1
|
Fernández-García R, Fraguas-Sánchez AI. Nanomedicines for Pulmonary Drug Delivery: Overcoming Barriers in the Treatment of Respiratory Infections and Lung Cancer. Pharmaceutics 2024; 16:1584. [PMID: 39771562 PMCID: PMC11677881 DOI: 10.3390/pharmaceutics16121584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The pulmonary route for drug administration has garnered a great deal of attention in therapeutics for treating respiratory disorders. It allows for the delivery of drugs directly to the lungs and, consequently, the maintenance of high concentrations at the action site and a reduction in systemic adverse effects compared to other routes, such as oral or intravenous. Nevertheless, the pulmonary administration of drugs is challenging, as the respiratory system tries to eliminate inhaled particles, being the main responsible mucociliary escalator. Nanomedicines represent a primary strategy to overcome the limitations of this route as they can be engineered to prolong pulmonary retention and avoid their clearance while reducing drug systemic distribution and, consequently, systemic adverse effects. This review analyses the use of pulmonary-administered nanomedicines to treat infectious diseases affecting the respiratory system and lung carcinoma, two pathologies that represent major health threats.
Collapse
Affiliation(s)
| | - Ana I. Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
2
|
Öztürk K, Kaplan M, Çalış S. Effects of nanoparticle size, shape, and zeta potential on drug delivery. Int J Pharm 2024; 666:124799. [PMID: 39369767 DOI: 10.1016/j.ijpharm.2024.124799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Nanotechnology has brought about a significant revolution in drug delivery, and research in this domain is increasingly focusing on understanding the role of nanoparticle (NP) characteristics in drug delivery efficiency. First and foremost, we center our attention on the size of nanoparticles. Studies have indicated that NP size significantly influences factors such as circulation time, targeting capabilities, and cellular uptake. Secondly, we examine the significance of nanoparticle shape. Various studies suggest that NPs of different shapes affect cellular uptake mechanisms and offer potential advantages in directing drug delivery. For instance, cylindrical or needle-like NPs may facilitate better cellular uptake compared to spherical NPs. Lastly, we address the importance of nanoparticle charge. Zeta potential can impact the targeting and cellular uptake of NPs. Positively charged NPs may be better absorbed by negatively charged cells, whereas negatively charged NPs might perform more effectively in positively charged cells. This review provides essential insights into understanding the role of nanoparticles in drug delivery. The properties of nanoparticles, including size, shape, and charge, should be taken into consideration in the rational design of drug delivery systems, as optimizing these characteristics can contribute to more efficient targeting of drugs to the desired tissues. Thus, research into nanoparticle properties will continue to play a crucial role in the future of drug delivery.
Collapse
Affiliation(s)
- Kıvılcım Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Türkiye
| | - Meryem Kaplan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Türkiye; Department of Pharmaceutical Technology, Faculty of Pharmacy, Süleyman Demirel University, 32260 Isparta, Türkiye
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Türkiye.
| |
Collapse
|
3
|
Hazt B, Read DJ, Harlen OG, Poon WCK, O'Connell A, Sarkar A. Mucoadhesion across scales: Towards the design of protein-based adhesives. Adv Colloid Interface Sci 2024; 334:103322. [PMID: 39489118 DOI: 10.1016/j.cis.2024.103322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Mucoadhesion is a special case of bioadhesion in which a material adheres to soft mucosal tissues. This review elucidates our current understanding of mucoadhesion across length, time, and energy scales by focusing on relevant structural features of mucus. We highlight the importance of both covalent and non-covalent interactions that can be tailored to maximize mucoadhesive interactions, particularly concerning proteinaceous mucoadhesives, which have been explored only to a limited extent so far in the literature. In particular, we highlight the importance of thiol groups, hydrophobic moieties, and charged species inherent to proteins as key levers to fine tune mucoadhesive performance. Some aspects of protein surface modification by grafting specific functional groups or coupling with polysaccharides to influence mucoadhesive performance are examined. Insights from this review offer a physicochemical roadmap to inform the development of biocompatible, protein-based mucoadhesive systems that can fulfil dual roles for both adhesion and delivery of actives, enabling the fabrication of advanced biomedical, nutritional and allied soft material technologies.
Collapse
Affiliation(s)
- Bianca Hazt
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, LS2 9JT, UK
| | - Daniel J Read
- School of Mathematics, University of Leeds, LS2 9JT, UK
| | | | - Wilson C K Poon
- School of Physics and Astronomy, University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK
| | - Adam O'Connell
- Polymer Science Platform, Reckitt Benckiser Healthcare (UK) Ltd, Dansom Lane S, Hull, HU8 7DS, UK
| | - Anwesha Sarkar
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
4
|
Jiang AY, Lathwal S, Meng S, Witten J, Beyer E, McMullen P, Hu Y, Manan RS, Raji I, Langer R, Anderson DG. Zwitterionic Polymer-Functionalized Lipid Nanoparticles for the Nebulized Delivery of mRNA. J Am Chem Soc 2024; 146:32567-32574. [PMID: 39535145 DOI: 10.1021/jacs.4c11347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Lipid nanoparticles (LNPs) have great potential to enable inhaled delivery of mRNA to treat pulmonary diseases. However, this potential has been limited by the challenge of nebulizing the LNPs. Nebulization of LNPs can cause LNPs to aggregate and release encapsulated mRNA, limiting their delivery efficacy. To overcome this challenge, LNPs are developed whereby the PEG-lipid is fully replaced with a zwitterionic polymer (ZIP)-lipid conjugate to greatly enhance the nebulizer stability. LNPs formulated with ZIP-lipids (ZIP-LNPs) were stable to nebulization across a wide range of formulation parameters. The optimized ZIP-LNP formulation, containing reduced cholesterol content relative to traditional PEG-lipid LNPs, demonstrated improved inhaled mRNA delivery in both healthy and mucoobstructed mouse lungs. Repeat administration of the optimized ZIP-LNP formulation was well tolerated and did not result in pulmonary inflammation. This study demonstrates the potential of zwitterionic polymer-lipid conjugates for improving the performance of inhaled mRNA-LNP formulations.
Collapse
Affiliation(s)
- Allen Y Jiang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sushil Lathwal
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sabrina Meng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jacob Witten
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Emily Beyer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Patrick McMullen
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yizong Hu
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Rajith S Manan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Idris Raji
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts 02115, United States
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts 02115, United States
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts 02115, United States
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
5
|
Valei ZK, Wamsler K, Parker AJ, Obara TA, Klotz AR, Shendruk TN. Dynamics of polymers in coarse-grained nematic solvents. SOFT MATTER 2024. [PMID: 39545826 PMCID: PMC11612746 DOI: 10.1039/d4sm00968a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Polymers are a primary building block in many biomaterials, often interacting with anisotropic backgrounds. While previous studies have considered polymer dynamics within nematic solvents, rarely are the effects of anisotropic viscosity and polymer elongation differentiated. Here, we study polymers embedded in nematic liquid crystals with isotropic viscosity via numerical simulations to explicitly investigate the effect of nematicity on macromolecular conformation and how conformation alone can produce anisotropic dynamics. We employ a hybrid multi-particle collision dynamics and molecular dynamics technique that captures nematic orientation, thermal fluctuations and hydrodynamic interactions. The coupling of the polymer segments to the director field of the surrounding nematic elongates the polymer, producing anisotropic diffusion even in nematic solvents with isotropic viscosity. For intermediate coupling, the competition between background anisotropy and macromolecular entropy leads to hairpins - sudden kinks along the backbone of the polymer. Experiments of DNA embedded in a solution of rod-like fd viruses qualitatively support the role of hairpins in establishing characteristic conformational features that govern polymer dynamics. Hairpin diffusion along the backbone exponentially slows as coupling increases. Better understanding two-way coupling between polymers and their surroundings could allow the creation of more biomimetic composite materials.
Collapse
Affiliation(s)
- Zahra K Valei
- School of Physics and Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Karolina Wamsler
- School of Physics and Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Alex J Parker
- School of Mathematics, Loughborough University, Leicestershire LE11 3TU, UK
| | - Therese A Obara
- Department of Physics and Astronomy, California State University, Long Beach, Long Beach, California 90840, USA
| | - Alexander R Klotz
- Department of Physics and Astronomy, California State University, Long Beach, Long Beach, California 90840, USA
| | - Tyler N Shendruk
- School of Physics and Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| |
Collapse
|
6
|
Li ZA, Wen KC, Liu JH, Zhang C, Zhang F, Li FQ. Strategies for intravesical drug delivery: From bladder physiological barriers and potential transport mechanisms. Acta Pharm Sin B 2024; 14:4738-4755. [PMID: 39664414 PMCID: PMC11628814 DOI: 10.1016/j.apsb.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 12/13/2024] Open
Abstract
Intravesical drug delivery (IDD), as a noninvasive, local pathway of administration, has great clinical significance for bladder diseases, especially bladder cancer. Despite the many advantages of IDD such as enhanced focal drug exposure and avoidance of systemic adverse drug reactions, the effectiveness of drug delivery is greatly challenged by the physiological barriers of the bladder. In this review, the routes and barriers encountered in IDD are first discussed, and attention is paid to the potential internal/mucosal retention and absorption-transport mechanisms of drugs. On this basis, the avoidance, overcoming and utilization of the "three barriers" is further emphasized, and current design and fabrication strategies for intravesical drug delivery systems (IDDSs) are described mainly from the perspectives of constructing drug reservoirs, enhancing permeability and targeting, with the hope of providing systematic understanding and inspirations for the research of novel IDDSs and their treatment of bladder diseases.
Collapse
Affiliation(s)
- Zheng-an Li
- School of Medicine, Shanghai University, Shanghai 200444, China
- Department of Urology/Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Kai-chao Wen
- School of Medicine, Shanghai University, Shanghai 200444, China
- Department of Urology/Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Ji-heng Liu
- Department of Urology/Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Chuan Zhang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Feng Zhang
- Department of Urology/Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Feng-qian Li
- School of Medicine, Shanghai University, Shanghai 200444, China
- Department of Urology/Pharmaceutics, Shanghai Eighth People's Hospital, Shanghai 200235, China
| |
Collapse
|
7
|
Agles AA, Bourg IC. Structure and Dynamics of Water in Polysaccharide (Alginate) Solutions and Gels Explained by the Core-Shell Model. Biomacromolecules 2024; 25:6403-6415. [PMID: 39228282 PMCID: PMC11480987 DOI: 10.1021/acs.biomac.4c00447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
In both biological and engineered systems, polysaccharides offer a means of establishing structural stiffness without altering the availability of water. Notable examples include the extracellular matrix of prokaryotes and eukaryotes, artificial skin grafts, drug delivery materials, and gels for water harvesting. Proper design and modeling of these systems require detailed understanding of the behavior of water confined in pores narrower than about 1 nm. We use molecular dynamics simulations to investigate the properties of water in solutions and gels of the polysaccharide alginate as a function of the water content and polymer cross-linking. We find that a detailed understanding of the nanoscale dynamics of water in alginate solutions and gels requires consideration of the discrete nature of water. However, we also find that the trends in tortuosity, permeability, dielectric constant, and shear viscosity can be adequately represented using the "core-shell" conceptual model that considers the confined fluid as a continuum.
