1
|
Salman DM, Mohammad TAM. siRNA-based therapy for gastric adenocarcinoma: what's next step? Pathol Res Pract 2024; 258:155328. [PMID: 38744002 DOI: 10.1016/j.prp.2024.155328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Gastric cancer continues to have a high death rate despite advancements in their diagnosis and treatment. Novel treatment techniques are thus desperately needed. This is where double-stranded RNA molecules known as small interfering RNA (siRNA), which may selectively target the mRNA of disease-causing genes, may find use in medicine. For siRNAs to function properly in the human body, they must be shielded from deterioration. Furthermore, in order to maintain organ function, they must only target the tumor and spare normal tissue. siRNAs have been designed using clever delivery mechanisms including polymers and lipids to achieve these objectives. Although siRNA protection is not hard to acquire, it is still challenging to target cancer cells with them. Here, we first discuss the basic characteristics of gastric cancer before describing the properties of siRNA and typical delivery methods created specifically for gastric tumors. Lastly, we provide a succinct overview of research using siRNAs to treat gastric tumors.
Collapse
Affiliation(s)
- Dyar Mudhafar Salman
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Pharmacy department, School of Medicine, University of Kurdistan Hewlêr (UKH), Erbil, Kurdistan Region, Iraq.
| |
Collapse
|
2
|
Lu R, Yu P, Sui Y. A computational study of cell membrane damage and intracellular delivery in a cross-slot microchannel. SOFT MATTER 2024; 20:4057-4071. [PMID: 38578041 DOI: 10.1039/d4sm00047a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
We propose a three-dimensional computational framework to simulate the flow-induced cell membrane damage and the resulting enhanced intracellular mass transport in a cross-slot microchannel. We model the cell as a liquid droplet enclosed by a viscoelastic membrane and solve the cell deformation using a well-tested immersed-boundary lattice-Boltzmann method. The cell membrane damage, which is directly related to the membrane permeability, is considered using continuum damage mechanics. The transport of the diffusive solute into the cell is solved by a lattice-Boltzmann model. After validating the computational framework against several benchmark cases, we consider a cell flowing through a cross-slot microchannel, focusing on the effects of the flow strength, channel fluid viscosity and cell membrane viscosity on the membrane damage and enhanced intracellular transport. Interestingly, we find that under a comparable pressure drop across the device, for cells with low membrane viscosity, the inertial flow regime, which can be achieved by driving a low-viscosity liquid at a high speed, often leads to much larger membrane damage, compared with the high-viscosity low-speed viscous flow regime. However, the enhancement can be significantly reduced or even reversed by an increase of the cell membrane viscosity, which limits cell deformation, particularly in the inertial flow regime. Our computational framework and simulation results may guide the design and optimisation of microfluidic devices, which use cross-slot geometry to disrupt cell membranes to enhance intracellular delivery of solutes.
Collapse
Affiliation(s)
- Ruixin Lu
- School of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK.
| | - Peng Yu
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yi Sui
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK.
| |
Collapse
|
3
|
Agiba AM, Arreola-Ramírez JL, Carbajal V, Segura-Medina P. Light-Responsive and Dual-Targeting Liposomes: From Mechanisms to Targeting Strategies. Molecules 2024; 29:636. [PMID: 38338380 PMCID: PMC10856102 DOI: 10.3390/molecules29030636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
In recent years, nanocarriers have played an ever-increasing role in clinical and biomedical applications owing to their unique physicochemical properties and surface functionalities. Lately, much effort has been directed towards the development of smart, stimuli-responsive nanocarriers that are capable of releasing their cargos in response to specific stimuli. These intelligent-responsive nanocarriers can be further surface-functionalized so as to achieve active tumor targeting in a sequential manner, which can be simply modulated by the stimuli. By applying this methodological approach, these intelligent-responsive nanocarriers can be directed to different target-specific organs, tissues, or cells and exhibit on-demand controlled drug release that may enhance therapeutic effectiveness and reduce systemic toxicity. Light, an external stimulus, is one of the most promising triggers for use in nanomedicine to stimulate on-demand drug release from nanocarriers. Light-triggered drug release can be achieved through light irradiation at different wavelengths, either in the UV, visible, or even NIR region, depending on the photophysical properties of the photo-responsive molecule embedded in the nanocarrier system, the structural characteristics, and the material composition of the nanocarrier system. In this review, we highlighted the emerging functional role of light in nanocarriers, with an emphasis on light-responsive liposomes and dual-targeted stimuli-responsive liposomes. Moreover, we provided the most up-to-date photo-triggered targeting strategies and mechanisms of light-triggered drug release from liposomes and NIR-responsive nanocarriers. Lastly, we addressed the current challenges, advances, and future perspectives for the deployment of light-responsive liposomes in targeted drug delivery and therapy.
Collapse
Affiliation(s)
- Ahmed M. Agiba
- Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey 64849, Mexico;
| | - José Luis Arreola-Ramírez
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Mexico City 14080, Mexico; (J.L.A.-R.); (V.C.)
| | - Verónica Carbajal
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Mexico City 14080, Mexico; (J.L.A.-R.); (V.C.)
| | - Patricia Segura-Medina
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Mexico City 14080, Mexico; (J.L.A.-R.); (V.C.)
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Mexico City 14380, Mexico
| |
Collapse
|
4
|
Jagrosse ML, Baliga UK, Jones CW, Russell JJ, García CI, Najar RA, Rahman A, Dean DA, Nilsson BL. Impact of Peptide Sequence on Functional siRNA Delivery and Gene Knockdown with Cyclic Amphipathic Peptide Delivery Agents. Mol Pharm 2023; 20:6090-6103. [PMID: 37963105 DOI: 10.1021/acs.molpharmaceut.3c00455] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Short-interfering RNA (siRNA) oligonucleotide therapeutics that modify gene expression by accessing RNA-interference (RNAi) pathways have great promise for the treatment of a range of disorders; however, their application in clinical settings has been limited by significant challenges in cellular delivery. Herein, we report a structure-function study using a series of modified cyclic amphipathic cell-penetrating peptides (CAPs) to determine the impact of peptide sequence on (1) siRNA-binding efficiency, (2) cellular delivery and knockdown efficiency, and (3) the endocytic uptake mechanism. Nine cyclic peptides of the general sequence Ac-C[XZ]4CG-NH2 in which X residues are hydrophobic/aromatic (Phe, Tyr, Trp, or Leu) and Z residues are charged/hydrophilic (Arg, Lys, Ser, or Glu) are assessed along with one acyclic peptide, Ac-(WR)4G-NH2. Cyclization is enforced by intramolecular disulfide bond formation between the flanking Cys residues. Binding analyses indicate that strong cationic character and the presence of aromatic residues that are competent to participate in CH-π interactions lead to CAP sequences that most effectively interact with siRNA. CAP-siRNA binding increases in the following order as a function of CAP hydrophobic/aromatic content: His < Phe < Tyr < Trp. Both cationic charge and disulfide-constrained cyclization of CAPs improve uptake of siRNA in vitro. Net neutral CAPs and an acyclic peptide demonstrate less-efficient siRNA translocation compared to the cyclic, cationic CAPs tested. All CAPs tested facilitated efficient siRNA target gene knockdown of at least 50% (as effective as a lipofectamine control), with the best CAPs enabling >80% knockdown. Significantly, gene knockdown efficiency does not strongly correlate with CAP-siRNA internalization efficiency but moderately correlates with CAP-siRNA-binding affinity. Finally, utilization of small-molecule inhibitors and targeted knockdown of essential endocytic pathway proteins indicate that most CAP-siRNA nanoparticles facilitate siRNA delivery through clathrin- and caveolin-mediated endocytosis. These results provide insight into the design principles for CAPs to facilitate siRNA delivery and the mechanisms by which these peptides translocate siRNA into cells. These studies also demonstrate the nature of the relationships between peptide-siRNA binding, cellular delivery of siRNA cargo, and functional gene knockdown. Strong correlations between these properties are not always observed, which illustrates the complexity in the design of optimal next-generation materials for oligonucleotide delivery.
