - D Howard Fairbrother
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
4
|
Yu T, Xu Q, Chen X, Deng X, Chen N, Kou MT, Huang Y, Guo J, Xiao Z, Wang J. Biomimetic nanomaterials in myocardial infarction treatment: Harnessing bionic strategies for advanced therapeutics. Mater Today Bio 2024; 25:100957. [PMID: 38322664 PMCID: PMC10844134 DOI: 10.1016/j.mtbio.2024.100957] [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: 10/20/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Myocardial infarction (MI) and its associated poor prognosis pose significant risks to human health. Nanomaterials hold great potential for the treatment of MI due to their targeted and controlled release properties, particularly biomimetic nanomaterials. The utilization of biomimetic strategies based on extracellular vesicles (EVs) and cell membranes will serve as the guiding principle for the development of nanomaterial therapy in the future. In this review, we present an overview of research progress on various exosomes derived from mesenchymal stem cells, cardiomyocytes, or induced pluripotent stem cells in the context of myocardial infarction (MI) therapy. These exosomes, utilized as cell-free therapies, have demonstrated the ability to enhance the efficacy of reducing the size of the infarcted area and preventing ischaemic reperfusion through mechanisms such as oxidative stress reduction, polarization modulation, fibrosis inhibition, and angiogenesis promotion. Moreover, EVs can exert cardioprotective effects by encapsulating therapeutic agents and can be engineered to specifically target the infarcted myocardium. Furthermore, we discuss the use of cell membranes derived from erythrocytes, stem cells, immune cells and platelets to encapsulate nanomaterials. This approach allows the nanomaterials to camouflage themselves as endogenous substances targeting the region affected by MI, thereby minimizing toxicity and improving biocompatibility. In conclusion, biomimetic nano-delivery systems hold promise as a potentially beneficial technology for MI treatment. This review serves as a valuable reference for the application of biomimetic nanomaterials in MI therapy and aims to expedite the translation of NPs-based MI therapeutic strategies into practical clinical applications.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Qiaxin Xu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Xu Chen
- Department of Clinical Pharmacy, Daqing Oilfield General Hospital, Daqing, 163000, China
| | - Xiujiao Deng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Nenghua Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Man Teng Kou
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Jun Guo
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
5
|
Zhang X, Yi Y, Jiang Y, Liao J, Yang R, Deng X, Zhang L. Targeted Therapy of Acute Liver Injury via Cryptotanshinone-Loaded Biomimetic Nanoparticles Derived from Mesenchymal Stromal Cells Driven by Homing. Pharmaceutics 2023; 15:2764. [PMID: 38140104 PMCID: PMC10747007 DOI: 10.3390/pharmaceutics15122764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/16/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Acute liver injury (ALI) has the potential to compromise hepatic function rapidly, with severe cases posing a considerable threat to human health and wellbeing. Conventional treatments, such as the oral administration of antioxidants, can inadvertently lead to liver toxicity and other unwanted side effects. Mesenchymal stromal cells (MSCs) can target therapeutic agents directly to inflammatory sites owing to their homing effect, and they offer a promising avenue for the treatment of ALI. However, the efficacy and feasibility of these live cell products are hampered by challenges associated with delivery pathways and safety concerns. Therefore, in this work, MSC membranes were ingeniously harnessed as protective shells to encapsulate synthesized PLGA nanoparticle cores (PLGA/MSCs). This strategic approach enabled nanoparticles to simulate endogenous substances and yielded a core-shell nano-biomimetic structure. The biomimetic nanocarrier remarkably maintained the homing ability of MSCs to inflammatory sites. In this study, cryptotanshinone (CPT)-loaded PLGA/MSCs (CPT@PLGA/MSC) were prepared. These nanoparticles can be effectively internalized by LO2 cells. They reduced cellular oxidative stress and elevated inflammatory levels. In vivo results suggested that, after intravenous administration, CPT@PLGA/MSCs significantly reduced uptake by the reticuloendothelial system and immune recognition compared to PLGA nanoparticles without MSC membrane coatings, subsequently resulting in their targeted and enhanced accumulation in the liver. The effectiveness of CPT@PLGA/MSCs in alleviating carbon tetrachloride-induced oxidative stress and inflammation in a mouse model was unequivocally demonstrated through comprehensive histological examination and liver function tests. This study introduces a pioneering strategy with substantial potential for ALI treatment.
Collapse
Affiliation(s)
- Xin Zhang
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Yao Yi
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Yuanyuan Jiang
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Jinqiu Liao
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.L.); (R.Y.)
| | - Ruiwu Yang
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.L.); (R.Y.)
| | - Xuexue Deng
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Li Zhang
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| |
Collapse
|
6
|
Chen Y, Wang J, Xu J, Zhang J, Xu S, Zhang Q, Huang J, Peng J, Xu H, Du Q, Gong Z. Fabrication of a Polysaccharide-Protein/Protein Complex Stabilized Oral Nanoemulsion to Facilitate the Therapeutic Effects of 1,8-Cineole on Atherosclerosis. ACS NANO 2023; 17:9090-9109. [PMID: 37172004 DOI: 10.1021/acsnano.2c12230] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Atherosclerosis (AS) is a systemic disease characterized by lipid deposition in the blood vessel wall that urgently requires effective and safe therapeutic drugs for long-term treatment. An essential oil monomer-1,8-cineole (CIN) with ameliorative effects on vascular injuries has considerable potential for preventing the progression of AS because of its antioxidant, anti-inflammation, and cholesterol regulatory effects. However, the high volatility and instability of CIN result in low oral bioavailability and a short half-life, thereby limiting its clinical application. We formulated a nanoemulsion using a polysaccharide-protein/protein complex (dextran-bovine serum albumin/protamine, DEX5k-BSA/PTM) as an emulsifier, with vitamin B12 (VB12) as the ligand to facilitate the transportation across the small intestine. An emulsion preparation method using a microjet followed by ultraviolet irradiation was developed to obtain the CIN-loaded oral nanoemulsion CIN@DEX5k-BSA/PTM/VB12. The nanoemulsion improved the stability of CIN both in vitro and in vivo, prolonged the retention time in the gastrointestinal tract (GIT), and enhanced the permeability across the mucus layer and intestinal epithelial cells to increase oral bioavailability and plaque accumulation of CIN. Validated in an AS mouse model, CIN@DEX5k-BSA/PTM/VB12 achieved prominent therapeutic efficacy combating AS. This study highlights the advantages of DEX5k-BSA/PTM and VB12 in the development of nanoemulsions for CIN and provides a promising oral nanoplatform for the delivery of essential oils.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | | | | |
Collapse
|
7
|
Vasquez-Martínez N, Guillen D, Moreno-Mendieta SA, Sanchez S, Rodríguez-Sanoja R. The Role of Mucoadhesion and Mucopenetration in the Immune Response Induced by Polymer-Based Mucosal Adjuvants. Polymers (Basel) 2023; 15:1615. [PMID: 37050229 PMCID: PMC10097111 DOI: 10.3390/polym15071615] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Mucus is a viscoelastic gel that acts as a protective barrier for epithelial surfaces. The mucosal vehicles and adjuvants need to pass through the mucus layer to make drugs and vaccine delivery by mucosal routes possible. The mucoadhesion of polymer particle adjuvants significantly increases the contact time between vaccine formulations and the mucosa; then, the particles can penetrate the mucus layer and epithelium to reach mucosa-associated lymphoid tissues. This review presents the key findings that have aided in understanding mucoadhesion and mucopenetration while exploring the influence of physicochemical characteristics on mucus-polymer interactions. We describe polymer-based particles designed with mucoadhesive or mucopenetrating properties and discuss the impact of mucoadhesive polymers on local and systemic immune responses after mucosal immunization. In future research, more attention paid to the design and development of mucosal adjuvants could lead to more effective vaccines.