Collapse
Affiliation(s)
- Avery A. Agles
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Ian C. Bourg
- Department
of Civil and Environmental Engineering, Princeton University, Princeton, New Jersey 08544, United States
- High
Meadows Environmental Institute, Princeton
University, Princeton, New Jersey 08544, United States
| |
Collapse
|
8
|
Bohley M, Leroux J. Gastrointestinal Permeation Enhancers Beyond Sodium Caprate and SNAC - What is Coming Next? ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400843. [PMID: 38884149 PMCID: PMC11434117 DOI: 10.1002/advs.202400843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/13/2024] [Indexed: 06/18/2024]
Abstract
Oral peptide delivery is trending again. Among the possible reasons are the recent approvals of two oral peptide formulations, which represent a huge stride in the field. For the first time, gastrointestinal (GI) permeation enhancers (PEs) are leveraged to overcome the main limitation of oral peptide delivery-low permeability through the intestinal epithelium. Despite some success, the application of current PEs, such as salcaprozate sodium (SNAC), sodium caprylate (C8), and sodium caprate (C10), is generally resulting in relatively low oral bioavailabilities (BAs)-even for carefully selected therapeutics. With several hundred peptide-based drugs presently in the pipeline, there is a huge unmet need for more effective PEs. Aiming to provide useful insights for the development of novel PEs, this review summarizes the biological hurdles to oral peptide delivery with special emphasis on the epithelial barrier. It describes the concepts and action modes of PEs and mentions possible new targets. It further states the benchmark that is set by current PEs, while critically assessing and evaluating emerging PEs regarding translatability, safety, and efficacy. Additionally, examples of novel PEs under preclinical and clinical evaluation and future directions are discussed.
Collapse
Affiliation(s)
- Marilena Bohley
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Jean‐Christophe Leroux
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| |
Collapse
|
9
|
Arapidi GP, Urban AS, Osetrova MS, Shender VO, Butenko IO, Bukato ON, Kuznetsov AA, Saveleva TM, Nos GA, Ivanova OM, Lopukhov LV, Laikov AV, Sharova NI, Nikonova MF, Mitin AN, Martinov AI, Grigorieva TV, Ilina EN, Ivanov VT, Govorun VM. Non-human peptides revealed in blood reflect the composition of intestinal microbiota. BMC Biol 2024; 22:178. [PMID: 39183269 PMCID: PMC11346180 DOI: 10.1186/s12915-024-01975-1] [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: 07/25/2023] [Accepted: 08/07/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The previously underestimated effects of commensal gut microbiota on the human body are increasingly being investigated using omics. The discovery of active molecules of interaction between the microbiota and the host may be an important step towards elucidating the mechanisms of symbiosis. RESULTS Here, we show that in the bloodstream of healthy people, there are over 900 peptides that are fragments of proteins from microorganisms which naturally inhabit human biotopes, including the intestinal microbiota. Absolute quantitation by multiple reaction monitoring has confirmed the presence of bacterial peptides in the blood plasma and serum in the range of approximately 0.1 nM to 1 μM. The abundance of microbiota peptides reaches its maximum about 5 h after a meal. Most of the peptides correlate with the bacterial composition of the small intestine and are likely obtained by hydrolysis of membrane proteins with trypsin, chymotrypsin and pepsin - the main proteases of the gastrointestinal tract. The peptides have physicochemical properties that likely allow them to selectively pass the intestinal mucosal barrier and resist fibrinolysis. CONCLUSIONS The proposed approach to the identification of microbiota peptides in the blood, after additional validation, may be useful for determining the microbiota composition of hard-to-reach intestinal areas and monitoring the permeability of the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Georgij P Arapidi
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation.
| | - Anatoly S Urban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Maria S Osetrova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Victoria O Shender
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Ivan O Butenko
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
- Research Institute for Systems Biology and Medicine, Nauchny Proezd 18, Moscow, 117246, Russian Federation
| | - Olga N Bukato
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Alexandr A Kuznetsov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Tatjana M Saveleva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Grigorii A Nos
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Olga M Ivanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Leonid V Lopukhov
- Kazan Federal University, Kremlyovskaya Str. 18, Kazan, 420008, Russian Federation
| | - Alexander V Laikov
- Kazan Federal University, Kremlyovskaya Str. 18, Kazan, 420008, Russian Federation
| | - Nina I Sharova
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Margarita F Nikonova
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Alexander N Mitin
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Alexander I Martinov
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Tatiana V Grigorieva
- Kazan Federal University, Kremlyovskaya Str. 18, Kazan, 420008, Russian Federation
| | - Elena N Ilina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
- Research Institute for Systems Biology and Medicine, Nauchny Proezd 18, Moscow, 117246, Russian Federation
| | - Vadim T Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Vadim M Govorun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
- Research Institute for Systems Biology and Medicine, Nauchny Proezd 18, Moscow, 117246, Russian Federation
| |
Collapse
|
10
|
Alam MI, Paget T, Moosa NY, Alghurairy H, Elkordy AA. Liposomal Drug Delivery against Helicobacter pylori Using Furazolidone and N-Acetyl Cysteine in Augmented Therapy. Pharmaceutics 2024; 16:1123. [PMID: 39339161 PMCID: PMC11435436 DOI: 10.3390/pharmaceutics16091123] [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: 05/31/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Helicobacter pylori (H. pylori) infection is a significant global health concern, affecting approximately 50% of the world's population and leading to gastric ulcers, gastritis, and gastric cancer. The increase in antibiotic resistance has compromised the efficacy of existing therapeutic regimens, necessitating novel approaches for effective eradication. This study aimed to develop a targeted liposomal drug delivery system incorporating furazolidone and N-acetylcysteine (NAC) to enhance mucopenetration and improve Helicobacter pylori eradication. Liposomes were formulated with furazolidone, NAC, and Pluronic F-127 using a modified reverse-phase evaporation technique. The formulations were categorized based on charge as neutral, negative, and positive and tested for mucopenetration using a modified silicon tube method with coumarin-6 as a fluorescent marker. The encapsulation efficiency and particle size were analyzed using HPLC and an Izon q-nano particle size analyzer. The results indicated that charged liposomes showed a higher encapsulation efficiency than neutral liposomes with Pluronic F-127. Notably, combining furazolidone with 1% NAC achieved complete eradication of H. pylori in 2.5 h, compared to six hours without NAC. The findings of this study suggest that incorporating NAC and Pluronic F-127 into liposomal formulations significantly enhances mucopenetration and antimicrobial efficacy.
Collapse
Affiliation(s)
- Muhammad Irfan Alam
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK
| | - Timothy Paget
- School of Medicine, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK
| | - Najla Yussuf Moosa
- School of Medicine, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK
| | | | - Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK
| |
Collapse
|
11
|
He ZJ, Huang B, Cai LH. Bottlebrush Polyethylene Glycol Nanocarriers Translocate across Human Airway Epithelium via Molecular Architecture-Enhanced Endocytosis. ACS NANO 2024; 18:17586-17599. [PMID: 38932624 PMCID: PMC11238595 DOI: 10.1021/acsnano.4c01983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Pulmonary drug delivery is critical for the treatment of respiratory diseases. However, the human airway surface presents multiple barriers to efficient drug delivery. Here, we report a bottlebrush poly(ethylene glycol) (PEG-BB) nanocarrier that can translocate across all barriers within the human airway surface. Guided by a molecular theory, we design a PEG-BB molecule consisting of a linear backbone densely grafted by many (∼1000) low molecular weight (∼1000 g/mol) polyethylene glycol (PEG) chains; this results in a highly anisotropic, wormlike nanocarrier featuring a contour length of ∼250 nm, a cross-section of ∼20 nm, and a hydrodynamic diameter of ∼40 nm. Using the classic air-liquid-interface culture system to recapitulate essential biological features of the human airway surface, we show that PEG-BB rapidly penetrates through endogenous airway mucus and periciliary brush layer (mesh size of 20-40 nm) to be internalized by cells across the whole epithelium. By quantifying the cellular uptake of polymeric carriers of various molecular architectures and manipulating cell proliferation and endocytosis pathways, we show that the translocation of PEG-BB across the epithelium is driven by bottlebrush architecture-enhanced endocytosis. Our results demonstrate that large, wormlike bottlebrush PEG polymers, if properly designed, can be used as a carrier for pulmonary and mucosal drug delivery.
Collapse
Affiliation(s)
- Zhi-Jian He
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Baiqiang Huang
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Li-Heng Cai
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
12
|
Faria MJ, González-Méijome JM, Real Oliveira MECD, Carracedo G, Lúcio M. Recent advances and strategies for nanocarrier-mediated topical therapy and theranostic for posterior eye disease. Adv Drug Deliv Rev 2024; 210:115321. [PMID: 38679293 DOI: 10.1016/j.addr.2024.115321] [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: 02/16/2024] [Revised: 04/08/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
Posterior eye disorders, such as age-related macular degeneration, diabetic retinopathy, and glaucoma, have a significant impact on human quality of life and are the primary cause of age-related retinal diseases among adults. There is a pressing need for innovative topical approaches to treat posterior eye disorders, as current methods often rely on invasive procedures with inherent risks. Limited success was attained in the realm of topical ophthalmic delivery through non-invasive means. Additionally, there exists a dearth of literature that delves into the potential of this approach for drug delivery and theranostic purposes, or that offers comprehensive design strategies for nanocarrier developers to surmount the significant physiological ocular barriers. This review offers a thorough and up-to-date state-of-the-art overview of 40 studies on therapeutic loaded nanocarriers and theranostic devices that, to the best of our knowledge, represent all successful works that reached posterior eye segments through a topical non-invasive administration. Most importantly, based on the successful literature studies, this review provides a comprehensive summary of the potential design strategies that can be implemented during nanocarrier development to overcome each ocular barrier.