Collapse
Affiliation(s)
- Melissa L Jagrosse
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
| | - Uday K Baliga
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| | - Christopher W Jones
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
| | - Jade J Russell
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
| | - Claudia I García
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
| | - Rauf Ahmad Najar
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| | - Arshad Rahman
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| | - David A Dean
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
- Materials Science Program, University of Rochester, Rochester, New York 14627, United States
| |
Collapse
|
5
|
Tracey SR, Smyth P, Herron UM, Burrows JF, Porter AJ, Barelle CJ, Scott CJ. Development of a cationic polyethyleneimine-poly(lactic- co-glycolic acid) nanoparticle system for enhanced intracellular delivery of biologics. RSC Adv 2023; 13:33721-33735. [PMID: 38020041 PMCID: PMC10654694 DOI: 10.1039/d3ra06050k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/05/2023] [Indexed: 12/01/2023] Open
Abstract
Intracellular delivery of proteins, peptides and biologics is an emerging field which has the potential to provide novel opportunities to target intracellular proteins, previously deemed 'undruggable'. However, the delivery of proteins intracellularly remains a challenge. Here, we present a cationic nanoparticle delivery system for enhanced cellular delivery of proteins through use of a polyethyleneimine and poly-(lactic-co-glycolic acid) polymer blend. Cationic nanoparticles were shown to provide increased cellular uptake compared to anionic and neutral nanoparticles, successfully delivering Variable New Antigen Receptors (vNARs), entrapped within the nanoparticle core, to the cell interior. vNARs were identified as ideal candidates for nanoparticle entrapment due to their remarkable stability. The optimised 10% PEI-PLGA nanoparticle formulation displayed low toxicity, was uniform in size and possessed appropriate cationic charge to limit cellular toxicity, whilst being capable of escaping the endo/lysosomal system and delivering their cargo to the cytosol. This work demonstrates the ability of cationic nanoparticles to facilitate intracellular delivery of vNARs, novel biologic agents with potential utility towards intracellular targets.
Collapse
Affiliation(s)
- Shannon R Tracey
- Queen's University Belfast, The Patrick G Johnston Centre for Cancer Research 97 Lisburn Road Belfast Northern Ireland BT9 7AE UK
| | - Peter Smyth
- Queen's University Belfast, The Patrick G Johnston Centre for Cancer Research 97 Lisburn Road Belfast Northern Ireland BT9 7AE UK
| | - Una M Herron
- Queen's University Belfast, The Patrick G Johnston Centre for Cancer Research 97 Lisburn Road Belfast Northern Ireland BT9 7AE UK
| | - James F Burrows
- Queen's University Belfast, School of Pharmacy Belfast BT9 7BL UK
| | | | | | - Christopher J Scott
- Queen's University Belfast, The Patrick G Johnston Centre for Cancer Research 97 Lisburn Road Belfast Northern Ireland BT9 7AE UK
| |
Collapse
|
6
|
Xu M, Li S. Nano-drug delivery system targeting tumor microenvironment: A prospective strategy for melanoma treatment. Cancer Lett 2023; 574:216397. [PMID: 37730105 DOI: 10.1016/j.canlet.2023.216397] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Melanoma, the most aggressive form of cutaneous malignancy arising from melanocytes, is frequently characterized by metastasis. Despite considerable progress in melanoma therapies, patients with advanced-stage disease often have a poor prognosis due to the limited efficacy, off-target effects, and toxicity associated with conventional drugs. Nanotechnology has emerged as a promising approach to address these challenges with nanoparticles capable of delivering therapeutic agents specifically to the tumor microenvironment (TME). However, the clinical approval of nanomedicines for melanoma treatment remains limited, necessitating further research to develop nanoparticles with improved biocompatibility and precise targeting capabilities. This comprehensive review provides an overview of the current research on nano-drug delivery systems for melanoma treatment, focusing on liposomes, polymeric nanoparticles, and inorganic nanoparticles. It discusses the potential of these nanoparticles for targeted drug delivery, as well as their ability to enhance the efficacy of conventional drugs while minimizing toxicity. Furthermore, this review emphasizes the significance of interdisciplinary collaboration between researchers from various fields to advance the development of nanomedicines. Overall, this review serves as a valuable resource for researchers and clinicians interested in the potential of nano-drug delivery systems for melanoma treatment and offers insights into future directions for research in this field.
Collapse
Affiliation(s)
- Mengdan Xu
- Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, China.
| |
Collapse
|
7
|
Agoni C, Stavropoulos I, Kirwan A, Mysior MM, Holton T, Kranjc T, Simpson JC, Roche HM, Shields DC. Cell-Penetrating Milk-Derived Peptides with a Non-Inflammatory Profile. Molecules 2023; 28:6999. [PMID: 37836842 PMCID: PMC10574647 DOI: 10.3390/molecules28196999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Milk-derived peptides are known to confer anti-inflammatory effects. We hypothesised that milk-derived cell-penetrating peptides might modulate inflammation in useful ways. Using computational techniques, we identified and synthesised peptides from the milk protein Alpha-S1-casein that were predicted to be cell-penetrating using a machine learning predictor. We modified the interpretation of the prediction results to consider the effects of histidine. Peptides were then selected for testing to determine their cell penetrability and anti-inflammatory effects using HeLa cells and J774.2 mouse macrophage cell lines. The selected peptides all showed cell penetrating behaviour, as judged using confocal microscopy of fluorescently labelled peptides. None of the peptides had an effect on either the NF-κB transcription factor or TNFα and IL-1β secretion. Thus, the identified milk-derived sequences have the ability to be internalised into the cell without affecting cell homeostatic mechanisms such as NF-κB activation. These peptides are worthy of further investigation for other potential bioactivities or as a naturally derived carrier to promote the cellular internalisation of other active peptides.
Collapse
Affiliation(s)
- Clement Agoni
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- School of Medicine, University College Dublin, Belfield, D04 W6F6 Dublin 4, Ireland
- Discipline of Pharmaceutical Sciences, University of KwaZulu Natal, Durban 4041, South Africa
| | - Ilias Stavropoulos
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- School of Medicine, University College Dublin, Belfield, D04 W6F6 Dublin 4, Ireland
| | - Anna Kirwan
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- School of Biology and Environmental Science, University College Dublin, Belfield, D04 N2E5 Dublin 4, Ireland
| | - Margharitha M. Mysior
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- Institute of Food and Health, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland
| | - Therese Holton
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- Institute of Food and Health, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland
| | - Tilen Kranjc
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- Institute of Food and Health, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland
| | - Jeremy C. Simpson
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- School of Biology and Environmental Science, University College Dublin, Belfield, D04 N2E5 Dublin 4, Ireland
| | - Helen M. Roche
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK
| | - Denis C. Shields
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland (M.M.M.); (J.C.S.)
- School of Medicine, University College Dublin, Belfield, D04 W6F6 Dublin 4, Ireland
| |
Collapse
|
8
|
Choi J, Kim DI, Kim JY, Pané S, Nelson BJ, Chang YT, Choi H. Magnetically Enhanced Intracellular Uptake of Superparamagnetic Iron Oxide Nanoparticles for Antitumor Therapy. ACS NANO 2023; 17:15857-15870. [PMID: 37477428 DOI: 10.1021/acsnano.3c03780] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely employed in biomedical fields, including targeted delivery of antitumor therapy. Conventional magnetic tumor targeting has used simple static magnetic fields (SMFs), which cause SPIONs to linearly aggregate into a long chain-like shape. Such agglomeration greatly hinders the intracellular targeting of SPIONs into tumors, thus reducing the therapeutic efficacy. In this study, we investigated the enhancement of the intracellular uptake of SPIONs through the application of rotating magnetic fields (RMFs). Based on the physical principles of SPION chain disassembly, we investigated physical parameters to predict the chain length favorable for intracellular uptake. Our prediction was validated by clear visualization of the intracellular distributions of SPIONs in tumor cells at both cellular and three-dimensional microtissue levels. To identify the potential therapeutic effects of enhanced intracellular uptake, magnetic hyperthermia as antitumor therapy was investigated under varying conditions of magnetic hyperthermia and RMFs. The results showed that enhanced intracellular uptake reduced magnetic hyperthermia time and strength as well as particle concentration. The proposed method will be useful in the development of techniques to determine the optimized physical conditions for the enhanced intracellular uptake of SPIONs in antitumor therapy.
Collapse
Affiliation(s)
- Junhee Choi
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Dong-In Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jin-Young Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Robotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Division of Biotechnology, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- IMsystem Co., Ltd., Daegu 42988, Republic of Korea
| | - Salvador Pané
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Bradley J Nelson
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Young-Tae Chang
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, Gyeongbuk 37673, Republic of Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hongsoo Choi
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Robotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| |
Collapse
|
9
|
Yihunie W, Nibret G, Aschale Y. Recent Advances in Messenger Ribonucleic Acid (mRNA) Vaccines and Their Delivery Systems: A Review. Clin Pharmacol 2023; 15:77-98. [PMID: 37554660 PMCID: PMC10405914 DOI: 10.2147/cpaa.s418314] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
Messenger ribonucleic acid (mRNA) was found as the intermediary that transfers genetic information from DNA to ribosomes for protein synthesis in 1961. The emergency use authorization of the two covid-19 mRNA vaccines, BNT162b2 and mRNA-1273, is a significant achievement in the history of vaccine development. Because they are generated in a cell-free environment using the in vitro transcription (IVT) process, mRNA vaccines are risk-free. Moreover, chemical modifications to the mRNA molecule, such as cap structures and changed nucleosides, have proved critical in overcoming immunogenicity concerns, achieving sustained stability, and achieving effective, accurate protein production in vivo. Several vaccine delivery strategies (including protamine, lipid nanoparticles (LNPs), polymers, nanoemulsions, and cell-based administration) were also optimized to load and transport RNA into the cytosol. LNPs, which are composed of a cationic or a pH-dependent ionizable lipid layer, a polyethylene glycol (PEG) component, phospholipids, and cholesterol, are the most advanced systems for delivering mRNA vaccines. Moreover, modifications of the four components that make up the LNPs showed to increase vaccine effectiveness and reduce side effects. Furthermore, the introduction of biodegradable lipids improved LNP biocompatibility. Furthermore, mRNA-based therapies are expected to be effective treatments for a variety of refractory conditions, including infectious diseases, metabolic genetic diseases, cancer, cardiovascular and cerebrovascular diseases. Therefore, the present review aims to provide the scientific community with up-to-date information on mRNA vaccines and their delivery systems.