Collapse
Affiliation(s)
- Nathaly Vasquez-Martínez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
- Programa de Doctorado en Ciencia Bioquímicas, Universidad Nacional Autónoma de México, Circuito de Posgrado, C.U., Coyoacán, Mexico City 04510, Mexico
| | - Daniel Guillen
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
| | - Silvia Andrea Moreno-Mendieta
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
- Programa de Doctorado en Ciencia Bioquímicas, Universidad Nacional Autónoma de México, Circuito de Posgrado, C.U., Coyoacán, Mexico City 04510, Mexico
- Consejo Nacional de Ciencia y Tecnología, Benito Juárez, Mexico City 03940, Mexico
| | - Sergio Sanchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
| |
Collapse
|
8
|
Fan L, Wei A, Gao Z, Mu X. Current progress of mesenchymal stem cell membrane-camouflaged nanoparticles for targeted therapy. Biomed Pharmacother 2023; 161:114451. [PMID: 36870279 DOI: 10.1016/j.biopha.2023.114451] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Nanodrug delivery systems have been widely used in disease treatment. However, weak drug targeting, easy to be cleared by the immune system, and low biocompatibility are great obstacles for drug delivery. As an important part of cell information transmission and behavior regulation, cell membrane can be used as drug coating material which represents a promising strategy and can overcome these limitations. Mesenchymal stem cell (MSC) membrane, as a new carrier, has the characteristics of active targeting and immune escape of MSC, and has broad application potential in tumor treatment, inflammatory disease, tissue regeneration and other fields. Here, we review recent progress on the use of MSC membrane-coated nanoparticles for therapy and drug delivery, aiming to provide guidance for the design and clinical application of membrane carrier in the future.
Collapse
Affiliation(s)
- Lianlian Fan
- Department of Pharmacy, China-Japan Union Hospital, Jilin University, Changchun130033, China
| | - Anhui Wei
- Department of Regenerative Medicine, College of Pharmacy, Jilin University, Changchun130021, China
| | - Zihui Gao
- Changchun City Experimental High School, Changchun130117, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun130033, China.
| |
Collapse
|
9
|
Kannaujiya VK, De Rubis G, Paudel KR, Manandhar B, Chellappan DK, Singh SK, MacLoughlin R, Gupta G, Xenaki D, Kumar P, Hansbro PM, Oliver BGG, Wich PR, Dua K. Anticancer activity of NFκB decoy oligonucleotide-loaded nanoparticles against human lung cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
10
|
Imran M, Gowd V, Saha P, Rashid S, Ahmad Chaudhary A, Mohamed MYA, Alawam AS, Khan R. Biologically inspired stealth - Camouflaged strategies in nanotechnology for the improved therapies in various diseases. Int J Pharm 2023; 631:122407. [PMID: 36402290 DOI: 10.1016/j.ijpharm.2022.122407] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/27/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022]
Abstract
Nanotechnology has received increasing attention in the past decade and it's being used as a model for developing better treatments for a variety of diseases. Despite the fact that nanotechnology-based therapy has greatly improved treatment regimens, it still faces challenges such as inadequate circulation, insufficient accumulation at the target region, and undesired toxicity. In this regard, scientists are working on producing cell-membrane camouflaged nanoparticles as a biomimetic technique for modifying the surface of existing nanoparticles to produce significant therapeutic benefits following imparting myriad of desired functionalities. Membranes originating from erythrocytes, white blood cells, cancer cells, stem cells, platelets, or bacterial cells have been used to coat nanoparticle surfaces and create biologically inspired camouflaged nanoparticles. These biomemitic delivery systems have been proven to have potential applications in diagnosing and treating vaiorus diseases, including drug administration, immunisation, immunological regulation, and detoxification. From its inception to the present, we provide a complete description of this advanced technique for functionalizing nanoparticle surfaces. The method of making these membrane coated nanoparticles as well as their characterisation have been thoroughly discussed. Following that, we focused on the diversity of cell membranes derived from distinct cells in the evolution of nanoparticles, emphasising how these biologically inspired stealth - camouflaged techniques have led to increased therapeutic efficacy in a variety of disease states.
Collapse
Affiliation(s)
- Mohammad Imran
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India
| | - Vemana Gowd
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India
| | - Puspita Saha
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh 11623, Saudi Arabia
| | - Marwa Yousry A Mohamed
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh 11623, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh 11623, Saudi Arabia
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge, City, Sector-81, Mohali 140306, Punjab, India.
| |
Collapse
|
11
|
Deka Dey A, Yousefiasl S, Kumar A, Dabbagh Moghaddam F, Rahimmanesh I, Samandari M, Jamwal S, Maleki A, Mohammadi A, Rabiee N, Cláudia Paiva‐Santos A, Tamayol A, Sharifi E, Makvandi P. miRNA-encapsulated abiotic materials and biovectors for cutaneous and oral wound healing: Biogenesis, mechanisms, and delivery nanocarriers. Bioeng Transl Med 2023; 8:e10343. [PMID: 36684081 PMCID: PMC9842058 DOI: 10.1002/btm2.10343] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 01/25/2023] Open
Abstract
MicroRNAs (miRNAs) as therapeutic agents have attracted increasing interest in the past decade owing to their significant effectiveness in treating a wide array of ailments. These polymerases II-derived noncoding RNAs act through post-transcriptional controlling of different proteins and their allied pathways. Like other areas of medicine, researchers have utilized miRNAs for managing acute and chronic wounds. The increase in the number of patients suffering from either under-healing or over-healing wound demonstrates the limited efficacy of the current wound healing strategies and dictates the demands for simpler approaches with greater efficacy. Various miRNA can be designed to induce pathway beneficial for wound healing. However, the proper design of miRNA and its delivery system for wound healing applications are still challenging due to their limited stability and intracellular delivery. Therefore, new miRNAs are required to be identified and their delivery strategy needs to be optimized. In this review, we discuss the diverse roles of miRNAs in various stages of wound healing and provide an insight on the most recent findings in the nanotechnology and biomaterials field, which might offer opportunities for the development of new strategies for this chronic condition. We also highlight the advances in biomaterials and delivery systems, emphasizing their challenges and resolutions for miRNA-based wound healing. We further review various biovectors (e.g., adenovirus and lentivirus) and abiotic materials such as organic and inorganic nanomaterials, along with dendrimers and scaffolds, as the delivery systems for miRNA-based wound healing. Finally, challenges and opportunities for translation of miRNA-based strategies into clinical applications are discussed.