Collapse
Affiliation(s)
- Maria João Faria
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - José M González-Méijome
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal; CEORLab - Clinical and Experimental Optometry Research Lab, Centre of Physics, Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - M Elisabete C D Real Oliveira
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - Gonzalo Carracedo
- Department of Optometry and Vision, Faculty of Optics and Optometry, University Complutense of Madrid, C/Arcos de Jalon 118, Madrid 28037, Spain.
| | - Marlene Lúcio
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal; CBMA - Centre of Molecular and Environmental Biology, Department of Biology, Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| |
Collapse
|
13
|
Zou H, Boboltz A, Cheema Y, Song D, Cahn D, Duncan GA. Synthetic mucus barrier arrays as a nanoparticle formulation screening platform. RSC PHARMACEUTICS 2024; 1:218-226. [PMID: 38899149 PMCID: PMC11185047 DOI: 10.1039/d3pm00057e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/08/2024] [Indexed: 06/21/2024]
Abstract
A mucus gel layer lines the luminal surface of tissues throughout the body to protect them from infectious agents and particulates. As a result, nanoparticle drug delivery systems delivered to these sites may become trapped in mucus and subsequently cleared before they can reach target cells. As such, optimizing the properties of nanoparticle delivery vehicles, such as their surface chemistry and size, is essential to improving their penetration through the mucus barrier. In previous work, we developed a mucin-based hydrogel that has viscoelastic properties like that of native mucus which can be further tailored to mimic specific mucosal tissues and disease states. Using this biomimetic hydrogel system, a 3D-printed array containing synthetic mucus barriers was created that is compatible with a 96-well plate enabling its use as a high-throughput screening platform for nanoparticle drug delivery applications. To validate this system, we evaluated several established design parameters to determine their impact on nanoparticle penetration through synthetic mucus barriers. Consistent with the literature, we found nanoparticles of smaller size and coated with a protective PEG layer more efficiently penetrated through synthetic mucus barriers. In addition, we evaluated a mucolytic (tris(2-carboxyethyl) phosphine, TCEP) for use as a permeation enhancer for mucosal drug delivery. In comparison to N-acetyl cysteine (NAC), we found TCEP significantly improved nanoparticle penetration through a disease-like synthetic mucus barrier. Overall, our results establish a new high-throughput screening approach using synthetic mucus barrier arrays to identify promising nanoparticle formulation strategies for drug delivery to mucosal tissues.
Collapse
Affiliation(s)
- Harry Zou
- Fischell Department of Bioengineering, University of Maryland College Park MD 20742 USA
| | - Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland College Park MD 20742 USA
| | - Yahya Cheema
- Fischell Department of Bioengineering, University of Maryland College Park MD 20742 USA
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland College Park MD 20742 USA
| | - Devorah Cahn
- Fischell Department of Bioengineering, University of Maryland College Park MD 20742 USA
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland College Park MD 20742 USA
| |
Collapse
|
14
|
Jiang AY, Witten J, Raji IO, Eweje F, MacIsaac C, Meng S, Oladimeji FA, Hu Y, Manan RS, Langer R, Anderson DG. Combinatorial development of nebulized mRNA delivery formulations for the lungs. NATURE NANOTECHNOLOGY 2024; 19:364-375. [PMID: 37985700 PMCID: PMC10954414 DOI: 10.1038/s41565-023-01548-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/16/2023] [Indexed: 11/22/2023]
Abstract
Inhaled delivery of mRNA has the potential to treat a wide variety of diseases. However, nebulized mRNA lipid nanoparticles (LNPs) face several unique challenges including stability during nebulization and penetration through both cellular and extracellular barriers. Here we develop a combinatorial approach addressing these barriers. First, we observe that LNP formulations can be stabilized to resist nebulization-induced aggregation by altering the nebulization buffer to increase the LNP charge during nebulization, and by the addition of a branched polymeric excipient. Next, we synthesize a combinatorial library of ionizable, degradable lipids using reductive amination, and evaluate their delivery potential using fully differentiated air-liquid interface cultured primary lung epithelial cells. The final combination of ionizable lipid, charge-stabilized formulation and stability-enhancing excipient yields a significant improvement in lung mRNA delivery over current state-of-the-art LNPs and polymeric nanoparticles.
Collapse
Affiliation(s)
- Allen Y Jiang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob Witten
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Idris O Raji
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
| | - Feyisayo Eweje
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT MD-PhD Program, Boston, MA, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sabrina Meng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Favour A Oladimeji
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yizong Hu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rajith S Manan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
15
|
He ZJ, Huang B, Cai LH. Bottlebrush polyethylene glycol nanocarriers translocate across human airway epithelium via molecular architecture enhanced endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580508. [PMID: 38405944 PMCID: PMC10888750 DOI: 10.1101/2024.02.15.580508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Pulmonary drug delivery is critical to the treatment of respiratory diseases. However, the human airway surface presents multiscale barriers to efficient drug delivery. Here we report a bottlebrush polyethylene glycol (PEG-BB) nanocarrier that can translocate across all barriers within the human airway surface. Guided by the molecular theory, we design a PEG-BB molecule consisting of a linear backbone densely grafted by many (∼1,000) low molecular weight (∼1000 g/mol) PEG chains; this results in a highly anisotropic, wormlike nanocarrier featuring a contour length of ∼250 nm, a cross-section of ∼20 nm, and a hydrodynamic diameter of ∼40 nm. Using the classic air-liquid-interface culture system to recapitulate essential biological features of the human airway surface, we show that PEG-BB rapidly penetrates through endogenous airway mucus and periciliary brush layer (mesh size of 20-40 nm) to be internalized by cells across the whole epithelium. By quantifying the cellular uptake of polymeric carriers of various molecular architectures and manipulating cell proliferation and endocytosis pathways, we show that the translocation of PEG-BB across the epithelium is driven by bottlebrush architecture enhanced endocytosis. Our results demonstrate that large, wormlike bottlebrush PEG polymers, if properly designed, can be used as a novel carrier for pulmonary and mucosal drug delivery. Table of Contents
Collapse
|
16
|
Okmen Altas B, Kalaycioglu GD, Lifshiz-Simon S, Talmon Y, Aydogan N. Tadpole-Like Anisotropic Polymer/Lipid Janus Nanoparticles for Nose-to-Brain Drug Delivery: Importance of Geometry, Elasticity on Mucus-Penetration Ability. Mol Pharm 2024; 21:633-650. [PMID: 38164788 DOI: 10.1021/acs.molpharmaceut.3c00773] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Asymmetric geometry (aspect ratio >1), moderate stiffness (i.e., semielasticity), large surface area, and low mucoadhesion of nanoparticles are the main features to reach the brain by penetrating across the nasal mucosa. Herein, a new application has been presented for the use of multifunctional Janus nanoparticles (JNPs) with controllable geometry and size as a nose-to-brain (N2B) delivery system by changing proportions of Precirol ATO 5 and polycaprolactone compartments and other operating conditions. To bring to light the N2B application of JNPs, the results are presented in comparison with polymer and solid lipid nanoparticles, which are frequently used in the literature regarding their biopharmaceutical aspects: mucoadhesion and permeability through the nasal mucosa. The morphology and geometry of JPs were observed via cryogenic-temperature transmission electron microscopy images, and their particle sizes were verified by dynamic light scattering, atomic force microscopy, and scanning electron microscopy. Although all NPs showed penetration across the mucus barrier, the best increase in penetration was observed with asymmetric and semielastic JNPs, which have low interaction ability with the mucus layer. This study presents a new and promising field of application for a multifunctional system suitable for N2B delivery, potentially benefiting the treatment of brain tumors and other central nervous system diseases.
Collapse
Affiliation(s)
- Burcu Okmen Altas
- Department of Chemical Engineering, Hacettepe University, Beytepe, 06800 Ankara, Turkey
| | | | - Sapir Lifshiz-Simon
- Department of Chemical Engineering, and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yeshayahu Talmon
- Department of Chemical Engineering, and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Nihal Aydogan
- Department of Chemical Engineering, Hacettepe University, Beytepe, 06800 Ankara, Turkey
| |
Collapse
|
17
|
Li T, Han K, Feng G, Guo J, Wan Z, Yang X. Condensation of Soy Protein Peptides Contributes to Sequester Bile Acids and Mitigate LPS-Induced Inflammation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1266-1275. [PMID: 38109330 DOI: 10.1021/acs.jafc.3c06480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Soy protein is widely known to have serum triglyceride (TG) and cholesterol-lowering effects associated with a reduced risk of cardiovascular disease. Recent studies highlighted that the extension region (ER) domain of soy 7S globulin (β-conglycinin) is a key component responsible for the serum TG-lowering effect via modulation of bile acid (BA) homeostasis. Here, we studied the sequestration of BAs by ER peptides during intestinal digestion in vitro and assessed the anti-inflammatory effects of ER peptides using Caco-2/HT29-MTX/RAW264.7 triple-cell cocultures as an intestine cell model. Results show that ER peptides, which share characteristics of intrinsically disordered regions (IDRs), are capable of forming peptide condensates and exhibit the capability to sequester BA-containing colloidal structures during intestinal digestion in vitro. Moreover, BAs enhance the penetration of peptide condensates within the mucus layer, enabling ER peptides to mitigate lipopolysaccharide (LPS)-induced gut inflammation. These results provide a possible explanation for the molecular mechanisms underlying the modulation of BA homeostasis by soybean proteins.
Collapse
Affiliation(s)
- Tanghao Li
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Kaining Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Guangxin Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jian Guo
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Zhili Wan
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
18
|
Chen X, Yang Y, Mai Q, Ye G, Liu Y, Liu J. Pillar arene Se nanozyme therapeutic systems with dual drive power effectively penetrated mucus layer combined therapy acute lung injury. Biomaterials 2024; 304:122384. [PMID: 38016334 DOI: 10.1016/j.biomaterials.2023.122384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/09/2023] [Accepted: 11/01/2023] [Indexed: 11/30/2023]
Abstract
siRNA has demonstrated a promising paradigm for therapy of acute lung injury(ALI). However, the pulmonary mucus layer barrier powerfully hinders the therapeutic efficacy. Herein, we proposed to use dual drive power to enhance the mucus permeation of siRNA by constructing the neutral and targeted selenium nanozymes therapeutic system. The multifunctional selenium nanozymes (CWP-Se@Man) were synthesized by modifying with cationic water-soluble pillar arene (CWP) and mannose (Man). After loading CCR2-siRNA, the CWP-Se@Man reached electroneutrality that co-driven by electroneutrality and targeting, the mucus permeation capacity of CWP-Se@Man enhanced by ∼15 fold, thus effectively penetrate pulmonary mucus layer and deliver CCR2-siRNA into macrophages. Moreover, with optimizing the composition of CWP-Se@Man made of CWP (Slutsky, 2013) [5] or CWP (Ichikado et al., 2012) [6], the therapeutic system CWP (Ichikado et al., 2012) [6]-Se@Man showed better biological activities due to smaller size. In inflamed modes, the CWP-Se@Man nanotherapeutic systems loading CCR2-siRNA not only exerted pronounced anti-inflammatory effect through combining inhibit the chemotactic effect and ROS, but also effectively against ALI after blocking the circulatory effect of ROS and inflammatory cytokines. Therefore, this strategy of dual-driving force penetration mucus renders a unique approach for mediating trans-mucus nucleic acid delivery in lungs, and provide a promising treatment for the acute lung injury therapy.