Collapse
Affiliation(s)
- Wubetu Yihunie
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getinet Nibret
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
10
|
Shahiwala A. Physiological determinants and plausible '6R' roadmap for clinical success of nanomedicines. Nanomedicine (Lond) 2023; 18:1207-1222. [PMID: 37650539 DOI: 10.2217/nnm-2023-0114] [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] [Indexed: 09/01/2023] Open
Abstract
Despite the promising features and aggressive research, the success of nanoparticles in clinical trials is minimal. This manuscript discusses the complex biological barriers that impede the journey of nanoparticles to the target site and the approaches used to overcome them. The '6R' framework (right route, right target, right design, right patient, right combination and right technology) is proposed to improve the clinical translation of nanomedicines. Disease-driven approach contrary to the traditional formulation-driven approach is suggested. Data-driven methods can analyze the relationships between various diseases, patient pathophysiology and the physicochemical properties of different nanomedicines, aiding in the precise selection of the most appropriate treatment options. Further research is needed to evaluate and refine these approaches to develop nanomedicines for clinical success.
Collapse
Affiliation(s)
- Aliasgar Shahiwala
- Department of Pharmaceutics, Dubai Pharmacy College for Girls, Muhaisnah 1, Al Mizhar, Dubai, United Arab Emirates
| |
Collapse
|
11
|
Nanomaterial-mediated photoporation for intracellular delivery. Acta Biomater 2023; 157:24-48. [PMID: 36584801 DOI: 10.1016/j.actbio.2022.12.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Translocation of extrinsic molecules into living cells is becoming increasingly crucial in biological studies ranging from cell engineering to biomedical applications. The concerns regarding biosafety and immunogenicity for conventional vectors and physical methods yet challenge effective intracellular delivery. Here, we begin with an overview of approaches for trans-membrane delivery up to now. These methods are featured with a relatively mature application but usually encounter low cell survival. Our review then proposes an advanced application for nanomaterial-sensitized photoporation triggered with a laser. We cover the mechanisms, procedures, and outcomes of photoporation-induced intracellular delivery with a highlight on its versatility to different living cells. We hope the review discussed here encourages researchers to further improvement and applications for photoporation-induced intracellular delivery. STATEMENT OF SIGNIFICANCE.
Collapse
|
12
|
Application of mRNA Technology in Cancer Therapeutics. Vaccines (Basel) 2022; 10:vaccines10081262. [PMID: 36016150 PMCID: PMC9415393 DOI: 10.3390/vaccines10081262] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
mRNA-based therapeutics pose as promising treatment strategies for cancer immunotherapy. Improvements in materials and technology of delivery systems have helped to overcome major obstacles in generating a sufficient immune response required to fight a specific type of cancer. Several in vivo models and early clinical studies have suggested that various mRNA treatment platforms can induce cancer-specific cytolytic activity, leading to numerous clinical trials to determine the optimal method of combinations and sequencing with already established agents in cancer treatment. Nevertheless, further research is required to optimize RNA stabilization, delivery platforms, and improve clinical efficacy by interacting with the tumor microenvironment to induce a long-term antitumor response. This review provides a comprehensive summary of the available evidence on the recent advances and efforts to overcome existing challenges of mRNA-based treatment strategies, and how these efforts play key roles in offering perceptive insights into future considerations for clinical application.
Collapse
|
13
|
Kamalkazemi E, Abedi-Gaballu F, Mohammad Hosseini TF, Mohammadi A, Mansoori B, Dehghan G, Baradaran B, Sheibani N. Glimpse into Cellular Internalization and Intracellular Trafficking of Lipid-Based Nanoparticles in Cancer Cells. Anticancer Agents Med Chem 2021; 22:1897-1912. [PMID: 34488605 DOI: 10.2174/1871520621666210906101421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/14/2021] [Accepted: 06/27/2021] [Indexed: 11/22/2022]
Abstract
Lipid-based nanoparticles as drug delivery carriers have been mainly used for delivery of anti-cancer therapeutic agents. Lipid-based nanoparticles, due to their smaller particle size and similarity to cell membranes, are readily internalized into cancer cells. Interestingly, cancer cells also overexpress receptors for specific ligands including folic acid, hyaluronic acid, and transferrin on their surface. This allows the use of these ligands for surface modification of the lipid-based nanoparticle. These modifications then allow the specific recognition of these ligand-coated nanoparticles by their receptors on cancer cells allowing the targeted gradual intracellular accumulation of the functionalized nanoplatforms. These interactions could eventually enhance the internalization of desired drugs via increasing ligand-receptor mediated cellular uptake of the nanoplatforms. The cellular internalization of the nanoplatforms also varies and depends on their physicochemical properties including particle size, zeta potential, and shape. The cellular uptake is also influenced by the types of ligand internalization pathway utilized by cells such as phagocytosis, macropinocytosis, and multiple endocytosis pathways. In this review, we will classify and discuss lipid based nanoparticles engineered to express specific ligands, and are recognized by their receptors on cancer cell, and their cellular internalization pathways. Moreover, the intracellular fate of nanoparticles decorated with specific ligands and the best internalization pathways (caveolae mediated endocytosis) for safe cargo delivery will be discussed.
Collapse
Affiliation(s)
- Elham Kamalkazemi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | | | | | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI . United States
| |
Collapse
|
14
|
Shoari A, Tooyserkani R, Tahmasebi M, Löwik DWPM. Delivery of Various Cargos into Cancer Cells and Tissues via Cell-Penetrating Peptides: A Review of the Last Decade. Pharmaceutics 2021; 13:1391. [PMID: 34575464 PMCID: PMC8470549 DOI: 10.3390/pharmaceutics13091391] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023] Open
Abstract
Cell-penetrating peptides (CPPs), also known as protein transduction domains, are a class of diverse amino acid sequences with the ability to cross cellular membranes. CPPs can deliver several bioactive cargos, including proteins, peptides, nucleic acids and chemotherapeutics, into cells. Ever since their discovery, synthetic and natural CPPs have been utilized in therapeutics delivery, gene editing and cell imaging in fundamental research and clinical experiments. Over the years, CPPs have gained significant attention due to their low cytotoxicity and high transduction efficacy. In the last decade, multiple investigations demonstrated the potential of CPPs as carriers for the delivery of therapeutics to treat various types of cancer. Besides their remarkable efficacy owing to fast and efficient delivery, a crucial benefit of CPP-based cancer treatments is delivering anticancer agents selectively, rather than mediating toxicities toward normal tissues. To obtain a higher therapeutic index and to improve cell and tissue selectivity, CPP-cargo constructions can also be complexed with other agents such as nanocarriers and liposomes to obtain encouraging outcomes. This review summarizes various types of CPPs conjugated to anticancer cargos. Furthermore, we present a brief history of CPP utilization as delivery systems for anticancer agents in the last decade and evaluate several reports on the applications of CPPs in basic research and preclinical studies.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Raheleh Tooyserkani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Mehdi Tahmasebi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
| | - Dennis W. P. M. Löwik
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| |
Collapse
|
15
|
Christopherson CJ, Paisley NR, Xiao Z, Algar WR, Hudson ZM. Red-Emissive Cell-Penetrating Polymer Dots Exhibiting Thermally Activated Delayed Fluorescence for Cellular Imaging. J Am Chem Soc 2021; 143:13342-13349. [PMID: 34382775 DOI: 10.1021/jacs.1c06290] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fluorescence imaging in living cells is key to understanding many biological processes, yet autofluorescence from the sample can lower sensitivity and hinder high-resolution imaging. Time-gated measurements using phosphorescent metal complexes can improve imaging, at the cost of potential toxicity from the use of heavy metals. Here, we describe orange/red-emitting polymer dots (Pdots) exhibiting thermally activated delayed fluorescence (TADF) for time-gated imaging. Inspired by the cell invasion mechanism of the HIV TAT protein, the Pdots were formed from block copolymers composed of a hydrophilic guanidine-rich block as a cell-penetrating peptide mimic, and a rigid organic semiconductor block to provide efficient delayed fluorescence. These all-organic polymer nanoparticles were shown to efficiently enter HeLa, CHO, and HepG2 cells within 30 min, with cell viabilities remaining high for Pdot concentrations up to 25 mg mL-1. Pdot quantum yields were as high as 0.17 in aerated water, with the Pdot structure effectively shielding the TADF emitters from quenching by oxygen. Colocalization experiments revealed that the Pdots primarily accumulate outside of lysosomes, minimizing lysosomal degradation. When used for fixed cellular imaging, Pdot-incubated cells showed high signal-to-background ratios compared to control samples with no Pdot exposure. Using time-resolved spectroscopy, the delayed emission of the TADF materials was effectively separated from that of both a biological serum and a secondary fluorescent dye.