Collapse
Affiliation(s)
| | - Satar Yousefiasl
- School of DentistryHamadan University of Medical SciencesHamadanIran
| | - Arun Kumar
- Chitkara College of PharmacyChitkara UniversityPunjabIndia
| | - Farnaz Dabbagh Moghaddam
- Department of Biology, Science and Research BranchIslamic Azad UniversityTehranIran
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100RomeItaly
| | - Ilnaz Rahimmanesh
- Applied Physiology Research CenterCardiovascular Research Institute, Isfahan University of Medical SciencesIsfahanIran
| | | | - Sumit Jamwal
- Department of Psychiatry, Yale School of MedicineYale UniversityNew HavenConnecticutUSA
| | - Aziz Maleki
- Department of Pharmaceutical Nanotechnology, School of PharmacyZanjan University of Medical SciencesZanjanIran
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjanIran
- Cancer Research CentreShahid Beheshti University of Medical SciencesTehranIran
| | | | - Navid Rabiee
- Department of PhysicsSharif University of TechnologyTehranIran
- School of EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| | - Ana Cláudia Paiva‐Santos
- Department of Pharmaceutical TechnologyFaculty of Pharmacy of the University of Coimbra, University of CoimbraCoimbraPortugal
- LAQV, REQUIMTE, Department of Pharmaceutical TechnologyFaculty of Pharmacy of the University of Coimbra, University of CoimbraCoimbraPortugal
| | - Ali Tamayol
- Department of Biomedical EngineeringUniversity of ConnecticutFarmingtonConnecticutUSA
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and TechnologiesHamadan University of Medical SciencesHamadanIran
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials InterfacesPontederaItaly
- School of Chemistry, Damghan UniversityDamghanIran
| |
Collapse
|
12
|
Michely L, Chesneau C, Dika E, Evrard T, Belbekhouche S. Easy way for fabricating calcium carbonate hybrid microparticles-supported carrier: Focus on the loading of several hydrosoluble cargos all at once. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Li S, Ju Y, Zhou J, Faria M, Ang CS, Mitchell AJ, Zhong QZ, Zheng T, Kent SJ, Caruso F. Protein precoating modulates biomolecular coronas and nanocapsule-immune cell interactions in human blood. J Mater Chem B 2022; 10:7607-7621. [PMID: 35713277 DOI: 10.1039/d2tb00672c] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The biomolecular corona that forms on particles upon contact with blood plays a key role in the fate and utility of nanomedicines. Recent studies have shown that precoating nanoparticles with serum proteins can improve the biocompatibility and stealth properties of nanoparticles. However, it is not fully clear how precoating influences biomolecular corona formation and downstream biological responses. Herein, we systematically examine three precoating strategies by coating bovine serum albumin (single protein), fetal bovine serum (FBS, mixed proteins without immunoglobulins), or bovine serum (mixed proteins) on three nanoparticle systems, namely supramolecular template nanoparticles, metal-phenolic network (MPN)-coated template (core-shell) nanoparticles, and MPN nanocapsules (obtained after template removal). The effect of protein precoating on biomolecular corona compositions and particle-immune cell interactions in human blood was characterized. In the absence of a pre-coating, the MPN nanocapsules displayed lower leukocyte association, which correlated to the lower amount (by 2-3 fold) of adsorbed proteins and substantially fewer immunoglobulins (more than 100 times) in the biomolecular corona relative to the template and core-shell nanoparticles. Among the three coating strategies, FBS precoating demonstrated the most significant reduction in leukocyte association (up to 97% of all three nanoparticles). A correlation analysis highlights that immunoglobulins and apolipoproteins may regulate leukocyte recognition. This study demonstrates the impact of different precoating strategies on nanoparticle-immune cell association and the role of immunoglobulins in bio-nano interactions.
Collapse
Affiliation(s)
- Shiyao Li
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia. .,School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Jiajing Zhou
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Matthew Faria
- Systems Biology Laboratory, School of Mathematics and Statistics, and the Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andrew J Mitchell
- Department of Chemical Engineering, Materials Characterisation and Fabrication Platform, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Qi-Zhi Zhong
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Tian Zheng
- Department of Chemical Engineering, Materials Characterisation and Fabrication Platform, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
14
|
Konhäuser M, Kannaujiya VK, Steiert E, Schwickert K, Schirmeister T, Wich PR. Co-Encapsulation of l-Asparaginase and Etoposide in Dextran Nanoparticles for Synergistic Effect in Chronic Myeloid Leukemia Cells. Int J Pharm 2022; 622:121796. [PMID: 35525474 DOI: 10.1016/j.ijpharm.2022.121796] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022]
Abstract
Co-encapsulation of multiple therapeutic drugs in a single nanocarrier has the potential to enable synergistic interactions, increase drug efficacy, and reduce side effects. The enzyme l-asparaginase and the small molecule drug etoposide have a known synergistic effect against selected cancer types. However, both drugs differ significantly in size, molecular weight, and solubility, which often results in challenges when a simultaneous delivery is required. In this study, we present the co-encapsulation of a large hydrophilic enzyme l-asparaginase and the small hydrophobic drug etoposide into a biodegradable, biocompatible, and acid-responsive dextran-based nanoparticle system. These dual drug-loaded nanoparticles show an excellent cellular uptake in chronic myeloid leukemia (CML) K562 cells and a stepwise release of the cytotoxic payloads in a pH-dependent manner. In activity tests, the dual drug-loaded formulation has shown a significant effect on cell viability (down to 31%) compared to those incubated only with l-asparaginase (92%) or etoposide (82%) at a particle concentration of 125 μg∙mL-1. These results show that the simultaneous co-delivery of these two drugs in K562 cells leads to synergistic cytotoxicity, indicating a great potential for the treatment of CML.
Collapse
Affiliation(s)
- M Konhäuser
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - V K Kannaujiya
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - E Steiert
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - K Schwickert
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - T Schirmeister
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - P R Wich
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany; School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
15
|
Imran M, Akhileshwar Jha L, Hasan N, Shrestha J, Pangeni R, Parvez N, Mohammed Y, Kumar Jha S, Raj Paudel K. “Nanodecoys”- Future of drug delivery by encapsulating nanoparticles in natural cell membranes. Int J Pharm 2022; 621:121790. [DOI: 10.1016/j.ijpharm.2022.121790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/12/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022]
|
16
|
Zeng Y, Xiang Y, Sheng R, Tomás H, Rodrigues J, Gu Z, Zhang H, Gong Q, Luo K. Polysaccharide-based nanomedicines for cancer immunotherapy: A review. Bioact Mater 2021; 6:3358-3382. [PMID: 33817416 PMCID: PMC8005658 DOI: 10.1016/j.bioactmat.2021.03.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy is an effective antitumor approach through activating immune systems to eradicate tumors by immunotherapeutics. However, direct administration of "naked" immunotherapeutic agents (such as nucleic acids, cytokines, adjuvants or antigens without delivery vehicles) often results in: (1) an unsatisfactory efficacy due to suboptimal pharmacokinetics; (2) strong toxic and side effects due to low targeting (or off-target) efficiency. To overcome these shortcomings, a series of polysaccharide-based nanoparticles have been developed to carry immunotherapeutics to enhance antitumor immune responses with reduced toxicity and side effects. Polysaccharides are a family of natural polymers that hold unique physicochemical and biological properties, as they could interact with immune system to stimulate an enhanced immune response. Their structures offer versatility in synthesizing multifunctional nanocomposites, which could be chemically modified to achieve high stability and bioavailability for delivering therapeutics into tumor tissues. This review aims to highlight recent advances in polysaccharide-based nanomedicines for cancer immunotherapy and propose new perspectives on the use of polysaccharide-based immunotherapeutics.