Collapse
Affiliation(s)
- Xu Chen
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Yonglan Yang
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Qiongmei Mai
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Gang Ye
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Yanan Liu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China.
| | - Jie Liu
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
19
|
Erstling JA, Bag N, Gardinier TC, Kohle FFE, DomNwachukwu N, Butler SD, Kao T, Ma K, Turker MZ, Feuer GB, Lee R, Naguib N, Tallman JF, Malarkey HF, Tsaur L, Moore WL, Chapman DV, Aubert T, Mehta S, Cerione RA, Weiss RS, Baird BA, Wiesner UB. Overcoming Barriers Associated with Oral Delivery of Differently Sized Fluorescent Core-Shell Silica Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305937. [PMID: 37689973 DOI: 10.1002/adma.202305937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/07/2023] [Indexed: 09/11/2023]
Abstract
Oral delivery, while a highly desirable form of nanoparticle-drug administration, is limited by challenges associated with overcoming several biological barriers. Here, the authors study how fluorescent and poly(ethylene glycol)-coated (PEGylated) core-shell silica nanoparticles sized 5 to 50 nm interact with major barriers including intestinal mucus, intestinal epithelium, and stomach acid. From imaging fluorescence correlation spectroscopy studies using quasi-total internal reflection fluorescence microscopy, diffusion of nanoparticles through highly scattering mucus is progressively hindered above a critical hydrodynamic size around 20 nm. By studying Caco-2 cell monolayers mimicking the intestinal epithelia, it is observed that ultrasmall nanoparticles below 10 nm diameter (Cornell prime dots, [C' dots]) show permeabilities correlated with high absorption in humans from primarily enhanced passive passage through tight junctions. Particles above 20 nm diameter exclusively show active transport through cells. After establishing C' dot stability in artificial gastric juice, in vivo oral gavage experiments in mice demonstrate successful passage through the body followed by renal clearance without protein corona formation. Results suggest C' dots as viable candidates for oral administration to patients with a proven pathway towards clinical translation and may generate renewed interest in examining silica as a food additive and its effects on nutrition and health.
Collapse
Affiliation(s)
- Jacob A Erstling
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Nirmalya Bag
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Thomas C Gardinier
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Ferdinand F E Kohle
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Naedum DomNwachukwu
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Scott D Butler
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Teresa Kao
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Kai Ma
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Melik Z Turker
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Grant B Feuer
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Rachel Lee
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Nada Naguib
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - James F Tallman
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Henry F Malarkey
- Department of Applied and Engineering Physics, Cornell University, Ithaca, NY, 14853, USA
| | - Lieihn Tsaur
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - William L Moore
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Dana V Chapman
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Tangi Aubert
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Saurabh Mehta
- Center for Precision Nutrition and Health, Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Richard A Cerione
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Robert S Weiss
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Barbara A Baird
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Ulrich B Wiesner
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
20
|
Hanio S, Möllmert S, Möckel C, Choudhury S, Höpfel AI, Zorn T, Endres S, Schlauersbach J, Scheller L, Keßler C, Scherf-Clavel O, Bellstedt P, Schubert US, Pöppler AC, Heinze KG, Guck J, Meinel L. Bile Is a Selective Elevator for Mucosal Mechanics and Transport. Mol Pharm 2023; 20:6151-6161. [PMID: 37906224 DOI: 10.1021/acs.molpharmaceut.3c00550] [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: 11/02/2023]
Abstract
Mucus mechanically protects the intestinal epithelium and impacts the absorption of drugs, with a largely unknown role for bile. We explored the impacts of bile on mucosal biomechanics and drug transport within mucus. Bile diffused with square-root-of-time kinetics and interplayed with mucus, leading to transient stiffening captured in Brillouin images and a concentration-dependent change from subdiffusive to Brownian-like diffusion kinetics within the mucus demonstrated by differential dynamic microscopy. Bile-interacting drugs, Fluphenazine and Perphenazine, diffused faster through mucus in the presence of bile, while Metoprolol, a drug with no bile interaction, displayed consistent diffusion. Our findings were corroborated by rat studies, where co-dosing of a bile acid sequestrant substantially reduced the bioavailability of Perphenazine but not Metoprolol. We clustered over 50 drugs based on their interactions with bile and mucin. Drugs that interacted with bile also interacted with mucin but not vice versa. This study detailed the dynamics of mucus biomechanics under bile exposure and linked the ability of a drug to interact with bile to its abbility to interact with mucus.
Collapse
Affiliation(s)
- Simon Hanio
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Stephanie Möllmert
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Conrad Möckel
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Susobhan Choudhury
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Andreas I Höpfel
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Theresa Zorn
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Sebastian Endres
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Jonas Schlauersbach
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Lena Scheller
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Christoph Keßler
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Oliver Scherf-Clavel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Peter Bellstedt
- Institute of Organic Chemistry, University of Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Institute for Clinical Chemistry, University of Zürich,Rämistrasse 100, 8091 Zurich, Switzerland
| | - Ulrich S Schubert
- Institute of Organic Chemistry, University of Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), University of Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ann-Christin Pöppler
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Josef-Schneider-Strasse 2, 97080 Wuerzburg, Germany
| |
Collapse
|
21
|
Wang D, Jiang Q, Dong Z, Meng T, Hu F, Wang J, Yuan H. Nanocarriers transport across the gastrointestinal barriers: The contribution to oral bioavailability via blood circulation and lymphatic pathway. Adv Drug Deliv Rev 2023; 203:115130. [PMID: 37913890 DOI: 10.1016/j.addr.2023.115130] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
Oral administration is the preferred route of drug delivery in clinical practice due to its noninvasiveness, safety, convenience, and high patient compliance. The gastrointestinal tract (GIT) plays a crucial role in facilitating the targeted delivery of oral drugs. However, the GIT presents multiple barriers that impede drug absorption, including the gastric barrier in the stomach and the mucus and epithelial barriers in the intestine. In recent decades, nanotechnology has emerged as a promising approach for overcoming these challenges by utilizing nanocarrier-based drug delivery systems such as liposomes, micelles, polymeric nanoparticles, solid lipid nanoparticles, and inorganic nanoparticles. Encapsulating drugs within nanocarriers not only protects them from degradation but also enhances their transport and absorption across the GIT, ultimately improving oral bioavailability. The aim of this review is to elucidate the mechanisms underlying nanocarrier-mediated transportation across the GIT into systemic circulation via both the blood circulation and lymphatic pathway.
Collapse
Affiliation(s)
- Ding Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Zhefan Dong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Jianwei Wang
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China; China Jinhua Institute of Zhejiang University, Jinhua 321299, PR China.
| |
Collapse
|
22
|
Zou H, Boboltz A, Cheema Y, Song D, Duncan GA. Synthetic mucus barrier arrays as a nanoparticle formulation screening platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569212. [PMID: 38076819 PMCID: PMC10705391 DOI: 10.1101/2023.11.29.569212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
A mucus gel layer lines the luminal surface of tissues throughout the body to protect them from infectious agents and particulates. As a result, nanoparticle drug delivery systems delivered to these sites may become trapped in mucus and subsequently cleared before they can reach target cells. As such, optimizing the properties of nanoparticle delivery vehicles, such as their surface chemistry and size, is essential to improving their penetration through the mucus barrier. In previous work, we developed a mucin-based hydrogel that has viscoelastic properties like that of native mucus which can be further tailored to mimic specific mucosal tissues and disease states. Using this biomimetic hydrogel system, a 3D-printed array containing synthetic mucus barriers was created that is compatible with a 96-well plate enabling its use as a high-throughput screening platform for nanoparticle drug delivery applications. To validate this system, we evaluated several established design parameters to determine their impact on nanoparticle penetration through synthetic mucus barriers. Consistent with the literature, we found nanoparticles of smaller size and coated with a protective PEG layer more efficiently penetrated through synthetic mucus barriers. In addition, we evaluated a mucolytic (tris (2-carboxyethyl) phosphine, TCEP) for use as a permeation enhancer for mucosal drug delivery. In comparison to N-acetyl cysteine (NAC), we found TCEP significantly improved nanoparticle penetration through a disease-like synthetic mucus barrier. Overall, our results establish a new high-throughput screening approach using synthetic mucus barrier arrays to identify promising nanoparticle formulation strategies for drug delivery to mucosal tissues.
Collapse
Affiliation(s)
- Harry Zou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Yahya Cheema
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Gregg A. Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
23
|
Donahue R, Sahoo JK, Rudolph S, Chen Y, Kaplan DL. Mucosa-Mimetic Materials for the Study of Intestinal Homeostasis and Disease. Adv Healthc Mater 2023; 12:e2300301. [PMID: 37329337 DOI: 10.1002/adhm.202300301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Mucus is a viscoelastic hydrogel that lines and protects the epithelial surfaces of the body that houses commensal microbiota and functions in host defense against pathogen invasion. As a first-line physical and biochemical barrier, intestinal mucus is involved in immune surveillance and spatial organization of the microbiome, while dysfunction of the gut mucus barrier is implicated in several diseases. Mucus can be collected from a variety of mammalian sources for study, however, established methods are challenging in terms of scale and efficiency, as well as with regard to rheological similarity to native human mucus. Therefore, there is a need for mucus-mimetic hydrogels that more accurately reflect the physical and chemical profile of the in vivo human epithelial environment to enable the investigation of the role of mucus in human disease and interactions with the intestinal microbiome. This review will evaluate the material properties of synthetic mucus mimics to date designed to address the above need, with a focus toward an improved understanding of the biochemical and immunological functions of these biopolymers related to utility for research and therapeutic applications.
Collapse
Affiliation(s)
- Rebecca Donahue
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|
24
|
Bustos NA, Ribbeck K, Wagner CE. The role of mucosal barriers in disease progression and transmission. Adv Drug Deliv Rev 2023; 200:115008. [PMID: 37442240 DOI: 10.1016/j.addr.2023.115008] [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: 08/31/2022] [Revised: 05/22/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Mucus is a biological hydrogel that coats and protects all non-keratinized wet epithelial surfaces. Mucins, the primary structural components of mucus, are critical components of the gel layer that protect against invading pathogens. For communicable diseases, pathogen-mucin interactions contribute to the pathogen's fate and the potential for disease progression in-host, as well as the potential for onward transmission. We begin by reviewing in-host mucus filtering mechanisms, including size filtering and interaction filtering, which regulate the permeability of mucus barriers to all molecules including pathogens. Next, we discuss the role of mucins in communicable diseases at the point of transmission (i.e. how the encapsulation of pathogens in emitted mucosal droplets externally to hosts may modulate pathogen infectivity and viability). Overall, mucosal barriers modulate both host susceptibility as well as the dynamics of population-level disease transmission. The study of mucins and their use in models and experimental systems are therefore crucial for understanding the mechanistic biophysical principles underlying disease transmission and the early stages of host infection.
Collapse
Affiliation(s)
- Nicole A Bustos
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline E Wagner
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Afrin H, Geetha Bai R, Kumar R, Ahmad SS, Agarwal SK, Nurunnabi M. Oral delivery of RNAi for cancer therapy. Cancer Metastasis Rev 2023; 42:699-724. [PMID: 36971908 PMCID: PMC10040933 DOI: 10.1007/s10555-023-10099-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Cancer is a major health concern worldwide and is still in a continuous surge of seeking for effective treatments. Since the discovery of RNAi and their mechanism of action, it has shown promises in targeted therapy for various diseases including cancer. The ability of RNAi to selectively silence the carcinogenic gene makes them ideal as cancer therapeutics. Oral delivery is the ideal route of administration of drug administration because of its patients' compliance and convenience. However, orally administered RNAi, for instance, siRNA, must cross various extracellular and intracellular biological barriers before it reaches the site of action. It is very challenging and important to keep the siRNA stable until they reach to the targeted site. Harsh pH, thick mucus layer, and nuclease enzyme prevent siRNA to diffuse through the intestinal wall and thereby induce a therapeutic effect. After entering the cell, siRNA is subjected to lysosomal degradation. Over the years, various approaches have been taken into consideration to overcome these challenges for oral RNAi delivery. Therefore, understanding the challenges and recent development is crucial to offer a novel and advanced approach for oral RNAi delivery. Herein, we have summarized the delivery strategies for oral delivery RNAi and recent advancement towards the preclinical stages.