Collapse
Affiliation(s)
- Cheyenne J Christopherson
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada, V6T 1Z1
| | - Nathan R Paisley
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada, V6T 1Z1
| | - Zhujun Xiao
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada, V6T 1Z1
| | - W Russ Algar
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada, V6T 1Z1
| | - Zachary M Hudson
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, Canada, V6T 1Z1
| |
Collapse
|
16
|
Mimicry of Dopamine 1 Receptor Signaling with Cell-Penetrating Peptides. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-020-10066-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AbstractIn this study, through the use of protein mimicry, a peptide was developed to activate the dopamine 1 receptor signaling pathway from the inside of the cell and in absence of the natural extracellular ligand. The sequence was initially derived from the intracellular interaction site between the activated receptor and the alpha domain of its associated G-protein and subsequently modified to increase its cell-penetrating properties. The peptide was then synthesized via solid phase peptide synthesis, purified and tested on cell models. This novel lipopeptide proved to be capable of efficiently ubiquitously penetrating the cell without the need for transfection agents or chiral recognition by specific pathways. Furthermore, the peptide induced the cellular response normally achieved through the activation of the receptor in cells that had not been treated with the natural ligand. The peptide could work as a candidate substitute to l-DOPA, leading the way for a peptides-based treatment for Parkinson’s disease.
Collapse
|
17
|
Abstract
The provision of safe products from the meat industry has been considered as the major source of protein for maintaining human health. Meat-borne outbreaks are mainly due to Salmonella typhimurium (S. typhimurium), Staphylococcus aureus (S. aureus), Escherichia coli (E. coli), and Clostridium perfringens (C. perfringens), reducing the shelf life and consumer demands. A variety of vulnerable substances, including cholesterol oxidation products (COPs), are generated by the oxidation of meat induced by the microbial infestations. The use of certain biodegradable active packaging, including pullulan active packaging, is being focused by the meat industry due to their safety, stability, and negligible health risks. The potential of pullulan active packaging, incorporated with silver nanoparticles and essential oils, against E. coli, S. typhimurium, Mycoplasma, and other bacterial species is exclusive. Similarly, maintenance of organoleptic properties of meat with nominal oxidative rancidity and limited human health issues can be acquired by pullulan active packaging.
Collapse
|
18
|
Babaee N, Talebkhan Garoosi Y, Karimipoor M, Davami F, Bayat E, Safarpour H, Mahboudi F, Barkhordari F. DARPin Ec1-LMWP protein scaffold in targeted delivery of siRNA molecules through EpCAM cancer stem cell marker. Mol Biol Rep 2020; 47:7323-7331. [PMID: 32979162 DOI: 10.1007/s11033-020-05752-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022]
Abstract
This study is to investigate the binding ability of Designed Ankyrin Repeat Proteins type Ec1that was fused to Low Molecular Weight Protamine (DARPin Ec1-LMWP) protein scaffold to the Epithelial Cell Adhesion Molecule (EpCAM) Cancer Stem Cell (CSC) marker and its efficiency in targeted delivery of small interfering RNA (siRNA) molecules into the studied cells. Gene fragment encoding the DARPIn Ec1-LMWP fusion protein was subcloned into pET28a expression vector following molecular docking studies performed to examine the affinity of the fusion protein towards EpCAM marker. The binding of the siRNA to the expressed fusion protein was tested through native PAGE. The toxicity of the fusion protein was tested by MTT assay. Attachment of the complex to the EpCAM marker was investigated by flow cytometry and delivery of siRNA into the cells was assessed by fluorescence microscopy. The expressed 21.6 kDa DARPin Ec1-LMWP fusion protein was purified and showed no cytotoxicity on tested cells. Arginine rich LMWP was efficiently bounded to the negatively charged siRNA molecule. Successful attachment of the fusion protein:siRNA complex to the EpCAM marker and its internalization into MCF-7 breast cancer cell line were confirmed. Here for the first time the recombinant DARPin Ec1-LMWP protein scaffold was designed and tested for targeting EpCAM surface marker and successful internalization of the siRNA into MCF-7 cells.
Collapse
Affiliation(s)
- Nikta Babaee
- Biotechnology Research Center, Medical Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran.,Biotechnology Research Center, Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Yeganeh Talebkhan Garoosi
- Biotechnology Research Center, Medical Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Morteza Karimipoor
- Biotechnology Research Center, Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Fatemeh Davami
- Biotechnology Research Center, Medical Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Bayat
- Biotechnology Research Center, Medical Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fereidoun Mahboudi
- Biotechnology Research Center, Medical Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Farzaneh Barkhordari
- Biotechnology Research Center, Medical Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
19
|
Wang S, Cao Z, Li M, Yue Y. G-DipC: An Improved Feature Representation Method for Short Sequences to Predict the Type of Cargo in Cell-Penetrating Peptides. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:739-747. [PMID: 31352350 DOI: 10.1109/tcbb.2019.2930993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cell-penetrating peptides (CPPs) are functional short peptides with high carrying capacity. CPP sequences with targeting functions for the highly efficient delivery of drugs to target cells. In this paper, which is focused on the prediction of the cargo category of CPPs, a biocomputational model is constructed to efficiently distinguish the category of cargo carried by CPPs as macromolecular carriers among the seven known deliverable cargo categories. Based on dipeptide composition (DipC), an improved feature representation method, general dipeptide composition (G-DipC) is proposed for short peptide sequences and can effectively increase the abundance of features represented. Then linear discriminant analysis (LDA) is applied to mine some important low-dimensional features of G-DipC and a predictive model is built with the XGBoost algorithm. Experimental results with five-fold cross validation show that G-DipC improves accuracy by 25 and 5 percent compared with amino acid composition (AAC) and DipC, respectively. G-DipC is even found to be better than tripeptide composition (TipC). Thus, the proposed model provides a novel resource for the study of cell-penetrating peptides, and the improved dipeptide composition G-DipC can be widely adapted to determine the feature representation of other biological sequences.
Collapse
|
20
|
Jagrosse ML, Dean DA, Rahman A, Nilsson BL. RNAi therapeutic strategies for acute respiratory distress syndrome. Transl Res 2019; 214:30-49. [PMID: 31401266 PMCID: PMC7316156 DOI: 10.1016/j.trsl.2019.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Acute respiratory distress syndrome (ARDS), replacing the clinical term acute lung injury, involves serious pathophysiological lung changes that arise from a variety of pulmonary and nonpulmonary injuries and currently has no pharmacological therapeutics. RNA interference (RNAi) has the potential to generate therapeutic effects that would increase patient survival rates from this condition. It is the purpose of this review to discuss potential targets in treating ARDS with RNAi strategies, as well as to outline the challenges of oligonucleotide delivery to the lung and tactics to circumvent these delivery barriers.
Collapse
Affiliation(s)
| | - David A Dean
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Arshad Rahman
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, New York.
| |
Collapse
|
21
|
Abstract
The integration of drugs into nanocarriers favorably altered their pharmacodynamics and pharmacokinetics compared to free drugs, and increased their therapeutic index. However, selective cellular internalization in diseased tissues rather than normal tissues still presents a formidable challenge. In this chapter I will cover solutions involving environment-responsive cell-penetrating peptides (CPPs). I will discuss properties of CPPs as universal cellular uptake enhancers, and the modifications imparted to CPP-modified nanocarriers to confine CPP activation to diseased tissues.