Collapse
Affiliation(s)
- Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yufan Xiang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruilong Sheng
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - Helena Tomás
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - João Rodrigues
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
17
|
Losada-Garcia N, Garcia-Sanz C, Andreu A, Velasco-Torrijos T, Palomo JM. Glyconanomaterials for Human Virus Detection and Inhibition. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1684. [PMID: 34206886 PMCID: PMC8308178 DOI: 10.3390/nano11071684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/23/2023]
Abstract
Viruses are among the most infectious pathogens, responsible for the highest death toll around the world. Lack of effective clinical drugs for most viral diseases emphasizes the need for speedy and accurate diagnosis at early stages of infection to prevent rapid spread of the pathogens. Glycans are important molecules which are involved in different biological recognition processes, especially in the spread of infection by mediating virus interaction with endothelial cells. Thus, novel strategies based on nanotechnology have been developed for identifying and inhibiting viruses in a fast, selective, and precise way. The nanosized nature of nanomaterials and their exclusive optical, electronic, magnetic, and mechanical features can improve patient care through using sensors with minimal invasiveness and extreme sensitivity. This review provides an overview of the latest advances of functionalized glyconanomaterials, for rapid and selective biosensing detection of molecules as biomarkers or specific glycoproteins and as novel promising antiviral agents for different kinds of serious viruses, such as the Dengue virus, Ebola virus, influenza virus, human immunodeficiency virus (HIV), influenza virus, Zika virus, or coronavirus SARS-CoV-2 (COVID-19).
Collapse
Affiliation(s)
- Noelia Losada-Garcia
- Department of Biocatalysis, Institute of Catalysis (CSIC), Marie Curie 2, 28049 Madrid, Spain; (N.L.-G.); (C.G.-S.); (A.A.)
| | - Carla Garcia-Sanz
- Department of Biocatalysis, Institute of Catalysis (CSIC), Marie Curie 2, 28049 Madrid, Spain; (N.L.-G.); (C.G.-S.); (A.A.)
| | - Alicia Andreu
- Department of Biocatalysis, Institute of Catalysis (CSIC), Marie Curie 2, 28049 Madrid, Spain; (N.L.-G.); (C.G.-S.); (A.A.)
| | | | - Jose M. Palomo
- Department of Biocatalysis, Institute of Catalysis (CSIC), Marie Curie 2, 28049 Madrid, Spain; (N.L.-G.); (C.G.-S.); (A.A.)
| |
Collapse
|
18
|
Li Z, Bratlie KM. The Influence of Polysaccharides-Based Material on Macrophage Phenotypes. Macromol Biosci 2021; 21:e2100031. [PMID: 33969643 DOI: 10.1002/mabi.202100031] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 02/03/2023]
Abstract
Macrophage polarization is a key factor in determining the success of implanted tissue engineering scaffolds. Polysaccharides (derived from plants, animals, and microorganisms) are known to modulate macrophage phenotypes by recognizing cell membrane receptors. Numerous studies have developed polysaccharide-based materials into functional biomaterial substrates for tissue regeneration and pharmaceutical application due to their immunostimulatory activities and anti-inflammatory response. They are used as hydrogel substrates, surface coatings, and drug delivery carriers. In addition to their innate immunological functions, the newly endowed physical and chemical properties, including substrate modulus, pore size/porosity, surface binding chemistry, and the mole ratio of polysaccharides in hybrid materials may regulate macrophage phenotypes more precisely. Growing evidence indicates that the sulfation pattern of glycosaminoglycans and proteoglycans expressed on polarized macrophages leads to the changes in protein binding, which may alter macrophage phenotype and influence the immune response. A comprehensive understanding of how different types of polysaccharide-based materials alter macrophage phenotypic changes can be beneficial to predict transplantation/implantation outcomes. This review focuses on recent advances in promoting wound healing and balancing macrophage phenotypes using polysaccharide-based substrates/coatings and new directions to address the limitations in the current understanding of macrophage responses to polysaccharides.
Collapse
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA.,Department of Chemical & Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
19
|
Wang S, Fontana F, Shahbazi MA, Santos HA. Acetalated dextran based nano- and microparticles: synthesis, fabrication, and therapeutic applications. Chem Commun (Camb) 2021; 57:4212-4229. [PMID: 33913978 DOI: 10.1039/d1cc00811k] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acetalated dextran (Ac-DEX) is a pH-responsive dextran derivative polymer. Prepared by a simple acetalation reaction, Ac-DEX has tunable acid-triggered release profile. Despite its relatively short research history, Ac-DEX has shown great potential in various therapeutic applications. Furthermore, the recent functionalization of Ac-DEX makes versatile derivatives with additional properties. Herein, we summarize the cutting-edge development of Ac-DEX and related polymers. Specifically, we focus on the chemical synthesis, nano- and micro-particle fabrication techniques, the controlled-release mechanisms, and the rational design Ac-DEX-based of drug delivery systems in various biomedical applications. Finally, we briefly discuss the challenges and future perspectives in the field.
Collapse
Affiliation(s)
- Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran and Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
20
|
Zhang M, Cheng S, Jin Y, Zhang N, Wang Y. Membrane engineering of cell membrane biomimetic nanoparticles for nanoscale therapeutics. Clin Transl Med 2021; 11:e292. [PMID: 33635002 PMCID: PMC7819108 DOI: 10.1002/ctm2.292] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, cell membrane camouflaging technology has emerged as an important strategy of nanomedicine, and the modification on the membranes is also a promising approach to enhance the properties of the nanoparticles, such as cancer targeting, immune evasion, and phototherapy sensitivity. Indeed, diversified approaches have been exploited to re-engineer the membranes of nanoparticles in several studies. In this review, first we discuss direct modification strategy of cell membrane camouflaged nanoparticles (CM-NP) via noncovalent, covalent, and enzyme-involved methods. Second, we explore how the membranes of CM-NPs can be re-engineered at the cellular level using strategies such as genetic engineering and membranes fusion. Due to the innate biological properties and excellent biocompatibility, the functionalized cell membrane-camouflaged nanoparticles have been widely applied in the fields of drug delivery, imaging, detoxification, detection, and photoactivatable therapy.
Collapse
Affiliation(s)
- Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yue Jin
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Nan Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yu Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
- Shanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| |
Collapse
|
21
|
Plucinski A, Lyu Z, Schmidt BVKJ. Polysaccharide nanoparticles: from fabrication to applications. J Mater Chem B 2021; 9:7030-7062. [DOI: 10.1039/d1tb00628b] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The present review highlights the developments in polysaccharide nanoparticles with a particular focus on applications in biomedicine, cosmetics and food.