Collapse
Affiliation(s)
- Humayra Afrin
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Renu Geetha Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Chair of Biosystems Engineering, Institute of Forestry and Engineering, Estonian University of Life Sciences, Kreutzwaldi 56/1, 51006, Tartu, Estonia
| | - Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Sheikh Shafin Ahmad
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Md Nurunnabi
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA.
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
| |
Collapse
|
26
|
Ma Y, Guo Y, Liu S, Hu Y, Yang C, Cheng G, Xue C, Zuo YY, Sun B. pH-Mediated Mucus Penetration of Zwitterionic Polydopamine-Modified Silica Nanoparticles. NANO LETTERS 2023; 23:7552-7560. [PMID: 37494635 DOI: 10.1021/acs.nanolett.3c02128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Zwitterionic polymers have emerged as promising trans-mucus nanocarriers due to their superior antifouling properties. However, for pH-sensitive zwitterionic polymers, the effect of the pH microenvironment on their trans-mucus fate remains unclear. In this work, we prepared a library of zwitterionic polydopamine-modified silica nanoparticles (SiNPs-PDA) with an isoelectric point of 5.6. Multiple-particle tracking showed that diffusion of SiNPs-PDA in mucus with a pH value of 5.6 was 3 times faster than that in mucus with pH value 3.0 or 7.0. Biophysical analysis found that the trans-mucus behavior of SiNPs-PDA was mediated by hydrophobic and electrostatic interactions and hydrogen bonding between mucin and the particles. Furthermore, the particle distribution in the stomach, intestine, and lung demonstrated the pH-mediated mucus penetration behavior of the SiNPs-PDA. This study reveals the pH-mediated mucus penetration behavior of zwitterionic nanomaterials, which provides rational design strategies for zwitterionic polymers as nanocarriers in various mucus microenvironments.
Collapse
Affiliation(s)
- Yubin Ma
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yiyang Guo
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shan Liu
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Yu Hu
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Cheng Yang
- School of Chemistry, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Gang Cheng
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
27
|
Lucia CMD, Oliveira LA, Dias KA, Pereira SMS, da Conceição AR, Babu PVA. Scientific Evidence for the Beneficial Effects of Dietary Blueberries on Gut Health: A Systematic Review. Mol Nutr Food Res 2023; 67:e2300096. [PMID: 37428472 PMCID: PMC10538750 DOI: 10.1002/mnfr.202300096] [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: 02/18/2023] [Revised: 06/10/2023] [Indexed: 07/11/2023]
Abstract
Emerging evidence indicates the association between an unhealthy gut and chronic diseases. A healthy gut comprises an intact gut epithelium and balanced gut microbes. Diet is one of the critical factors that modulate gut health by positively or negatively affecting the intestinal barrier and gut microbes. Blueberries are an excellent source of health-promoting bioactive components, and this systematic review is conducted to evaluate the effect of dietary blueberries on gut health. A literature search is conducted on PubMed/MEDLINE, Scopus, Web of Science, and Embase databases to review relevant studies published between 2011 and 2022 according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The Systematic Review Center for Laboratory Animal Experimentation Risk of Bias (SYRCLE-RoB) tool is used for methodological quality assessments. Sixteen studies included from four countries are reviewed and the results are synthesized narratively. This data analysis indicates that blueberry supplementation improves gut health by improving intestinal morphology, reducing gut permeability, suppressing oxidative stress, ameliorating gut inflammation, and modulating the composition and function of gut microbes. However, there are significant knowledge gaps in this field. These findings indicate that further studies are needed to establish the beneficial effects of blueberries on gut health.
Collapse
Affiliation(s)
- Ceres Mattos Della Lucia
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Livya Alves Oliveira
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Kelly Aparecida Dias
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
28
|
Lenders V, Koutsoumpou X, Phan P, Soenen SJ, Allegaert K, de Vleeschouwer S, Toelen J, Zhao Z, Manshian BB. Modulation of engineered nanomaterial interactions with organ barriers for enhanced drug transport. Chem Soc Rev 2023; 52:4672-4724. [PMID: 37338993 DOI: 10.1039/d1cs00574j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
The biomedical use of nanoparticles (NPs) has been the focus of intense research for over a decade. As most NPs are explored as carriers to alter the biodistribution, pharmacokinetics and bioavailability of associated drugs, the delivery of these NPs to the tissues of interest remains an important topic. To date, the majority of NP delivery studies have used tumor models as their tool of interest, and the limitations concerning tumor targeting of systemically administered NPs have been well studied. In recent years, the focus has also shifted to other organs, each presenting their own unique delivery challenges to overcome. In this review, we discuss the recent advances in leveraging NPs to overcome four major biological barriers including the lung mucus, the gastrointestinal mucus, the placental barrier, and the blood-brain barrier. We define the specific properties of these biological barriers, discuss the challenges related to NP transport across them, and provide an overview of recent advances in the field. We discuss the strengths and shortcomings of different strategies to facilitate NP transport across the barriers and highlight some key findings that can stimulate further advances in this field.
Collapse
Affiliation(s)
- Vincent Lenders
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Xanthippi Koutsoumpou
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stefaan J Soenen
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, CN Rotterdam, 3015, The Netherlands
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B3000 Leuven, Belgium
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Steven de Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jaan Toelen
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| |
Collapse
|
29
|
Tang Y, Zhang L, Sun R, Luo B, Zhou Y, Zhang Y, Liang Y, Xiao B, Wang C. Pulmonary delivery of mucus-traversing PF127-modified silk fibroin nanoparticles loading with quercetin for lung cancer therapy. Asian J Pharm Sci 2023; 18:100833. [PMID: 37635802 PMCID: PMC10450418 DOI: 10.1016/j.ajps.2023.100833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
The mucosal barrier remains a major barrier in the pulmonary drug delivery system, as mucociliary clearance in the airway accelerates the removal of inhaled nanoparticles (NPs). Herein, we designed and developed the inhalable Pluronic F127-modified silk fibroin NPs loading with quercetin (marked as QR-SF (PF127) NPs), aiming to solve the airway mucus barrier and improve the cancer therapeutic effect of QR. The PF127 coating on the SF NPs could attenuate the interaction between NPs and mucin proteins, thus facilitating the diffusion of SF(PF127) NPs in the mucus layer. The QR-SF (PF127) NPs had particle sizes of approximately 200 nm with negatively charged surfaces and showed constant drug release properties. Fluorescence recovery after photobleaching (FRAP) assay and transepithelial transport test showed that QR-SF (PF127) NPs exhibited superior mucus-penetrating ability in artificial mucus and monolayer Calu-3 cell model. Notably, a large amount of QR-SF (PF127) NPs distributed uniformly in the mice airway section, indicating the good retention of NPs in the respiratory tract. The mice melanoma lung metastasis model was established, and the therapeutic effect of QR-SF (PF127) NPs was significantly improved in vivo. PF127-modified SF NPs may be a promising strategy to attenuate the interaction with mucin proteins and enhance mucus penetration efficiency in the pulmonary drug delivery system.
Collapse
Affiliation(s)
- Yu Tang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Lanfang Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Rui Sun
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Baiyi Luo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yu Zhou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yuqi Liang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Chenhui Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
30
|
Ben-David EA, Habibi M, Haddad E, Sammar M, Angel DL, Dror H, Lahovitski H, Booth AM, Sabbah I. Mechanism of nanoplastics capture by jellyfish mucin and its potential as a sustainable water treatment technology. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 869:161824. [PMID: 36720396 DOI: 10.1016/j.scitotenv.2023.161824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 06/18/2023]
Abstract
The accumulation of nanoplastics (NPs) in the environment has raised concerns about their impact on human health and the biosphere. The main aim of this study is to understand the mechanism that governs the capture of NPs by jellyfish mucus extracted from the jellyfish Aurelia sp. (A.a.) and compare the capture/removal efficiency to that of conventional coagulants and mucus from other organisms. The efficacy of A.a mucus to capture polystyrene and acrylic NPs (∼100 nm) from spiked wastewater treatment plant (WWTP) effluent was evaluated. The mucus effect on capture kinetics and destabilization of NPs of different polymer compositions, sizes and concentrations was quantified by means of fluorescent NPs, dynamic light scattering and zeta potential measurements and visualized by scanning electron microscopy. A dosing of A.a. mucus equivalent to protein concentrations of ∼2-4 mg L-1 led to a rapid change in zeta potential from a baseline of -30 mV to values close to 0 mV, indicating a marked change from a stable to a non-stable dispersion leading to a rapid (<10 min) and significant removal of NPs (60 %-90 %) from a stable suspension. The A.a. mucus outperformed all other mucus types (0-37 %) and coagulants (0 %-32 % for ferric chloride; 23-40 % for poly aluminum chlorohydrate), highlighting the potential for jellyfish mucus to be used as bio-flocculant. The results indicate a mucus-particle interaction consisting of adsorption-bridging and "mesh" filtration. Further insight is provided by carbohydrate composition and protein disruption analysis. Total protein disruption resulted in a complete loss of the A.a. mucus capacity to capture NPs, while the breaking of disulfide bonds and protein unfolding resulted in improved capture capacity. The study demonstrates that natural jellyfish mucin can capture and remove NPs in water and wastewater treatment systems more efficiently than conventional coagulants, highlighting the potential for development of a new type of bio-flocculant.