Collapse
|
22
|
Biasutto L, Mattarei A, La Spina M, Azzolini M, Parrasia S, Szabò I, Zoratti M. Strategies to target bioactive molecules to subcellular compartments. Focus on natural compounds. Eur J Med Chem 2019; 181:111557. [PMID: 31374419 DOI: 10.1016/j.ejmech.2019.07.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/04/2019] [Accepted: 07/21/2019] [Indexed: 02/06/2023]
Abstract
Many potential pharmacological targets are present in multiple subcellular compartments and have different pathophysiological roles depending on location. In these cases, selective targeting of a drug to the relevant subcellular domain(s) may help to sharpen its impact by providing topological specificity, thus limiting side effects, and to concentrate the compound where needed, thus increasing its effectiveness. We review here the state of the art in precision subcellular delivery. The major approaches confer "homing" properties to the active principle via permanent or reversible (in pro-drug fashion) modifications, or through the use of special-design nanoparticles or liposomes to ferry a drug(s) cargo to its desired destination. An assortment of peptides, substituents with delocalized positive charges, custom-blended lipid mixtures, pH- or enzyme-sensitive groups provide the main tools of the trade. Mitochondria, lysosomes and the cell membrane may be mentioned as the fronts on which the most significant advances have been made. Most of the examples presented here have to do with targeting natural compounds - in particular polyphenols, known as pleiotropic agents - to one or the other subcellular compartment.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121, Padova, Italy; Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy.
| | - Andrea Mattarei
- Dept. Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Martina La Spina
- Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Michele Azzolini
- Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Sofia Parrasia
- Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121, Padova, Italy; Dept. Biology, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121, Padova, Italy; Dept. Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy
| |
Collapse
|
23
|
Rostami B, Irani S, Bolhassani A, Cohan RA. M918: A Novel Cell Penetrating Peptide for Effective Delivery of HIV-1 Nef and Hsp20-Nef Proteins into Eukaryotic Cell Lines. Curr HIV Res 2019; 16:280-287. [PMID: 30520377 PMCID: PMC6416460 DOI: 10.2174/1570162x17666181206111859] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/30/2018] [Accepted: 12/02/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND HIV-1 Nef protein is a possible attractive target in the development of therapeutic HIV vaccines including protein-based vaccines. The most important disadvantage of protein-based vaccines is their low immunogenicity which can be improved by heat shock proteins (Hsps) as an immunomodulator, and cell-penetrating peptides (CPPs) as a carrier. METHODS In this study, the HIV-1 Nef and Hsp20-Nef proteins were generated in E.coli expression system for delivery into the HEK-293T mammalian cell line using a novel cell-penetrating peptide, M918, in a non-covalent fashion. The size, zeta potential and morphology of the peptide/protein complexes were studied by scanning electron microscopy (SEM) and Zeta sizer. The efficiency of Nef and Hsp20-Nef transfection using M918 was evaluated by western blotting in HEK-293T cell line. RESULTS The SEM data confirmed the formation of discrete nanoparticles with a diameter of approximately 200-250 nm and 50-80 nm for M918/Nef and M918/Hsp20-Nef, respectively. The dominant band of ~ 27 kDa and ~ 47 kDa was detected in the transfected cells with the Nef/ M918 and Hsp20-Nef/ M918 nanoparticles at a molar ratio of 1:20 using anti-HIV-1 Nef monoclonal antibody. These bands were not detected in the un-transfected and transfected cells with Nef or Hsp20- Nef protein alone indicating that M918 could increase the penetration of Nef and Hsp20-Nef proteins into the cells. CONCLUSION These data suggest that M918 CPP can be used to enter HIV-1 Nef and Hsp20-Nef proteins inside mammalian cells efficiently as a promising approach in HIV-1 vaccine development.
Collapse
Affiliation(s)
- Bahareh Rostami
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Pilot Nano-Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Kalmouni M, Al-Hosani S, Magzoub M. Cancer targeting peptides. Cell Mol Life Sci 2019; 76:2171-2183. [PMID: 30877335 PMCID: PMC11105397 DOI: 10.1007/s00018-019-03061-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/08/2019] [Accepted: 03/07/2019] [Indexed: 12/19/2022]
Abstract
Despite continuing advances in the development of biomacromolecules for therapeutic purposes, successful application of these often large and hydrophilic molecules has been hindered by their inability to efficiently traverse the cellular plasma membrane. In recent years, cell-penetrating peptides (CPPs) have received considerable attention as a promising class of delivery vectors due to their ability to mediate the efficient import of a large number of cargoes in vitro and in vivo. However, the lack of target specificity of CPPs remains a major obstacle to their clinical development. To address this issue, researchers have developed strategies in which chemotherapeutic drugs are conjugated to cancer targeting peptides (CTPs) that exploit the unique characteristics of the tumor microenvironment or cancer cells, thereby improving cancer cell specificity. This review highlights several of these strategies that are currently in use, and discusses how multi-component nanoparticles conjugated to CTPs can be designed to provide a more efficient cancer therapeutic delivery strategy.
Collapse
Affiliation(s)
- Mona Kalmouni
- Biology Program, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Sumaya Al-Hosani
- Biology Program, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Mazin Magzoub
- Biology Program, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
25
|
Deprey K, Becker L, Kritzer J, Plückthun A. Trapped! A Critical Evaluation of Methods for Measuring Total Cellular Uptake versus Cytosolic Localization. Bioconjug Chem 2019; 30:1006-1027. [PMID: 30882208 PMCID: PMC6527423 DOI: 10.1021/acs.bioconjchem.9b00112] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biomolecules have many properties that make them promising for intracellular therapeutic applications, but delivery remains a key challenge because large biomolecules cannot easily enter the cytosol. Furthermore, quantification of total intracellular versus cytosolic concentrations remains demanding, and the determination of delivery efficiency is thus not straightforward. In this review, we discuss strategies for delivering biomolecules into the cytosol and briefly summarize the mechanisms of uptake for these systems. We then describe commonly used methods to measure total cellular uptake and, more selectively, cytosolic localization, and discuss the major advantages and drawbacks of each method. We critically evaluate methods of measuring "cell penetration" that do not adequately distinguish total cellular uptake and cytosolic localization, which often lead to inaccurate interpretations of a molecule's cytosolic localization. Finally, we summarize the properties and components of each method, including the main caveats of each, to allow for informed decisions about method selection for specific applications. When applied correctly and interpreted carefully, methods for quantifying cytosolic localization offer valuable insight into the bioactivity of biomolecules and potentially the prospects for their eventual development into therapeutics.
Collapse
Affiliation(s)
- Kirsten Deprey
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford, Massachusetts 02155, United States
| | - Lukas Becker
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Joshua Kritzer
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford, Massachusetts 02155, United States
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
26
|
Fornaguera C, Castells-Sala C, Borrós S. Unraveling Polymeric Nanoparticles Cell Uptake Pathways: Two Decades Working to Understand Nanoparticles Journey to Improve Gene Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1288:117-138. [PMID: 31916235 DOI: 10.1007/5584_2019_467] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polymeric nanoparticles have aroused an increasing interest in the last decades as novel advanced delivery systems to improve the treatment of many diseases. Hard work has been performed worldwide designing and developing polymeric nanoparticles using different building blocks, which target specific cell types, trying to avoid bioaccumulation and degradation pathways. The main handicap of the design is to understand the final fate and the journey that the nanoparticle will follow, which is intimately ligated with the chemical and physical properties of the nanoparticles themselves and specific factors of the targeted cells. Although the huge number of published scientific articles regarding polymeric nanoparticles for biomedical applications, their use in clinics is still limited. This fact could be explained by the limited data reporting the interaction of the huge diversity of polymeric nanoparticles with cells. This knowledge is essential to understand nanoparticle uptake and trafficking inside cells to the subcellular target structure.In this chapter, we aim to contribute to this field of knowledge by: (1) summarizing the polymeric nanoparticles properties and cellular factors that influence nanoparticle endocytosis and (2) reviewing the endocytic pathways classified as a function of nanoparticle size and as a function of the receptor playing a role. The revision of previously reported endocytic pathways for particular polymeric nanoparticles could facilitate scientist involved in this field to easily delineate efficient delivery systems based on polymeric nanoparticles.
Collapse
Affiliation(s)
- C Fornaguera
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain.
| | - C Castells-Sala
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
| | - S Borrós
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
| |
Collapse
|
27
|
Givens BE, Naguib YW, Geary SM, Devor EJ, Salem AK. Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Therapeutics. AAPS J 2018; 20:108. [PMID: 30306365 PMCID: PMC6398936 DOI: 10.1208/s12248-018-0267-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
The recent progress in harnessing the efficient and precise method of DNA editing provided by CRISPR/Cas9 is one of the most promising major advances in the field of gene therapy. However, the development of safe and optimally efficient delivery systems for CRISPR/Cas9 elements capable of achieving specific targeting of gene therapy to the location of interest without off-target effects is a primary challenge for clinical therapeutics. Nanoparticles (NPs) provide a promising means to meet such challenges. In this review, we present the most recent advances in developing innovative NP-based delivery systems that efficiently deliver CRISPR/Cas9 constructs and maximize their effectiveness.
Collapse
Affiliation(s)
- Brittany E Givens
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Youssef W Naguib
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA.
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
28
|
Zeng J, Wu J, Li M, Wang P. A Novel Magnetic Nanoparticle for Early Detection of Amyloid Plaques in Alzheimer's Disease. Arch Med Res 2018; 49:282-285. [PMID: 30266531 DOI: 10.1016/j.arcmed.2018.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 09/14/2018] [Indexed: 01/15/2023]
Abstract
Early diagnosis of Alzheimer's disease (AD) remains challenging even with the assistance of imaging. Radiation exposure limits the application of positron emission tomography (PET) for amyloid imaging. Magnetic resonance imaging (MRI) offers superior spatial resolution without the disadvantage of radiation exposure. We developed Mn0.6Zn0.4Fe2O4 (MZF) modified by Pittsburgh compound B (PiB) to specifically bind to amyloid plaques. Coated with amphiphilic nanopolymer 6sPCL-b-P(MEO2MA-co-OEGMA), PiB-MZF was stable, biocompatible, and approximately 100 nm in size. The R2 relaxation rate of PiB-MZF was 169.93 mM-1S-1 demonstrating excellent superparamagnetism as a T2 negative contrast agent. PiB-MZF also showed no cytotoxicity in two cell lines. Immunohistochemistry indicated successful in vitro binding of PiB-MZF with Aβ plaque on 6 months old AD mice brain sections. In conclusion, PiB-MZF nanoparticles are preliminarily proven to have the potential for early detection of amyloid plaques and diagnosis of AD.