Collapse
Affiliation(s)
| | - Zan Lyu
- School of Chemistry, University of Glasgow, G12 8QQ Glasgow, UK
| | | |
Collapse
|
22
|
Huang Y, Nan L, Xiao C, Su F, Li K, Ji QA, Wei Q, Liu Y, Bao G. PEGylated nano-Rehmannia glutinosa polysaccharide induces potent adaptive immunity against Bordetella bronchiseptica. Int J Biol Macromol 2020; 168:507-517. [PMID: 33310103 DOI: 10.1016/j.ijbiomac.2020.12.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 12/18/2022]
Abstract
Vaccines, in many cases, stimulate only too weak immunogenicity to prevent infection. Therefore, adjuvants are required during their preparation to boost the immune response. We herein developed a PEGylated nano-adjuvant based on Rehmannia glutinosa polysaccharide (RGP). The addition of PEG layer exhibits enhanced immune performance of the nano-RGP. Stimulation of dendritic cells (DCs) with PEGylated nano-RGP (pRL) led to increased proliferation and cytokine production (IL-6, IL-12, IL-1β and TNF-α). The pRL was internalized into DCs via a rapid and efficient method. The mice immunized with pRL exhibited enhanced antigen-specific serum IgG and Th1-(IFN-γ), Th2-(IL-4), and Th17-(IL-17, IL-6) cytokine production, contributing to a good anti-infection performance. Furthermore, the pRL could effectively deliver the antigen to the lymph nodes (LNs), activate DC in the LN and produce enhanced CD4+and CD8+ T-cells-derived memory (CD44high CD62Lhigh), and effector (CD44high CD62Llow) as well as functional phenotypes. Our results revealed that pRL can act as a promising adjuvant with targeted delivery of antigen due to its effective activation and robust adaptive immunity induction of DCs.
Collapse
Affiliation(s)
- Yee Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Li Nan
- Zhejiang Normal University, Jinhua 321000, PR China
| | - Chenwen Xiao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Fei Su
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Ke Li
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Quan-An Ji
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Qiang Wei
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Yan Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China
| | - Guolian Bao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021, PR China.
| |
Collapse
|
23
|
Skalickova S, Horky P, Mlejnkova V, Skladanka J, Hosnedlova B, Ruttkay‐Nedecky B, Fernandez C, Kizek R. Theranostic Approach for the Protein Corona of Polysaccharide Nanoparticles. CHEM REC 2020; 21:17-28. [DOI: 10.1002/tcr.202000042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/22/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Sylvie Skalickova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy Masaryk University Palackeho 1946/1 612 00 Brno Czech Republic
- Department of Animal Nutrition and Forage Production Mendel University in Brno Zemedelska 1 613 00 Brno Czech Republic
| | - Pavel Horky
- Department of Animal Nutrition and Forage Production Mendel University in Brno Zemedelska 1 613 00 Brno Czech Republic
| | - Veronika Mlejnkova
- Department of Animal Nutrition and Forage Production Mendel University in Brno Zemedelska 1 613 00 Brno Czech Republic
| | - Jiri Skladanka
- Department of Animal Nutrition and Forage Production Mendel University in Brno Zemedelska 1 613 00 Brno Czech Republic
| | - Bozena Hosnedlova
- Department of Research and Development Prevention Medicals Tovarni 342 742 13 Studenka-Butovice Czech Republic
- Department of Viticulture and Enology, Faculty of Horticulture Mendel University in Brno Valticka 337 CZ-691 44 Lednice Czech Republic
| | - Branislav Ruttkay‐Nedecky
- Department of Research and Development Prevention Medicals Tovarni 342 742 13 Studenka-Butovice Czech Republic
- Department of Viticulture and Enology, Faculty of Horticulture Mendel University in Brno Valticka 337 CZ-691 44 Lednice Czech Republic
- Department of Molecular Pharmacy, Faculty of Pharmacy Masaryk University Palackeho 1946/1 612 00 Brno Czech Republic
| | - Carlos Fernandez
- School of Pharmacy and Life Sciences Robert Gordon University Garthdee Road AB10 7QB Aberdeen UK
| | - Rene Kizek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy Masaryk University Palackeho 1946/1 612 00 Brno Czech Republic
- Department of Research and Development Prevention Medicals Tovarni 342 742 13 Studenka-Butovice Czech Republic
- Department of Viticulture and Enology, Faculty of Horticulture Mendel University in Brno Valticka 337 CZ-691 44 Lednice Czech Republic
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy with Division of Laboratory Medicine Wroclaw Medical University Borowska 211 50-556 Wroclaw Poland
| |
Collapse
|
24
|
Nguyen HV, Campbell K, Painter GF, Young SL, Walker GF. Nanoparticle System Based on Amino-Dextran as a Drug Delivery Vehicle: Immune-Stimulatory CpG-Oligonucleotide Loading and Delivery. Pharmaceutics 2020; 12:pharmaceutics12121150. [PMID: 33260874 PMCID: PMC7760314 DOI: 10.3390/pharmaceutics12121150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 11/30/2022] Open
Abstract
The aim of this study is to prepare and characterize an amino-dextran nanoparticle (aDNP) platform and investigate two loading strategies for unmethylated cytosine-phosphate-guanine (CpG) oligonucleotide. aDNP was prepared by desolvation of amino-dextran followed by the chemical crosslinking of amino groups. Size, surface charge, and surface morphology of aDNP was determined by dynamic light scattering and transmission electron microscopy. CpG was either loaded onto aDNP by adsorption (CpG-adsorbed-aDNP) or conjugated to aDNP (CpG-conjugated-aDNP). In vitro cytokine production by bone marrow-derived dendritic cells (BMDCs) was measured by flow cytometry. aDNPs size and zeta potential could be controlled to produce uniform particles in the size range of 50 to 300 nm, surface charge of −16.5 to +14 mV, and were spherical in shape. Formulation control parameters investigated included the anti-solvent, water-to-anti-solvent ratio, level of amine functionality of dextran, and the molar ratio of glutaraldehyde to amine. aDNP could be lyophilized without additional cryoprotectant. Unloaded cationic aDNP (+13 mV) showed acceptable in vitro hemolysis. Unloaded and CpG-loaded aDNPs showed no cytotoxicity on BMDCs. CpG-loaded nanoparticles stimulated cytokine production by BMDCs, the level of cytokine production was higher for CpG-conjugated-aDNP compared to CpG-absorbed-aDNP. aDNP is a promising new drug delivery platform as its offers versatility in loading and tuning of particle properties.