Collapse
Affiliation(s)
- Eric A Ben-David
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Maryana Habibi
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Elias Haddad
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Marei Sammar
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Dror L Angel
- Department of Maritime Civilizations, and Recanati Institute for Maritime Studies, University of Haifa, Haifa, Israel
| | - Hila Dror
- Department of Maritime Civilizations, and Recanati Institute for Maritime Studies, University of Haifa, Haifa, Israel
| | - Haim Lahovitski
- Department of Maritime Civilizations, and Recanati Institute for Maritime Studies, University of Haifa, Haifa, Israel
| | | | - Isam Sabbah
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel; The Institute of Applied Research, The Galilee Society, Shefa-Amr, Israel.
| |
Collapse
|
31
|
Mishima MDV, Martino HSD, Kolba N, Shah DD, Grancieri M, Dos Santos KMO, Lima JP, Da Silva BP, Gonzalez de Mejia E, Tako E. Effects of Intra-Amniotic Administration of the Hydrolyzed Protein of Chia ( Salvia hispanica L.) and Lacticaseibacillus paracasei on Intestinal Functionality, Morphology, and Bacterial Populations, In Vivo ( Gallus gallus). Nutrients 2023; 15:nu15081831. [PMID: 37111052 PMCID: PMC10144735 DOI: 10.3390/nu15081831] [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: 02/21/2023] [Revised: 03/20/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
As a protein source, chia contains high concentrations of bioactive peptides. Probiotics support a healthy digestive tract and immune system. Our study evaluated the effects of the intra-amniotic administration of the hydrolyzed chia protein and the probiotic Lacticaseibacillus paracasei on intestinal bacterial populations, the intestinal barrier, the inflammatory response, and brush border membrane functionality in ovo (Gallus gallus). Fertile broiler (Gallus gallus) eggs (n = 9/group) were divided into 5 groups: (NI) non-injected; (H2O) 18 MΩ H2O; (CP) 10 mg/mL hydrolyzed chia protein; (CPP) 10 mg/mL hydrolyzed chia protein + 106 colony-forming unit (CFU) L. paracasei; (P) 106 CFU L. paracasei. The intra-amniotic administration was performed on day 17 of incubation. At hatching (day 21), the animals were euthanized, and the duodenum and cecum content were collected. The probiotic downregulated the gene expression of NF-κβ, increased Lactobacillus and E. coli, and reduced Clostridium populations. The hydrolyzed chia protein downregulated the gene expression of TNF-α, increased OCLN, MUC2, and aminopeptidase, reduced Bifidobacterium, and increased Lactobacillus. The three experimental groups improved in terms of intestinal morphology. The current results suggest that the intra-amniotic administration of the hydrolyzed chia protein or a probiotic promoted positive changes in terms of the intestinal inflammation, barrier, and morphology, improving intestinal health.
Collapse
Affiliation(s)
| | - Hércia Stampini Duarte Martino
- Department of Nutrition and Health, Federal University of Viçosa, Av. Purdue, s/n, Campus Universitário, Viçosa 36570-900, MG, Brazil
| | - Nikolai Kolba
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA
| | | | - Mariana Grancieri
- Department of Nutrition and Health, Federal University of Viçosa, Av. Purdue, s/n, Campus Universitário, Viçosa 36570-900, MG, Brazil
| | | | - Janine Passos Lima
- Embrapa Agroindústria de Alimentos, Av. das Américas 29.501, Rio de Janeiro 23020-470, RJ, Brazil
| | - Bárbara Pereira Da Silva
- Department of Nutrition and Health, Federal University of Viçosa, Av. Purdue, s/n, Campus Universitário, Viçosa 36570-900, MG, Brazil
| | - Elvira Gonzalez de Mejia
- Department of Food Science & Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Elad Tako
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
32
|
Spleis H, Sandmeier M, Claus V, Bernkop-Schnürch A. Surface design of nanocarriers: Key to more efficient oral drug delivery systems. Adv Colloid Interface Sci 2023; 313:102848. [PMID: 36780780 DOI: 10.1016/j.cis.2023.102848] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
As nanocarriers (NCs) can improve the solubility of drugs, prevent their degradation by gastrointestinal (GI) enzymes and promote their transport across the mucus gel layer and absorption membrane, the oral bioavailability of these drugs can be substantially enhanced. All these properties of NCs including self-emulsifying drug delivery systems (SEDDS), solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), liposomes, polymeric nanoparticles, inorganic nanoparticles and polymeric micelles depend mainly on their surface chemistry. In particular, interaction with food, digestive enzymes, bile salts and electrolytes, diffusion behaviour across the mucus gel layer and fate on the absorption membrane are determined by their surface. Bioinert surfaces limiting interactions with gastrointestinal fluid and content as well as with mucus, adhesive surfaces providing an intimate contact with the GI mucosa and absorption enhancing surfaces can be designed. Furthermore, charge converting surfaces shifting their zeta potential from negative to positive directly at the absorption membrane and surfaces providing a targeted drug release are advantageous. In addition to these passive surfaces, even active surfaces cleaving mucus glycoproteins on their way through the mucus gel layer can be created. Within this review, we provide an overview on these different surfaces and discuss their impact on the performance of NCs in the GI tract.
Collapse
Affiliation(s)
- Helen Spleis
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Matthias Sandmeier
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Victor Claus
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria.
| |
Collapse
|
33
|
Wagner CE, Krupkin M, Smith-Dupont KB, Wu CM, Bustos NA, Witten J, Ribbeck K. Comparison of Physicochemical Properties of Native Mucus and Reconstituted Mucin Gels. Biomacromolecules 2023; 24:628-639. [PMID: 36727870 DOI: 10.1021/acs.biomac.2c01016] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Simulating native mucus with model systems such as gels made from reconstituted mucin or commercially available polymers presents experimental advantages including greater sample availability and reduced inter- and intradonor heterogeneity. Understanding whether these gels reproduce the complex physical and biochemical properties of native mucus at multiple length scales is critical to building relevant experimental models, but few systematic comparisons have been reported. Here, we compared bulk mechanical properties, microstructure, and biochemical responses of mucus from different niches, reconstituted mucin gels (with similar pH and polymer concentrations as native tissues), and commonly used commercially available polymers. To evaluate gel properties across these length scales, we used small-amplitude oscillatory shear, single-particle tracking, and microaffinity chromatography with small analytes. With the exception of human saliva, the mechanical response of mucin gels was qualitatively similar to that of native mucus. The transport behavior of charged peptides through native mucus gels was qualitatively reproduced in gels composed of corresponding isolated mucins. Compared to native mucus, we observed substantial differences in the physicochemical properties of gels reconstituted from commercially available mucins and the substitute carboxymethylcellulose, which is currently used in artificial tear and saliva treatments. Our study highlights the importance of selecting a mucus model system guided by the length scale relevant to the scientific investigation or disease application.
Collapse
Affiliation(s)
- Caroline E Wagner
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Miri Krupkin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Kathryn B Smith-Dupont
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Chloe M Wu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Nicole A Bustos
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Jacob Witten
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States.,Computational and Systems Biology Initiative, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| |
Collapse
|
34
|
Mucus adhesion vs. mucus penetration? Screening nanomaterials for nasal inhalation by MD simulation. J Control Release 2023; 353:366-379. [PMID: 36462640 DOI: 10.1016/j.jconrel.2022.11.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Nanocarrier-aided drug delivery techniques have improved the absorption and permeability of drugs in nose-to-brain delivery. However, the molecular properties of nanocarriers during the delivery process are of great interest; in particular, the characteristics when penetrating barriers in vivo are crucial for the screening and optimization of materials for nasal inhalation. In this study, we have focused on two types of delivery systems: mucoadhesive nanoparticles (MAPs) and mucopenetrating nanoparticles (MPPs); both have been widely used for mucosal delivery, although a method for selecting the more effective type of drug carriers for mucosal delivery has not been established. Molecular dynamics (MD) simulations were used to reveal the all-atom dynamic characteristics of the interaction between different delivery systems and the nasal mucus protein MUC5AC. Among the systems tested, hydroxypropyltrimethyl ammonium chloride chitosan (HTCC) had the strongest interaction with mucin, suggesting it had better mucoadhesive performance, and that it interacted with MUC5AC more strongly than unmodified chitosan. In contrast, the mucus-penetrating material polyethylene glycol-poly lactic acid-co-glycolic acid (PEG-PLGA), had almost no interaction with MUC5AC. The results of the MD simulations were verified by in vitro experiments on nanoparticles (NPs) and mucin binding. The drug delivery performance of the four types of NPs, analyzed by in vitro and ex vivo mucosal penetration, were all generally consistent with the properties of the material predicted from the MD simulation. These clues to the molecular mechanism of MAPs and MPPs may provide useful insight into the screening and optimization of nanomaterials suitable for nasal inhalation.
Collapse
|
35
|
Gustafsson JK, Johansson MEV. The role of goblet cells and mucus in intestinal homeostasis. Nat Rev Gastroenterol Hepatol 2022; 19:785-803. [PMID: 36097076 DOI: 10.1038/s41575-022-00675-x] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/08/2022]
Abstract
The intestinal tract faces numerous challenges that require several layers of defence. The tight epithelium forms a physical barrier that is further protected by a mucus layer, which provides various site-specific protective functions. Mucus is produced by goblet cells, and as a result of single-cell RNA sequencing identifying novel goblet cell subpopulations, our understanding of their various contributions to intestinal homeostasis has improved. Goblet cells not only produce mucus but also are intimately linked to the immune system. Mucus and goblet cell development is tightly regulated during early life and synchronized with microbial colonization. Dysregulation of the developing mucus systems and goblet cells has been associated with infectious and inflammatory conditions and predisposition to chronic disease later in life. Dysfunctional mucus and altered goblet cell profiles are associated with inflammatory conditions in which some mucus system impairments precede inflammation, indicating a role in pathogenesis. In this Review, we present an overview of the current understanding of the role of goblet cells and the mucus layer in maintaining intestinal health during steady-state and how alterations to these systems contribute to inflammatory and infectious disease.
Collapse
Affiliation(s)
- Jenny K Gustafsson
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemisty and Cell biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
36
|
Agrizzi Verediano T, Agarwal N, Stampini Duarte Martino H, Kolba N, Grancieri M, Dias Paes MC, Tako E. Effect of Black Corn Anthocyanin-Rich Extract ( Zea mays L.) on Cecal Microbial Populations In Vivo ( Gallus gallus). Nutrients 2022; 14:4679. [PMID: 36364942 PMCID: PMC9655515 DOI: 10.3390/nu14214679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 08/17/2023] Open
Abstract
Black corn has been attracting attention to investigate its biological properties due to its anthocyanin composition, mainly cyanidin-3-glucoside. Our study evaluated the effects of black corn extract (BCE) on intestinal morphology, gene expression, and the cecal microbiome. The BCE intra-amniotic administration was evaluated by an animal model in Gallus gallus. The eggs (n = 8 per group) were divided into: (1) no injection; (2) 18 MΩ H2O; (3) 5% black corn extract (BCE); and (4) 0.38% cyanidin-3-glucoside (C3G). A total of 1 mL of each component was injected intra-amniotic on day 17 of incubation. On day 21, the animals were euthanized after hatching, and the duodenum and cecum content were collected. The cecal microbiome changes were attributed to BCE administration, increasing the population of Bifidobacterium and Clostridium, and decreasing E. coli. The BCE did not change the gene expression of intestinal inflammation and functionality. The BCE administration maintained the villi height, Paneth cell number, and goblet cell diameter (in the villi and crypt), similar to the H2O injection but smaller than the C3G. Moreover, a positive correlation was observed between Bifidobacterium, Clostridium, E. coli, and villi GC diameter. The BCE promoted positive changes in the cecum microbiome and maintained intestinal morphology and functionality.