Collapse
Affiliation(s)
- Jiaqi Zeng
- Department of Radiology, Tongji Hospital of Tongji University, Shanghai, China
| | - Jiaqi Wu
- Department of Radiology, Tongji Hospital of Tongji University, Shanghai, China
| | - Minghua Li
- Department of Radiology, Tongji Hospital of Tongji University, Shanghai, China
| | - Peijun Wang
- Department of Radiology, Tongji Hospital of Tongji University, Shanghai, China.
| |
Collapse
|
29
|
Wu J, Li J, Wang H, Liu CB. Mitochondrial-targeted penetrating peptide delivery for cancer therapy. Expert Opin Drug Deliv 2018; 15:951-964. [PMID: 30173542 DOI: 10.1080/17425247.2018.1517750] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Mitochondria are promising targeting organelles for anticancer strategies; however, mitochondria are difficult for antineoplastic drugs to recognize and bind. Mitochondria-penetrating peptides (MPPs) are unique tools to gain access to the cell interior and deliver a bioactive cargo into mitochondria. MPPs have combined or delivered a variety of antitumor cargoes and obviously inhibited the tumor growth in vivo and in vitro. MPPs create new opportunities to develop new treatments for cancer. AREAS COVERED We review the target sites of mitochondria and the target-penetration mechanism of MPPs, different strategies, and various additional strategies decorated MPPs for tumor cell mitochondria targeting, the decorating mattes including metabolism molecules, RNA, DNA, and protein, which exploited considered as therapeutic combined with MPPs and target in human cancer treatment. EXPERT OPINION/COMMENTARY Therapeutic selectivity that preferentially targets the mitochondrial abnormalities in cancer cells without toxic impact on normal cells still need to be deepen. Moreover, it needs appropriate study designs for a correct evaluation of the target delivery outcome and the degradation rate of the drug in the cell. Generally, it is optimistic that the advances in mitochondrial targeting drug delivery by MPPs plasticity outlined here will ultimately help to the discovery of new approaches for the prevention and treatment of cancers.
Collapse
Affiliation(s)
- Jiao Wu
- a Affiliated Ren He Hospital of China Three Gorges University , Yichang , China.,b Hubei Key Lab. of Tumor Microenvironment and Immunotherapy , China Three Gorges University , Yichang , China.,c Medical School , China Three Gorges University , Yichang , China
| | - Jason Li
- d Institute for Cell Engineering , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Hu Wang
- b Hubei Key Lab. of Tumor Microenvironment and Immunotherapy , China Three Gorges University , Yichang , China.,c Medical School , China Three Gorges University , Yichang , China.,d Institute for Cell Engineering , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Chang-Bai Liu
- b Hubei Key Lab. of Tumor Microenvironment and Immunotherapy , China Three Gorges University , Yichang , China.,c Medical School , China Three Gorges University , Yichang , China
| |
Collapse
|
30
|
Manavalan B, Subramaniyam S, Shin TH, Kim MO, Lee G. Machine-Learning-Based Prediction of Cell-Penetrating Peptides and Their Uptake Efficiency with Improved Accuracy. J Proteome Res 2018; 17:2715-2726. [PMID: 29893128 DOI: 10.1021/acs.jproteome.8b00148] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cell-penetrating peptides (CPPs) can enter cells as a variety of biologically active conjugates and have various biomedical applications. To offset the cost and effort of designing novel CPPs in laboratories, computational methods are necessitated to identify candidate CPPs before in vitro experimental studies. We developed a two-layer prediction framework called machine-learning-based prediction of cell-penetrating peptides (MLCPPs). The first-layer predicts whether a given peptide is a CPP or non-CPP, whereas the second-layer predicts the uptake efficiency of the predicted CPPs. To construct a two-layer prediction framework, we employed four different machine-learning methods and five different compositions including amino acid composition (AAC), dipeptide composition, amino acid index, composition-transition-distribution, and physicochemical properties (PCPs). In the first layer, hybrid features (combination of AAC and PCP) and extremely randomized tree outperformed state-of-the-art predictors in CPP prediction with an accuracy of 0.896 when tested on independent data sets, whereas in the second layer, hybrid features obtained through feature selection protocol and random forest produced an accuracy of 0.725 that is better than state-of-the-art predictors. We anticipate that our method MLCPP will become a valuable tool for predicting CPPs and their uptake efficiency and might facilitate hypothesis-driven experimental design. The MLCPP server interface along with the benchmarking and independent data sets are freely accessible at www.thegleelab.org/MLCPP .
Collapse
Affiliation(s)
- Balachandran Manavalan
- Department of Physiology , Ajou University School of Medicine , Suwon 443380 , Republic of Korea
| | | | - Tae Hwan Shin
- Department of Physiology , Ajou University School of Medicine , Suwon 443380 , Republic of Korea.,Institute of Molecular Science and Technology , Ajou University , Suwon 443721 , Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 Plus), College of Natural Sciences , Gyeongsang National University , Jinju 52828 , Republic of Korea
| | - Gwang Lee
- Department of Physiology , Ajou University School of Medicine , Suwon 443380 , Republic of Korea.,Institute of Molecular Science and Technology , Ajou University , Suwon 443721 , Republic of Korea
| |
Collapse
|
31
|
Zhao H, To KKW, Chu H, Ding Q, Zhao X, Li C, Shuai H, Yuan S, Zhou J, Kok KH, Jiang S, Yuen KY. Dual-functional peptide with defective interfering genes effectively protects mice against avian and seasonal influenza. Nat Commun 2018; 9:2358. [PMID: 29907765 PMCID: PMC6004018 DOI: 10.1038/s41467-018-04792-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/25/2018] [Indexed: 11/09/2022] Open
Abstract
Limited efficacy of current antivirals and antiviral-resistant mutations impairs anti-influenza treatment. Here, we evaluate the in vitro and in vivo antiviral effect of three defective interfering genes (DIG-3) of influenza virus. Viral replication is significantly reduced in cell lines transfected with DIG-3. Mice treated with DIG-3 encoded by jetPEI-vector, as prophylaxis and therapeutics against A(H7N7) virus, respectively, have significantly better survivals (80% and 50%) than control mice (0%). We further develop a dual-functional peptide TAT-P1, which delivers DIG-3 with high efficiency and concomitantly exerts antiviral activity by preventing endosomal acidification. TAT-P1/DIG-3 is more effective than jetPEI/DIG-3 in treating A(H7N7) or A(H1N1)pdm09-infected mice and shows potent prophylactic protection on A(H7N7) or A(H1N1)pdm09-infected mice. The addition of P1 peptide, which prevents endosomal acidification, can enhance the protection of TAT-P1/DIG-3 on A(H1N1)pdm09-infected mice. Dual-functional TAT-P1 with DIG-3 can effectively protect or treat mice infected by avian and seasonal influenza virus.
Collapse
Affiliation(s)
- Hanjun Zhao
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kelvin K W To
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Qiulu Ding
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xiaoyu Zhao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Cun Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Huiping Shuai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, 200032, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
32
|
Mambelli-Lisboa NC, Sciani JM, Brandão Prieto da Silva AR, Kerkis I. Co-Localization of Crotamine with Internal Membranes and Accentuated Accumulation in Tumor Cells. Molecules 2018; 23:E968. [PMID: 29693555 PMCID: PMC6017820 DOI: 10.3390/molecules23040968] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 01/07/2023] Open
Abstract
Crotamine is a highly cationic; cysteine rich, cross-linked, low molecular mass cell penetrating peptide (CPP) from the venom of the South American rattlesnake. Potential application of crotamine in biomedicine may require its large-scale purification. To overcome difficulties related with the purification of natural crotamine (nCrot) we aimed in the present study to synthesize and characterize a crotamine analog (sCrot) as well investigate its CPP activity. Mass spectrometry analysis demonstrates that sCrot and nCrot have equal molecular mass and biological function—the capacity to induce spastic paralysis in the hind limbs in mice. sCrot CPP activity was evaluated in a wide range of tumor and non-tumor cell tests performed at different time points. We demonstrate that sCrot-Cy3 showed distinct co-localization patterns with intracellular membranes inside the tumor and non-tumor cells. Time-lapse microscopy and quantification of sCrot-Cy3 fluorescence signalss in living tumor versus non-tumor cells revealed a significant statistical difference in the fluorescence intensity observed in tumor cells. These data suggest a possible use of sCrot as a molecular probe for tumor cells, as well as, for the selective delivery of anticancer molecules into these tumors.
Collapse
Affiliation(s)
- Nicole Caroline Mambelli-Lisboa
- Laboratory of Genetics, Butantan Institute, Sao Paulo 05503-900, Brazil.