Collapse
Affiliation(s)
- Hien V. Nguyen
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand;
- Department of Pathology, University of Otago, Dunedin 9016, New Zealand;
| | - Katrin Campbell
- Department of Pathology, University of Otago, Dunedin 9016, New Zealand;
| | - Gavin F. Painter
- The Ferrier Research Institute, Victoria University of Wellington, Wellington 5040, New Zealand;
| | - Sarah L. Young
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2006, NSW, Australia;
| | - Greg F. Walker
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand;
- Correspondence: ; Tel.: +64-3-479-7133
| |
Collapse
|
25
|
Moore KM, Batty CJ, Stiepel RT, Genito CJ, Bachelder EM, Ainslie KM. Injectable, Ribbon-Like Microconfetti Biopolymer Platform for Vaccine Applications. ACS APPLIED MATERIALS & INTERFACES 2020; 12:38950-38961. [PMID: 32805875 PMCID: PMC7484345 DOI: 10.1021/acsami.0c10276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Previously, high-aspect- ratio ribbon-like microconfetti (MC) composed of acetalated dextran (Ace-DEX) have been shown to form a subcutaneous depot for sustained drug release. In this study, MC were explored as an injectable vaccine platform. Production of MC by electrospinning followed by high-shear homogenization allowed for precise control over MC fabrication. Three distinct sizes of MC, small (0.67 × 10.2 μm2), medium (1.28 × 20.7 μm2), and large (5.67 × 90.2 μm2), were fabricated and loaded with the adjuvant, resiquimod. Steady release rates of resiquimod were observed from MC, indicating their ability to create an immunostimulatory depot in vivo. Resiquimod-loaded MC stimulated inflammatory cytokine production in bone marrow-derived dendritic cells without incurring additional cytotoxicity in vitro. Interestingly, even medium and large MC were able to be internalized by antigen-presenting cells and facilitate antigen presentation when ovalbumin was adsorbed onto their surface. After subcutaneous injection in vivo with adsorbed ovalbumin, blank MC of all sizes were found to stimulate a humoral response. Adjuvant activity of resiquimod was enhanced by loading it into MC and small- and medium-sized MC effectively induced a Th1-skewed immune response. Antigen co-delivered with adjuvant-loaded MC of various sizes illustrates a new potential vaccine platform.
Collapse
Affiliation(s)
- Kathryn M. Moore
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Christopher J. Genito
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M. Ainslie
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
26
|
Wang H, Liu Y, He R, Xu D, Zang J, Weeranoppanant N, Dong H, Li Y. Cell membrane biomimetic nanoparticles for inflammation and cancer targeting in drug delivery. Biomater Sci 2020; 8:552-568. [PMID: 31769765 DOI: 10.1039/c9bm01392j] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanoparticle capture and elimination by the immune system are great obstacles for drug delivery. Camouflaging nanoparticles with cell membrane represents a promising strategy to communicate and negotiate with the immune system. As a novel class of nanotherapeutics, such biomimetic nanoparticles inherit specific biological functionalities of the source cells (e.g., erythrocytes, immune cells, cancer cells and platelets) in order to evade immune elimination, prolong circulation time, and even target a disease region by virtue of the homing tendency of the cell membrane protein. In this review, we begin with an overview of different cell membranes that can be utilized to create a biointerface on nanoparticles. Subsequently, we elaborate on the state-of-the-art of cell membrane biomimetic nanoparticles for drug delivery. In particular, a summary of data on circulation capacity and targeting efficiency by camouflaged nanoparticles is presented. In addition to cancer therapy, inflammation treatment, as an emerging application of biomimetic nanoparticles, is specifically included. The challenges and outlook of this technology are discussed.
Collapse
Affiliation(s)
- Huaiji Wang
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ghaeini-Hesaroeiye S, Razmi Bagtash H, Boddohi S, Vasheghani-Farahani E, Jabbari E. Thermoresponsive Nanogels Based on Different Polymeric Moieties for Biomedical Applications. Gels 2020; 6:E20. [PMID: 32635573 PMCID: PMC7559285 DOI: 10.3390/gels6030020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
Nanogels, or nanostructured hydrogels, are one of the most interesting materials in biomedical engineering. Nanogels are widely used in medical applications, such as in cancer therapy, targeted delivery of proteins, genes and DNAs, and scaffolds in tissue regeneration. One salient feature of nanogels is their tunable responsiveness to external stimuli. In this review, thermosensitive nanogels are discussed, with a focus on moieties in their chemical structure which are responsible for thermosensitivity. These thermosensitive moieties can be classified into four groups, namely, polymers bearing amide groups, ether groups, vinyl ether groups and hydrophilic polymers bearing hydrophobic groups. These novel thermoresponsive nanogels provide effective drug delivery systems and tissue regeneration constructs for treating patients in many clinical applications, such as targeted, sustained and controlled release.
Collapse
Affiliation(s)
- Sobhan Ghaeini-Hesaroeiye
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14115, Iran; (S.G.-H.); (H.R.B.)
| | - Hossein Razmi Bagtash
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14115, Iran; (S.G.-H.); (H.R.B.)
| | - Soheil Boddohi
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14115, Iran; (S.G.-H.); (H.R.B.)
| | - Ebrahim Vasheghani-Farahani
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14115, Iran; (S.G.-H.); (H.R.B.)
| | - Esmaiel Jabbari
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA;
| |
Collapse
|
28
|
Acid-responsive dextran-based therapeutic nanoplatforms for photodynamic-chemotherapy against multidrug resistance. Int J Biol Macromol 2020; 155:233-240. [DOI: 10.1016/j.ijbiomac.2020.03.197] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 11/21/2022]
|
29
|
Barry MA, Rubin JD, Lu SC. Retargeting adenoviruses for therapeutic applications and vaccines. FEBS Lett 2020; 594:1918-1946. [PMID: 31944286 PMCID: PMC7311308 DOI: 10.1002/1873-3468.13731] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022]
Abstract
Adenoviruses (Ads) are robust vectors for therapeutic applications and vaccines, but their use can be limited by differences in their in vitro and in vivo pharmacologies. This review emphasizes that there is not just one Ad, but a whole virome of diverse viruses that can be used as therapeutics. It discusses that true vector targeting involves not only retargeting viruses, but importantly also detargeting the viruses from off-target cells.
Collapse
Affiliation(s)
- Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Department of Immunology, Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey D Rubin
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Shao-Chia Lu
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
Braga CB, Kido LA, Lima EN, Lamas CA, Cagnon VHA, Ornelas C, Pilli RA. Enhancing the Anticancer Activity and Selectivity of Goniothalamin Using pH-Sensitive Acetalated Dextran (Ac-Dex) Nanoparticles: A Promising Platform for Delivery of Natural Compounds. ACS Biomater Sci Eng 2020; 6:2929-2942. [PMID: 33463303 DOI: 10.1021/acsbiomaterials.0c00057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Goniothalamin (GTN), a natural compound isolated from Goniothalamus species, has previously demonstrated cytotoxic activity against several cancer cell lines. However, similarly to many natural and synthetic anticancer compounds, GTN presents toxicity toward some healthy cells and low aqueous solubility, decreasing its bioavailability and precluding its application as an antineoplastic drug. In our efforts to improve the pharmacokinetic behavior and selectivity of GTN against cancer cells, we developed a polymeric nanosystem, in which rac-GTN was encapsulated in pH-responsive acetalated dextran (Ac-Dex) nanoparticles (NPs) with high loadings of the bioactive compound. Dynamic light scattering (DLS) analysis showed that the nanoparticles obtained presented a narrow size distribution of around 100 nm in diameter, whereas electron microscopy (EM) images showed nanoparticles with a regular spherical morphology in agreement with the size range obtained by DLS. Stability and release studies indicated that the GTN@Ac-Dex NPs presented high stability under physiological conditions (pH 7.4) and disassembled under slightly acidic conditions (pH 5.5), releasing the rac-GTN in a sustained manner. In vitro assays showed that GTN@Ac-Dex NPs significantly increased cytotoxicity and selectivity against cancer cells when compared with the empty Ac-Dex NPs and the free rac-GNT. Cellular uptake and morphology studies using MCF-7 cells demonstrated that GTN@Ac-Dex NPs are rapidly internalized into the cancer cells, causing cell death. In vivo investigation confirmed the efficient release of rac-GTN from GTN@Ac-Dex NPs, resulting in the delay of prostate cancer progression in transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Furthermore, liver histopathology evaluation after treatment with GTN@Ac-Dex NPs showed no evidence of toxicity. Therefore, the in vitro and in vivo findings suggest that the Ac-Dex NPs are a promising nanosystem for the sustained delivery of rac-GTN into tumors.