Collapse
Affiliation(s)
- Thaisa Agrizzi Verediano
- Nutrition and Health Department, Universidade Federal de Viçosa, Vicosa 36571-000, Minas Gerais, Brazil
| | - Nikita Agarwal
- Department of Food Science, Cornell University, Stocking Hall, Ithaca, NY 14853, USA
| | | | - Nikolai Kolba
- Department of Food Science, Cornell University, Stocking Hall, Ithaca, NY 14853, USA
| | - Mariana Grancieri
- Nutrition and Health Department, Universidade Federal de Viçosa, Vicosa 36571-000, Minas Gerais, Brazil
| | - Maria Cristina Dias Paes
- Empresa Brasileira de Pesquisa e Agropecuária (EMBRAPA), Sete Lagoas 35701-970, Minas Gerais, Brazil
| | - Elad Tako
- Department of Food Science, Cornell University, Stocking Hall, Ithaca, NY 14853, USA
| |
Collapse
|
37
|
Magnetically Driven Muco-Inert Janus Nanovehicles for Enhanced Mucus Penetration and Cellular Uptake. Molecules 2022; 27:molecules27217291. [DOI: 10.3390/molecules27217291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
One of the main challenges of transmucosal drug delivery is that of enabling particles and molecules to move across the mucosal barrier of the mucosal epithelial surface. Inspired by nanovehicles and mucus-penetrating nanoparticles, a magnetically driven, mucus-inert Janus-type nanovehicle (Janus-MMSN-pCB) was fabricated by coating the zwitterionic polymer poly(carboxybetaine methacrylate) (pCB) on the mesoporous silica nanorod, which was grown on one side of superparamagnetic Fe3O4 nanoparticle using the sol–gel method. X-ray diffraction, transmission electron microscopy, vibrating sample magnetometry, and Fourier infrared spectroscopy were used to characterize the structure and morphology of the nanovehicles, proving the success of each synthesis step. The in vitro cell viability assessment of these composites using Calu-3 cell lines indicates that the nanovehicles are biocompatible in nature. Furthermore, the multiparticle tracking, Transwell® system, and cell imaging experimental results demonstrate that both the modification of pCB and the application of a magnetic field effectively accelerated the diffusion of the nanovehicles in the mucus and improved the endocytosis through Calu-3. The favorable cell uptake performance of Janus-MMSN-pCB in mucus systems with/without magnetic driving proves its potential role in the diagnosis, treatment, and imaging of mucosal-related diseases.
Collapse
|
38
|
Zhao K, Xie Y, Lin X, Xu W. The Mucoadhesive Nanoparticle-Based Delivery System in the Development of Mucosal Vaccines. Int J Nanomedicine 2022; 17:4579-4598. [PMID: 36199476 PMCID: PMC9527817 DOI: 10.2147/ijn.s359118] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Mucosal tissue constitutes the largest interface between the body and the external environment, regulating the entry of pathogens, particles, and molecules. Mucosal immunization is the most effective way to trigger a protective mucosal immune response. However, the majority of the currently licensed vaccines are recommended to be administered by intramuscular injection, which has obvious shortcomings, such as high production costs, low patient compliance, and lack of mucosal immune response. Strategies for eliciting mucosal and systemic immune responses are being developed, including appropriate vaccine adjuvant, delivery system, and bacterial or viral vectors. Biodegradable mucoadhesive nanoparticles (NPs) are the most promising candidate for vaccine delivery systems due to their inherent immune adjuvant property and the ability to protect the antigen from degradation, sustain the release of loaded antigen, and increase the residence time of antigen at the administration site. The current review outlined the complex structure of mucosa, the mechanism of interaction between NPs and mucosa, factors affecting the mucoadhesion of NPs, and the application of the delivery system based on mucoadhesive NPs in the field of vaccines. Moreover, this review demonstrated that the biodegradable and mucoadhesive NP-based delivery system has the potential for mucosal administration of vaccines.
Collapse
Affiliation(s)
- Kai Zhao
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
- Correspondence: Kai Zhao, Tel +86 576 88660338, Email
| | - Yinzhuo Xie
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
| | - Xuezheng Lin
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
- Xuezheng Lin, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China, Email
| | - Wei Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, People’s Republic of China
| |
Collapse
|
39
|
Singh A, Yadagiri G, Javaid A, Sharma KK, Verma A, Singh OP, Sundar S, Mudavath SL. Hijacking the intrinsic vitamin B 12 pathway for the oral delivery of nanoparticles, resulting in enhanced in vivo anti-leishmanial activity. Biomater Sci 2022; 10:5669-5688. [PMID: 36017751 DOI: 10.1039/d2bm00979j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Surface-functionalized vitamin B12 (VB12) biocompatible nanoparticles exploit the well-characterized uptake pathway of VB12, shielding it from enzymatic degradation and inadequate absorption. In this perspective, subsequent to escalated mucus interaction and diffusion analysis, the nanoparticles were investigated by immunostaining with the anti-CD320 antibody, and their internalization mechanisms were examined by selectively blocking specific uptake processes. It was observed that their internalization occurred via an energy-dependent clathrin-mediated mechanism, simultaneously highlighting their remarkable ability to bypass the P-glycoprotein efflux. In particular, the synthesized nanoparticles were evaluated for their cytocompatibility by analyzing cellular proliferation, membrane viscoelasticity, and fluidity by fluorescence recovery after photobleaching and oxidative-stress detection, making them well-suited for successful translation to a clinical setup. Our previous in vitro antileishmanial results were paramount for their further in vivo and toxicity analysis, demonstrating their targeted therapeutic efficiency. The augmented surface hydrophilicity, which is attributed to VB12, and monomerization of amphotericin B in the lipid core strengthened the oral bioavailability and stability of the nanoparticles, as evidenced by the fluorescence resonance energy transfer analysis.
Collapse
Affiliation(s)
- Aakriti Singh
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India.
| | - Ganesh Yadagiri
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India.
| | - Aaqib Javaid
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India.
| | - Krishna Kumar Sharma
- Department of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Anurag Verma
- Department of Pharmacy, Teerthanker Mahaveer University, Moradabad, Uttar Pradesh 244001, India
| | - Om Prakash Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Shyam Sundar
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Shyam Lal Mudavath
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India.
| |
Collapse
|
40
|
Feng G, Han K, Yang Q, Feng W, Guo J, Wang J, Yang X. Interaction of Pyrogallol-Containing Polyphenols with Mucin Reinforces Intestinal Mucus Barrier Properties. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9536-9546. [PMID: 35852590 DOI: 10.1021/acs.jafc.2c03564] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
High consumption of polyphenol-rich green tea, coffee, fruits, and vegetables is associated with a low risk of human chronic diseases. Recent studies highlight the relevance of polyphenol-mediated gut microbiota modulation and its impact on mucus barrier. Herein, we study the direct interaction of epicatechin (EC), epigallocatechin gallate (EGCG), and tannic acid (TA) with intestinal mucin by isothermal titration calorimetry and multiple particle tracking and the impact on mucus barrier using ex vivo mucus and Caco-2/HT29-MTX cocultures. Results show that pyrogallol-containing polyphenols EGCG and TA exhibit strong binding to intestinal mucin and reinforce mucus barrier, whereas EC does not. ECGG and TA also mitigate gliadin-mediated cytotoxicity and inflammation. The chemical binding of EGCG and TA to the nucleophilic thiol groups of mucins shows their roles as cross-linkers of mucin networks. These results bring a novel understanding of the health benefits of polyphenols and provide support for the consumption of pyrogallol-containing beverages like green tea as a potential dietary therapy for gluten-related disorders.
Collapse
Affiliation(s)
- Guangxin Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Kaining Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qian Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Weiting Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jian Guo
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jinmei Wang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
41
|
Acupuncture for Female Infertility: Discussion on Action Mechanism and Application. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3854117. [PMID: 35832528 PMCID: PMC9273356 DOI: 10.1155/2022/3854117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/11/2022] [Indexed: 11/18/2022]
Abstract
A higher incidence of female infertility has been reported with an unexpectedly early appearance in recent years. The female infertility treatment and application of assisted reproductive technology have recently gained immense interest from scientists. Many studies have discussed the beneficial effects of acupuncture on female infertility. With advancements in science and medical technology, acupuncture-related research has increased in investigating its effectiveness in treating female infertility. This review focuses on a compilation of research in recent years on acupuncture for female infertility treatment and the exploration of the underlying mechanism. For this purpose, literature was searched using various search engines like PubMed, Web of Science, and Google Scholar. The search was refined by only focusing on recent studies on acupuncture effectiveness and mechanism in female infertility and evaluating pregnancy outcomes.
Collapse
|
42
|
Black corn (Zea mays L.) soluble extract showed anti-inflammatory effects and improved the intestinal barrier integrity in vivo (Gallus gallus). Food Res Int 2022; 157:111227. [DOI: 10.1016/j.foodres.2022.111227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/21/2022]
|
43
|
Oral delivery of polyester nanoparticles for brain-targeting: Challenges and opportunities. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
44
|
Pednekar DD, Liguori MA, Marques CNH, Zhang T, Zhang N, Zhou Z, Amoako K, Gu H. From Static to Dynamic: A Review on the Role of Mucus Heterogeneity in Particle and Microbial Transport. ACS Biomater Sci Eng 2022; 8:2825-2848. [PMID: 35696291 DOI: 10.1021/acsbiomaterials.2c00182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mucus layers (McLs) are on the front line of the human defense system that protect us from foreign abiotic/biotic particles (e.g., airborne virus SARS-CoV-2) and lubricates our organs. Recently, the impact of McLs on human health (e.g., nutrient absorption and drug delivery) and diseases (e.g., infections and cancers) has been studied extensively, yet their mechanisms are still not fully understood due to their high variety among organs and individuals. We characterize these variances as the heterogeneity of McLs, which lies in the thickness, composition, and physiology, making the systematic research on the roles of McLs in human health and diseases very challenging. To advance mucosal organoids and develop effective drug delivery systems, a comprehensive understanding of McLs' heterogeneity and how it impacts mucus physiology is urgently needed. When the role of airway mucus in the penetration and transmission of coronavirus (CoV) is considered, this understanding may also enable a better explanation and prediction of the CoV's behavior. Hence, in this Review, we summarize the variances of McLs among organs, health conditions, and experimental settings as well as recent advances in experimental measurements, data analysis, and model development for simulations.
Collapse
Affiliation(s)
- Dipesh Dinanath Pednekar
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| | - Madison A Liguori
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| | | | - Teng Zhang
- Department of Mechanical and Aerospace Engineering, Syracuse University, Syracuse, New York 13244, United States.,BioInspired Syracuse, Syracuse University, Syracuse, New York 13244, United States
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zejian Zhou
- Department of Electrical and Computer Engineering and Computer Science, University of New Haven, West Haven, Connecticut 06516, United States
| | - Kagya Amoako
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| | - Huan Gu
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| |
Collapse
|
45
|
Kaler L, Joyner K, Duncan GA. Machine learning-informed predictions of nanoparticle mobility and fate in the mucus barrier. APL Bioeng 2022; 6:026103. [PMID: 35757278 PMCID: PMC9217165 DOI: 10.1063/5.0091025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/30/2022] [Indexed: 11/14/2022] Open
Abstract
Nanomaterial diffusion through mucus is important to basic and applied areas of research such as drug delivery. However, it is often challenging to interpret nanoparticle dynamics within the mucus gel due to its heterogeneous microstructure and biochemistry. In this study, we measured the diffusion of polyethylene glycolylated nanoparticles (NPs) in human airway mucus ex vivo using multiple particle tracking and utilized machine learning to classify diffusive vs sub-diffusive NP movement. Using mathematic models that account for the mode of NP diffusion, we calculate the percentage of NPs that would cross the mucus barrier over time in airway mucus with varied total solids concentration. From this analysis, we predict rapidly diffusing NPs will cross the mucus barrier in a physiological timespan. Although less efficient, sub-diffusive "hopping" motion, a characteristic of a continuous time random walk, may also enable NPs to cross the mucus barrier. However, NPs exhibiting fractional Brownian sub-diffusion would be rapidly removed from the airways via mucociliary clearance. In samples with increased solids concentration (>5% w/v), we predict up to threefold reductions in the number of nanoparticles capable of crossing the mucus barrier. We also apply this approach to explore diffusion and to predict the fate of influenza A virus within human mucus. We predict only a small fraction of influenza virions will cross the mucus barrier presumably due to physical obstruction and adhesive interactions with mucin-associated glycans. These results provide new tools to evaluate the extent of synthetic and viral nanoparticle penetration through mucus in the lung and other tissues.