- CENTD-Center of Excellence in New Target Discovery, Butantan Institute, Sao Paulo 05503-900, Brazil.
| | - Juliana Mozer Sciani
- CENTD-Center of Excellence in New Target Discovery, Butantan Institute, Sao Paulo 05503-900, Brazil.
- Biochemistry and Biophysics Laboratory, Butantan Institute, Sao Paulo 05503-900, Brazil.
| | - Alvaro Rossan Brandão Prieto da Silva
- Laboratory of Genetics, Butantan Institute, Sao Paulo 05503-900, Brazil.
- CENTD-Center of Excellence in New Target Discovery, Butantan Institute, Sao Paulo 05503-900, Brazil.
| | - Irina Kerkis
- Laboratory of Genetics, Butantan Institute, Sao Paulo 05503-900, Brazil.
- CENTD-Center of Excellence in New Target Discovery, Butantan Institute, Sao Paulo 05503-900, Brazil.
| |
Collapse
|
33
|
van Lith SA, van den Brand D, Wallbrecher R, Wübbeke L, van Duijnhoven SM, Mäkinen PI, Hoogstad-van Evert JS, Massuger L, Ylä-Herttuala S, Brock R, Leenders WP. The effect of subcellular localization on the efficiency of EGFR-targeted VHH photosensitizer conjugates. Eur J Pharm Biopharm 2018; 124:63-72. [DOI: 10.1016/j.ejpb.2017.12.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/19/2022]
|
34
|
Mammalian display screening of diverse cystine-dense peptides for difficult to drug targets. Nat Commun 2017; 8:2244. [PMID: 29269835 PMCID: PMC5740061 DOI: 10.1038/s41467-017-02098-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Protein:protein interactions are among the most difficult to treat molecular mechanisms of disease pathology. Cystine-dense peptides have the potential to disrupt such interactions, and are used in drug-like roles by every clade of life, but their study has been hampered by a reputation for being difficult to produce, owing to their complex disulfide connectivity. Here we describe a platform for identifying target-binding cystine-dense peptides using mammalian surface display, capable of interrogating high quality and diverse scaffold libraries with verifiable folding and stability. We demonstrate the platform’s capabilities by identifying a cystine-dense peptide capable of inhibiting the YAP:TEAD interaction at the heart of the oncogenic Hippo pathway, and possessing the potency and stability necessary for consideration as a drug development candidate. This platform provides the opportunity to screen cystine-dense peptides with drug-like qualities against targets that are implicated for the treatment of diseases, but are poorly suited for conventional approaches. Pathologies related to protein:protein interaction are hard to treat but cystine-dense peptides have the potential to disrupt such interactions. Here the authors develop a high-diversity mammalian cell screen for cystine-dense peptides with drug potential and use it to identify a YAP:TEAD inhibitor.
Collapse
|
35
|
Salatin S, Yari Khosroushahi A. Overviews on the cellular uptake mechanism of polysaccharide colloidal nanoparticles. J Cell Mol Med 2017; 21:1668-1686. [PMID: 28244656 PMCID: PMC5571529 DOI: 10.1111/jcmm.13110] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/30/2016] [Indexed: 12/15/2022] Open
Abstract
Nanoparticulate drug/gene carriers have gained much attention in the past decades because of their versatile and tunable properties. However, efficacy of the therapeutic agents can be further enhanced using naturally occurring materials-based nanoparticles. Polysaccharides are an emerging class of biopolymers; therefore, they are generally considered to be safe, non-toxic, biocompatible and biodegradable. Considering that the target of nanoparticle-based therapeutic strategies is localization of biomedical agents in subcellular compartments, a detailed understanding of the cellular mechanism involved in the uptake of polysaccharide-based nanoparticles is essential for safe and efficient therapeutic applications. Uptake of the nanoparticles by the cellular systems occurs with a process known as endocytosis and is influenced by the physicochemical characteristics of nanoparticles such as size, shape and surface chemistry as well as the employed experimental conditions. In this study, we highlight the main endocytosis mechanisms responsible for the cellular uptake of polysaccharide nanoparticles containing drug/gene.
Collapse
Affiliation(s)
- Sara Salatin
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Breger JC, Muttenthaler M, Delehanty JB, Thompson DA, Oh E, Susumu K, Deschamps JR, Anderson GP, Field LD, Walper SA, Dawson PE, Medintz IL. Nanoparticle cellular uptake by dendritic wedge peptides: achieving single peptide facilitated delivery. NANOSCALE 2017; 9:10447-10464. [PMID: 28703833 DOI: 10.1039/c7nr03362a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Significant efforts are being undertaken to optimize the cargo carrying capacity and especially the cellular delivery efficiency of functionalized nanoparticles for applications in biological research and pharmacological delivery. One approach to increasing nanoparticle surface cargo display capacity is to decrease the number of moieties required for mediating cellular delivery by improving their efficiency. We describe a series of multivalent cell penetrating peptide (CPP) dendrimers that facilitate rapid cellular delivery of prototypical nanoparticle-semiconductor quantum dots (QDs). The modular CPP dendrimers were assembled through an innovative convergent oxime ligation strategy between (Arg9)n motifs and a dendritic QD-coordination scaffold. Dendrimeric peptides sequentially incorporate a terminal (His)6 motif for metal-affinity QD coordination, a Pro9 spacer, a branching poly-lysine scaffold, and wedged display of (Arg9)n binding motifs with n = 1×, 2×, 4×, 8×, 16× multivalency. QD dendrimer display capacity was estimated using structural simulations and QD-(Arg9)1-16 conjugates characterized by dynamic light scattering along with surface plasmon resonance-based binding assays to heparan sulfate proteoglycan surfaces. Cellular uptake via endocytosis was confirmed and peptide delivery kinetics investigated as a function of QD-(Arg9)1-16 conjugate exposure time and QD assembly ratio where cellular viability assays reflected no overt cytotoxicity. The ability of single dendrimer conjugates to facilitate cellular uptake was confirmed for QD-(Arg9)2-16 repeats along with the ability to deliver >850 kDa of protein cargo per QD. Minimizing the number of CPPs required for cellular uptake is critical for expanding nanoparticle cargo carrying capacity and can allow for inclusion of additional sensors, therapeutics and contrast agents on their surface.
Collapse
Affiliation(s)
- Joyce C Breger
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA. and American Society for Engineering Education, Washington, DC 20036, USA
| | - Markus Muttenthaler
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA and Institute for Molecular Bioscience, The University of Queensland, 4072 St Lucia, Brisbane, Australia
| | - James B Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - Darren A Thompson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA and University of Idaho, Coeur d'Alene, ID 83814, USA
| | - Eunkeu Oh
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, DC 20375, USA and Sotera Defense Solutions, Inc., Columbia, MD 21046, USA
| | - Kimihiro Susumu
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, DC 20375, USA and Sotera Defense Solutions, Inc., Columbia, MD 21046, USA
| | - Jeffrey R Deschamps
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - George P Anderson
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - Lauren D Field
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA. and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Scott A Walper
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - Philip E Dawson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| |
Collapse
|
37
|
Kerkis I, de Brandão Prieto da Silva AR, Pompeia C, Tytgat J, de Sá Junior PL. Toxin bioportides: exploring toxin biological activity and multifunctionality. Cell Mol Life Sci 2017; 74:647-661. [PMID: 27554773 PMCID: PMC11107510 DOI: 10.1007/s00018-016-2343-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/27/2016] [Accepted: 08/15/2016] [Indexed: 10/21/2022]
Abstract
Toxins have been shown to have many biological functions and to constitute a rich source of drugs and biotechnological tools. We focus on toxins that not only have a specific activity, but also contain residues responsible for transmembrane penetration, which can be considered bioportides-a class of cell-penetrating peptides that are also intrinsically bioactive. Bioportides are potential tools in pharmacology and biotechnology as they help deliver substances and nanoparticles to intracellular targets. Bioportides characterized so far are peptides derived from human proteins, such as cytochrome c (CYCS), calcitonin receptor (camptide), and endothelial nitric oxide synthase (nosangiotide). However, toxins are usually disregarded as potential bioportides. In this review, we discuss the inclusion of some toxins and molecules derived thereof as a new class of bioportides based on structure activity relationship, minimization, and biological activity studies. The comparative analysis of the amino acid residue composition of toxin-derived bioportides and their short molecular variants is an innovative analytical strategy which allows us to understand natural toxin multifunctionality in vivo and plan novel pharmacological and biotechnological products. Furthermore, we discuss how many bioportide toxins have a rigid structure with amphiphilic properties important for both cell penetration and bioactivity.