Collapse
Affiliation(s)
- Carolyne B Braga
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), P.O. Box 6154, CEP 13083-970 Campinas, São Paulo, Brazil
| | - Larissa A Kido
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, CEP 13083-865 Campinas, São Paulo, Brazil
| | - Ellen N Lima
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, CEP 13083-865 Campinas, São Paulo, Brazil
| | - Celina A Lamas
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, CEP 13083-865 Campinas, São Paulo, Brazil
| | - Valéria H A Cagnon
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, CEP 13083-865 Campinas, São Paulo, Brazil
| | - Catia Ornelas
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), P.O. Box 6154, CEP 13083-970 Campinas, São Paulo, Brazil
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), P.O. Box 6154, CEP 13083-970 Campinas, São Paulo, Brazil
| |
Collapse
|
31
|
Clickable modular polysaccharide nanoparticles for selective cell-targeting. Carbohydr Polym 2020; 234:115901. [PMID: 32070522 DOI: 10.1016/j.carbpol.2020.115901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/03/2020] [Accepted: 01/20/2020] [Indexed: 12/11/2022]
Abstract
A therapeutic nanocarrier capable of cell targeting has the potential to reduce off-target effects of otherwise effective drugs. Nanoparticle surface modification can be tailored for specific cells, however multistep surface modification can prove slow and difficult for a variety of cell types. Here, we designed drug carrying polysaccharide based nanoparticles with a layered structure for clickable surface modification. The center of nanoparticle was composed of cationic macromer (e.g., poly-l-lysine) and anionic polysaccharide (e.g., heparin). Furthermore, a 'clickable' polysaccharide was installed on the surface of the nanoparticles to permit a wide range of bioconjugation via norbornene-tetrazine click chemistry. The utilities of these layered nanoparticles were demonstrated via enhanced protein sequestration, selective cell targeting (via PEGylation or altering polysaccharide coating), as well as loading and release of chemotherapeutic. The drug-loaded nanocarriers proved cytotoxic to J774A.1 monocytes and MOLM-14 leukemia cells.
Collapse
|
32
|
Schulz E, Karagianni A, Koch M, Fuhrmann G. Hot EVs - How temperature affects extracellular vesicles. Eur J Pharm Biopharm 2019; 146:55-63. [PMID: 31805356 DOI: 10.1016/j.ejpb.2019.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/15/2019] [Accepted: 11/27/2019] [Indexed: 12/25/2022]
Abstract
In recent years, extracellular vesicles (EVs) and outer membrane vesicles (OMVs) have become an extensive and diverse field of research. They hold potential as diagnostic markers, therapeutics and for fundamental biological understanding. Despite ongoing studies, numerous information regarding function, content and stability of EVs remains unclear. If EVs and OMVs ought to be used as therapeutics and in clinical environments, their stability is one of the most important factors to be considered. Especially for formulation development, EVs and OMVs need to be stable at higher temperatures. To the best of our knowledge, very little work has been published regarding heat stability of neither EVs nor OMVs. In the present study, we investigated B lymphoblastoid cell-derived EVs and OMVs derived from myxobacterial species Sorangiineae as model vesicles. We exposed the vesicles to 37 °C, 50 °C, 70 °C and 100 °C for 1 h, 6 h and 24 h, and also autoclaved them. Interestingly, physico-chemical analyses such as size, particle concentration and protein concentration showed minor alterations, particularly at 37 °C. Flow cytometry analysis emphasised these results suggesting that after heat impact, EVs and OMVs were still able to be taken up by macrophage-like dTHP-1 cells. These data indicate that both mammalian and bacterial vesicles show intrinsic stability at physiological temperature. Our findings are important to consider for vesicle formulation and for advanced bioengineering approaches.
Collapse
Affiliation(s)
- Eilien Schulz
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken 66123, Germany
| | - Anna Karagianni
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2.2, Saarbrücken 66123, Germany
| | - Gregor Fuhrmann
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken 66123, Germany.
| |
Collapse
|
33
|
Butzbach K, Konhäuser M, Fach M, Bamberger DN, Breitenbach B, Epe B, Wich PR. Receptor-mediated Uptake of Folic Acid-functionalized Dextran Nanoparticles for Applications in Photodynamic Therapy. Polymers (Basel) 2019; 11:polym11050896. [PMID: 31100893 PMCID: PMC6572481 DOI: 10.3390/polym11050896] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 01/26/2023] Open
Abstract
In photodynamic therapy (PDT), photosensitizers and light are used to cause photochemically induced cell death. The selectivity and the effectiveness of the phototoxicity in cancer can be increased by a specific uptake of the photosensitizer into tumor cells. A promising target for this goal is the folic acid receptor α (FRα), which is overexpressed on the surface of many tumor cells and mediates an endocytotic uptake. Here, we describe a polysaccharide-based nanoparticle system suitable for targeted uptake and its photochemical and photobiological characterization. The photosensitizer 5, 10, 15, 20-tetraphenyl-21H, 23H-porphyrine (TPP) was encapsulated in spermine- and acetal-modified dextran (SpAcDex) nanoparticles and conjugated with folic acid (FA) on the surface [SpAcDex(TPP)-FA]. The particles are successfully taken up by human HeLa-KB cells, and a light-induced cytotoxicity is observable. An excess of free folate as the competitor for the FRα-mediated uptake inhibits the phototoxicity. In conclusion, folate-modified SpAcDex particles are a promising drug delivery system for a tumor cell targeted photodynamic therapy.
Collapse
Affiliation(s)
- Kathrin Butzbach
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudingerweg 5, 55128 Mainz, Germany
| | - Matthias Konhäuser
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudingerweg 5, 55128 Mainz, Germany
| | - Matthias Fach
- Department of Health Technology, Technical University of Denmark, Produktionstorvet Building 423, 2800 Lyngby, Denmark
| | - Denise N Bamberger
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudingerweg 5, 55128 Mainz, Germany
| | - Benjamin Breitenbach
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudingerweg 5, 55128 Mainz, Germany
| | - Bernd Epe
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudingerweg 5, 55128 Mainz, Germany
| | - Peter R Wich
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudingerweg 5, 55128 Mainz, Germany.