Collapse
Affiliation(s)
- Logan Kaler
- Biophysics Program, University of Maryland, College Park, Maryland 20742, USA
| | - Katherine Joyner
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | | |
Collapse
|
46
|
Fröhlich E. Non-Cellular Layers of the Respiratory Tract: Protection against Pathogens and Target for Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14050992. [PMID: 35631578 PMCID: PMC9143813 DOI: 10.3390/pharmaceutics14050992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 12/10/2022] Open
Abstract
Epithelial barriers separate the human body from the environment to maintain homeostasis. Compared to the skin and gastrointestinal tract, the respiratory barrier is the thinnest and least protective. The properties of the epithelial cells (height, number of layers, intercellular junctions) and non-cellular layers, mucus in the conducting airways and surfactant in the respiratory parts determine the permeability of the barrier. The review focuses on the non-cellular layers and describes the architecture of the mucus and surfactant followed by interaction with gases and pathogens. While the penetration of gases into the respiratory tract is mainly determined by their hydrophobicity, pathogens use different mechanisms to invade the respiratory tract. Often, the combination of mucus adhesion and subsequent permeation of the mucus mesh is used. Similar mechanisms are also employed to improve drug delivery across the respiratory barrier. Depending on the payload and target region, various mucus-targeting delivery systems have been developed. It appears that the mucus-targeting strategy has to be selected according to the planned application.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria; ; Tel.: +43-316-38573011
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
47
|
Cheng YE, Wu IE, Chen YC, Chu IM. Thermo-Sensitive mPEG-PA-PLL Hydrogel for Drug Release of Calcitonin. Gels 2022; 8:282. [PMID: 35621580 PMCID: PMC9141101 DOI: 10.3390/gels8050282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
The oral route is the most popular way of drug administration because of good patient compliance and ease of use. However, the oral delivery of peptides and proteins is difficult, mainly due to poor oral bioavailability. In past decades, researchers have developed several strategies to improve oral bioavailability by avoiding losing activity in the gastrointestinal (GI) tract and enhancing the intestinal permeability of these drugs. Methoxy poly(ethylene glycol)-poly(l-alanine) (mPEG-PA) is a thermo-sensitive hydrogel exhibiting a sol-to-gel phase transition property. This characteristic is appropriate for encapsulating peptide or protein drugs. To enhance the adhesion ability to intestinal mucus, a thermo-sensitive polymer, mPEG-PA, modified with charged amino acid lysine was developed. This positively charged material would help to bind the negatively charged mucin in mucus. The synthesis was conducted by individually synthesizing mPEG-PA and poly(l-lysine) (PLL) of different lengths via ring-opening polymerization. Then, mPEG-PA and PLL were combined using an NHS ester reaction to synthesize the triblock copolymer (mPEG-PA-PLL). Biocompatibility and the release of calcitonin from the synthesized hydrogel particles under different pH were examined. The initial data showed that the newly design material had a promising potential for the oral delivery of peptide drugs.
Collapse
Affiliation(s)
| | | | | | - I-Ming Chu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan; (Y.-E.C.); (I.-E.W.); (Y.-C.C.)
| |
Collapse
|
48
|
Add Sugar to Chitosan: Mucoadhesion and In Vitro Intestinal Permeability of Mannosylated Chitosan Nanocarriers. Pharmaceutics 2022; 14:pharmaceutics14040830. [PMID: 35456664 PMCID: PMC9024478 DOI: 10.3390/pharmaceutics14040830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
Crosslinked chitosan nanocarriers (140–160 nm) entrapping coumarin-6 (λex/em = 455/508 nm) with or without surface mannosylation were synthesized and assessed for cytotoxicity, adherence and cellular uptake in Caco-2 cells, flux across Caco-2 monolayers, and mucoadhesion to porcine mucin. Mannosylated and non-mannosylated nanocarriers demonstrated biocompatibility with slow release of coumarin-6 at pH 6.8 and 7.4 over 24 h. Adherence of the non-mannosylated nanocarriers (50 and 150 µg/mL) to Caco-2 cells was ~10% over 24 h, whereas cellular uptake of 25–30% was noted at 4 h. The mannosylated nanocarriers showed a similar adherence to non-mannosylated nanocarriers after 24 h, but a lower cellular uptake (~20%) at 1 h, comparable uptake at 4 h, and a higher uptake (~25–30%) at 24 h. Overall, the nanocarriers did not affect the integrity of Caco-2 monolayers. Mannosylated nanocarriers elicited higher Papp of 1.6 × 10−6 cm/s (50 µg/mL) and 1.2 × 10−6 (150 µg/mL) than the non-mannosylated ones: 9.8 × 10−7 cm/s (50 µg/mL) and 1.0 × 10−6 (150 µg/mL) after 2 h. Non-mannosylated chitosan nanocarriers elicited enhanced adhesion to porcine gut mucin via mucin-filled microchannels due to higher cationic charge density. These results underpin the importance of surface chemistry in the biological interactions of nanocarriers, while highlighting the role of surface hydrophilicity in mucopermeation due to mannosylation.
Collapse
|
49
|
Watchorn J, Clasky AJ, Prakash G, Johnston IAE, Chen PZ, Gu FX. Untangling Mucosal Drug Delivery: Engineering, Designing, and Testing Nanoparticles to Overcome the Mucus Barrier. ACS Biomater Sci Eng 2022; 8:1396-1426. [PMID: 35294187 DOI: 10.1021/acsbiomaterials.2c00047] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mucus is a complex viscoelastic gel and acts as a barrier covering much of the soft tissue in the human body. High vascularization and accessibility have motivated drug delivery to various mucosal surfaces; however, these benefits are hindered by the mucus layer. To overcome the mucus barrier, many nanomedicines have been developed, with the goal of improving the efficacy and bioavailability of drug payloads. Two major nanoparticle-based strategies have emerged to facilitate mucosal drug delivery, namely, mucoadhesion and mucopenetration. Generally, mucoadhesive nanoparticles promote interactions with mucus for immobilization and sustained drug release, whereas mucopenetrating nanoparticles diffuse through the mucus and enhance drug uptake. The choice of strategy depends on many factors pertaining to the structural and compositional characteristics of the target mucus and mucosa. While there have been promising results in preclinical studies, mucus-nanoparticle interactions remain poorly understood, thus limiting effective clinical translation. This article reviews nanomedicines designed with mucoadhesive or mucopenetrating properties for mucosal delivery, explores the influence of site-dependent physiological variation among mucosal surfaces on efficacy, transport, and bioavailability, and discusses the techniques and models used to investigate mucus-nanoparticle interactions. The effects of non-homeostatic perturbations on protein corona formation, mucus composition, and nanoparticle performance are discussed in the context of mucosal delivery. The complexity of the mucosal barrier necessitates consideration of the interplay between nanoparticle design, tissue-specific differences in mucus structure and composition, and homeostatic or disease-related changes to the mucus barrier to develop effective nanomedicines for mucosal delivery.
Collapse
Affiliation(s)
- Jeffrey Watchorn
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Gayatri Prakash
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Ian A E Johnston
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Paul Z Chen
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Frank X Gu
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
50
|
Bhattacharjee S. Molecular Descriptors as a Facile Tool toward Designing Surface-Functionalized Nanoparticles for Drug Delivery. Mol Pharm 2022; 19:1168-1175. [PMID: 35316069 PMCID: PMC8985240 DOI: 10.1021/acs.molpharmaceut.1c00940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/20/2022] [Accepted: 03/08/2022] [Indexed: 11/29/2022]
Abstract
Modulating the surface chemistry of nanoparticles, often by grafting hydrophilic polymer brushes (e.g., polyethylene glycol) to prepare nanoformulations that can resist opsonization in a hematic environment and negotiate with the mucus barrier, is a popular strategy toward developing biocompatible and effective nano-drug delivery systems. However, there is a need for tools that can screen multiple surface ligands and cluster them based on both structural similarity and physicochemical attributes. Molecular descriptors offer numerical readouts based on molecular properties and provide a fertile ground for developing quick screening platforms. Thus, a study was conducted with 14 monomers/repeating blocks of polymeric chains, namely, oxazoline, acrylamide, vinylpyrrolidone, glycerol, acryloyl morpholine, dimethyl acrylamide, hydroxypropyl methacrylamide, hydroxyethyl methacrylamide, sialic acid, carboxybetaine acrylamide, carboxybetaine methacrylate, sulfobetaine methacrylate, methacryloyloxyethyl phosphorylcholine, and vinyl-pyridinio propanesulfonate, capable of imparting hydrophilicity to a surface when assembled as polymeric brushes. Employing free, Web-based, and user-friendly platforms, such as SwissADME and ChemMine tools, a series of molecular descriptors and Tanimoto coefficient of molecular pairs were determined, followed by hierarchical clustering analyses. Molecular pairs of oxazoline/dimethyl acrylamide, hydroxypropyl methacrylamide/hydroxyethyl methacrylamide, acrylamide/glycerol, carboxybetaine acrylamide/vinyl-pyridinio propanesulfonate, and sulfobetaine methacrylate/methacryloyloxyethyl phosphorylcholine were clustered together. Similarly, the molecular pair of hydroxypropyl methacrylamide/hydroxyethyl methacrylamide demonstrated a high Tanimoto coefficient of >0.9, whereas the pairs oxazoline/vinylpyrrolidone, acrylamide/dimethyl acrylamide, acryloyl morpholine/dimethyl acrylamide, acryloyl morpholine/hydroxypropyl methacrylamide, acryloyl morpholine/hydroxyethyl methacrylamide, carboxybetaine methacrylate/sulfobetaine methacrylate, and glycerol/hydroxypropyl methacrylamide had a Tanimoto coefficient of >0.8. The analyzed data not only demonstrated the ability of such in silico tools as a facile technique in clustering molecules of interest based on their structure and physicochemical characteristics but also provided vital information on their behavior within biological systems, including the ability to engage an array of possible molecular targets when the monomers are self-assembled on nanoparticulate surfaces.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin 4, Ireland
| |
Collapse
|