Collapse
Affiliation(s)
- Irina Kerkis
- Laboratório de Genética, Instituto Butantan, Av. Vital Brasil 1500, São Paulo, SP, 05503-900, Brazil.
| | | | - Celine Pompeia
- Laboratório de Genética, Instituto Butantan, Av. Vital Brasil 1500, São Paulo, SP, 05503-900, Brazil
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Louvain, Belgium
| | - Paulo L de Sá Junior
- Laboratório de Genética, Instituto Butantan, Av. Vital Brasil 1500, São Paulo, SP, 05503-900, Brazil.
| |
Collapse
|
38
|
Kharrat N, Belmabrouk S, Abdelhedi R, Benmarzoug R, Assidi M, Al Qahtani MH, Rebai A. Screening for clusters of charge in human virus proteomes. BMC Genomics 2016; 17:758. [PMID: 27766959 PMCID: PMC5073957 DOI: 10.1186/s12864-016-3086-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background The identification of charge clusters (runs of charged residues) in proteins and their mapping within the protein structure sequence is an important step toward a comprehensive analysis of how these particular motifs mediate, via electrostatic interactions, various molecular processes such as protein sorting, translocation, docking, orientation and binding to DNA and to other proteins. Few algorithms that specifically identify these charge clusters have been designed and described in the literature. In this study, 197 distinctive human viral proteomes were screened for the occurrence of charge clusters (CC) using a new computational approach. Results Three hundred and seventy three CC have been identified within the 2549 viral protein sequences screened. The number of protein sequences that are CC-free is 2176 (85.3 %) while 150 and 180 proteins contained positive charge (PCC) and negative charge clusters (NCC), respectively. The NCCs (211 detected) were more prevalent than PCC (162). PCC-containing proteins are significantly longer than those having NCCs (p = 2.10-16). The most prevalent virus families having PCC and NCC were Herpesviridae followed by Papillomaviridae. However, the single-strand RNA group has in average three times more NCC than PCC. According to the functional domain classification, a significant difference in distribution was observed between PCC and NCC (p = 2. 10−8) with the occurrence of NCCs being more frequent in C-terminal region while PCC more often fall within functional domains. Only 29 proteins sequences contained both NCC and PCC. Moreover, 101 NCC were conserved in 84 proteins while only 62 PCC were conserved in 60 protein sequences. To understand the mechanism by which the membrane translocation functionalities are embedded in viral proteins, we screened our PCC for sequences corresponding to cell-penetrating peptides (CPPs) using two online databases: CellPPd and CPPpred. We found that all our PCCs, having length varying from 7 to 30 amino-acids were predicted as CPPs. Experimental validation is required to improve our understanding of the role of these PCCs in viral infection process. Conclusions Screening distinctive cluster charges in viral proteomes suggested a functional role of these protein regions and might provide potential clues to improve the current understanding of viral diseases in order to tailor better preventive and therapeutic approaches. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3086-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Najla Kharrat
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia.
| | - Sabrine Belmabrouk
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| | - Rania Abdelhedi
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| | - Riadh Benmarzoug
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| | - Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed H Al Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Rebai
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| |
Collapse
|
39
|
Delivery of RNAi Therapeutics to the Airways-From Bench to Bedside. Molecules 2016; 21:molecules21091249. [PMID: 27657028 PMCID: PMC6272875 DOI: 10.3390/molecules21091249] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/05/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
RNA interference (RNAi) is a potent and specific post-transcriptional gene silencing process. Since its discovery, tremendous efforts have been made to translate RNAi technology into therapeutic applications for the treatment of different human diseases including respiratory diseases, by manipulating the expression of disease-associated gene(s). Similar to other nucleic acid-based therapeutics, the major hurdle of RNAi therapy is delivery. Pulmonary delivery is a promising approach of delivering RNAi therapeutics directly to the airways for treating local conditions and minimizing systemic side effects. It is a non-invasive route of administration that is generally well accepted by patients. However, pulmonary drug delivery is a challenge as the lungs pose a series of anatomical, physiological and immunological barriers to drug delivery. Understanding these barriers is essential for the development an effective RNA delivery system. In this review, the different barriers to pulmonary drug delivery are introduced. The potential of RNAi molecules as new class of therapeutics, and the latest preclinical and clinical studies of using RNAi therapeutics in different respiratory conditions are discussed in details. We hope this review can provide some useful insights for moving inhaled RNAi therapeutics from bench to bedside.
Collapse
|
40
|
Shorter SA, Gollings AS, Gorringe-Pattrick MAM, Coakley JE, Dyer PDR, Richardson SCW. The potential of toxin-based drug delivery systems for enhanced nucleic acid therapeutic delivery. Expert Opin Drug Deliv 2016; 14:685-696. [DOI: 10.1080/17425247.2016.1227781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Roma-Rodrigues C, Heuer-Jungemann A, Fernandes AR, Kanaras AG, Baptista PV. Peptide-coated gold nanoparticles for modulation of angiogenesis in vivo. Int J Nanomedicine 2016; 11:2633-9. [PMID: 27354794 PMCID: PMC4907718 DOI: 10.2147/ijn.s108661] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In this work, peptides designed to selectively interact with cellular receptors involved in the regulation of angiogenesis were anchored to oligo-ethylene glycol-capped gold nanoparticles (AuNPs) and used to evaluate the modulation of vascular development using an ex ovo chick chorioallantoic membrane assay. These nanoparticles alter the balance between naturally secreted pro- and antiangiogenic factors, under various biological conditions, without causing toxicity. Exposure of chorioallantoic membranes to AuNP–peptide activators of angiogenesis accelerated the formation of new arterioles when compared to scrambled peptide-coated nanoparticles. On the other hand, antiangiogenic AuNP–peptide conjugates were able to selectively inhibit angiogenesis in vivo. We demonstrated that AuNP vectorization is crucial for enhancing the effect of active peptides. Our data showed for the first time the effective control of activation or inhibition of blood vessel formation in chick embryo via AuNP-based formulations suitable for the selective modulation of angiogenesis, which is of paramount importance in applications where promotion of vascular growth is desirable (eg, wound healing) or ought to be contravened, as in cancer development.
Collapse
Affiliation(s)
- Catarina Roma-Rodrigues
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Amelie Heuer-Jungemann
- Institute for Life Sciences, Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Antonios G Kanaras
- Institute for Life Sciences, Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
42
|
Morshed RA, Muroski ME, Dai Q, Wegscheid ML, Auffinger B, Yu D, Han Y, Zhang L, Wu M, Cheng Y, Lesniak MS. Cell-Penetrating Peptide-Modified Gold Nanoparticles for the Delivery of Doxorubicin to Brain Metastatic Breast Cancer. Mol Pharm 2016; 13:1843-54. [PMID: 27169484 DOI: 10.1021/acs.molpharmaceut.6b00004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As therapies continue to increase the lifespan of patients with breast cancer, the incidence of brain metastases has steadily increased, affecting a significant number of patients with metastatic disease. However, a major barrier toward treating these lesions is the inability of therapeutics to penetrate into the central nervous system and accumulate within intracranial tumor sites. In this study, we designed a cell-penetrating gold nanoparticle platform to increase drug delivery to brain metastatic breast cancer cells. TAT peptide-modified gold nanoparticles carrying doxorubicin led to improved cytotoxicity toward two brain metastatic breast cancer cell lines with a decrease in the IC50 of at least 80% compared to free drug. Intravenous administration of these particles led to extensive accumulation of particles throughout diffuse intracranial metastatic microsatellites with cleaved caspase-3 activity corresponding to tumor foci. Furthermore, intratumoral administration of these particles improved survival in an intracranial MDA-MB-231-Br xenograft mouse model. Our results demonstrate the promising application of gold nanoparticles for improving drug delivery in the context of brain metastatic breast cancer.
Collapse
Affiliation(s)
- Ramin A Morshed
- The Brain Tumor Center, The University of Chicago , Chicago, Illinois United States
| | - Megan E Muroski
- Northwestern University Feinberg School of Medicine , 676 North Saint Clair Street, Suite 2210, Chicago, Illinois 60611, United States
| | - Qing Dai
- Department of Chemistry, Institute of Biophysics Dynamics and Howard Hughes Medical Institute, The University of Chicago , Chicago, Illinois United States
| | - Michelle L Wegscheid
- The Brain Tumor Center, The University of Chicago , Chicago, Illinois United States
| | - Brenda Auffinger
- The Brain Tumor Center, The University of Chicago , Chicago, Illinois United States
| | - Dou Yu
- Northwestern University Feinberg School of Medicine , 676 North Saint Clair Street, Suite 2210, Chicago, Illinois 60611, United States
| | - Yu Han
- Northwestern University Feinberg School of Medicine , 676 North Saint Clair Street, Suite 2210, Chicago, Illinois 60611, United States
| | - Lingjiao Zhang
- The Brain Tumor Center, The University of Chicago , Chicago, Illinois United States
| | - Meijing Wu
- Northwestern University Feinberg School of Medicine , 676 North Saint Clair Street, Suite 2210, Chicago, Illinois 60611, United States
| | - Yu Cheng
- Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine , Shanghai, China
| | - Maciej S Lesniak
- Northwestern University Feinberg School of Medicine , 676 North Saint Clair Street, Suite 2210, Chicago, Illinois 60611, United States
| |
Collapse
|