- School of Chemical Engineering, University of New South Wales, Science and Engineering Building, Sydney, NSW 2052, Australia.
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
34
|
Breitenbach BB, Steiert E, Konhäuser M, Vogt LM, Wang Y, Parekh SH, Wich PR. Double stimuli-responsive polysaccharide block copolymers as green macrosurfactants for near-infrared photodynamic therapy. SOFT MATTER 2019; 15:1423-1434. [PMID: 30662988 DOI: 10.1039/c8sm02204f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The NIR absorbing photosensitizer phthalocyanine zinc (PC(Zn)) was stabilized in aqueous media as water-dispersible nanoparticles with a reduction- and pH-responsive full polysaccharide block copolymer. A cellular uptake and also photo switchable intracellular activity of the cargo upon irradiation at wavelengths in the near infrared region were shown. The block copolymer was synthesized by applying a copper-free click strategy based on a thiol exchange reaction, creating an amphiphilic double-stimuli-responsive mixed disulfide. The dual-sensitive polysaccharide micelles represent a non-toxic and biodegradable green macrosurfactant for the delivery of phthalocyanine zinc. By encapsulation into micellar nanoparticles, the bioavailability of PC(Zn) increased significantly, enabling smart photodynamic therapy for future applications in cancer-related diseases.
Collapse
Affiliation(s)
- Benjamin B Breitenbach
- Institut für Pharmazie und Biochemie, Johannes Gutenberg-Universität Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Geskovski N, Sazdovska SD, Goracinova K. Macroalgal Polysaccharides in Biomimetic Nanodelivery Systems. Curr Pharm Des 2019; 25:1265-1289. [PMID: 31020934 DOI: 10.2174/1381612825666190423155116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Imitating nature in the design of bio-inspired drug delivery systems resulted in several success stories. However, the practical application of biomimicry is still largely unrealized owing to the fact that we tend to copy the shape more often than the whole biology. Interesting chemistry of polysaccharides provides endless possibilities for drug complex formation and creation of delivery systems with diverse morphological and surface properties. However, the type of biological response, which may be induced by these systems, remains largely unexploited. METHODS Considering the most current research for the given topic, in this review, we will try to present the integrative approaches for the design of biomimetic DDS's with improved therapeutic or theranostic effects based on different algal polysaccharides that exert multiple biological functions. RESULTS Algal polysaccharides may provide building blocks for bioinspired drug delivery systems capable of supporting the mechanical properties of nanomedicines and mimicking various biological processes by molecular interactions at the nanoscale. Numerous research studies demonstrate the efficacy and safety of multifunctional nanoparticles integrating several functions in one delivery system, composed of alginate, carrageenan, ulvan, fucoidan and their derivatives, intended to be used as bioartificial microenvironment or for diagnosis and therapy of different diseases. CONCLUSION Nanodimensional structure of polysaccharide DDS's shows substantial influence on the bioactive motifs potential availability for interaction with a variety of biomolecules and cells. Evaluation of the nano dimensional structure-activity relationship is crucial for unlocking the full potential of the future application of polysaccharide bio-mimicking DDS in modern diagnostic and therapeutic procedures.
Collapse
Affiliation(s)
- Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss Cyril and Methodius, Skopje, Republic of North Macedonia
| | - Simona Dimchevska Sazdovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss Cyril and Methodius, Skopje, Republic of North Macedonia
| | | |
Collapse
|
36
|
Wang JL, Du XJ, Yang JX, Shen S, Li HJ, Luo YL, Iqbal S, Xu CF, Ye XD, Cao J, Wang J. The effect of surface poly(ethylene glycol) length on in vivo drug delivery behaviors of polymeric nanoparticles. Biomaterials 2018; 182:104-113. [PMID: 30114562 DOI: 10.1016/j.biomaterials.2018.08.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/29/2022]
Abstract
Engineering nanoparticles of reasonable surface poly(ethylene glycol) (PEG) length is important for designing efficient drug delivery systems. Eliminating the disturbance by other nanoproperties, such as size, PEG density, etc., is crucial for systemically investigating the impact of surface PEG length on the biological behavior of nanoparticles. In the present study, nanoparticles with different surface PEG length but similar other nanoproperties were prepared by using poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) copolymers of different molecular weights and incorporating different contents of PCL3500 homopolymer. The molecular weight of PEG block in PEG-PCL was between 3400 and 8000 Da, the sizes of nanoparticles were around 100 nm, the terminal PEG density was controlled at 0.4 PEG/nm2 (or the frontal PEG density was controlled at 0.16 PEG/nm2). Using these nanoproperties well-designed nanoparticles, we demonstrated PEG length-dependent changes in the biological behaviors of nanoparticles and exhibited nonmonotonic improvements as the PEG molecular weight increased from 3400 to 8000 Da. Moreover, under the experimental conditions, we found nanoparticles with a surface PEG length of 13.8 nm (MW = 5000 Da) significantly decreased the absorption with serum protein and interaction with macrophages, which led to prolonged blood circulation time, enhanced tumor accumulation and improved antitumor efficacy. The present study will help to establish a relatively precise relationship between surface PEG length and the in vivo behavior of nanoparticles.
Collapse
Affiliation(s)
- Ji-Long Wang
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China
| | - Xiao-Jiao Du
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
| | - Jin-Xian Yang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Song Shen
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China
| | - Hong-Jun Li
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China
| | - Ying-Li Luo
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Shoaib Iqbal
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Cong-Fei Xu
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Xiao-Dong Ye
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Jie Cao
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, PR China.
| | - Jun Wang
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, PR China; Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
37
|
Bros M, Nuhn L, Simon J, Moll L, Mailänder V, Landfester K, Grabbe S. The Protein Corona as a Confounding Variable of Nanoparticle-Mediated Targeted Vaccine Delivery. Front Immunol 2018; 9:1760. [PMID: 30116246 PMCID: PMC6082927 DOI: 10.3389/fimmu.2018.01760] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023] Open
Abstract
Nanocarriers (NC) are very promising tools for cancer immunotherapy. Whereas conventional vaccines are based on the administration of an antigen and an adjuvant in an independent fashion, nanovaccines can facilitate cell-specific co-delivery of antigen and adjuvant. Furthermore, nanovaccines can be decorated on their surface with molecules that facilitate target-specific antigen delivery to certain antigen-presenting cell types or tumor cells. However, the target cell-specific uptake of nanovaccines is highly dependent on the modifications of the nanocarrier itself. One of these is the formation of a protein corona around NC after in vivo administration, which may potently affect cell-specific targeting and uptake of the NC. Understanding the formation and composition of the protein corona is, therefore, of major importance for the use of nanocarriers in vaccine approaches. This Mini Review will give a short overview of potential non-specific interactions of NC with body fluids or cell surfaces that need to be considered for the design of NC vaccines for immunotherapy of cancer.
Collapse
Affiliation(s)
- Matthias Bros
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Johanna Simon
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Lorna Moll
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Volker Mailänder
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|