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
|
Faurschou KL, Clasky AJ, Watchorn J, Tram Su J, Li NT, McGuigan AP, Gu FX. Lateral Assessment of Mucomimetic Hydrogels to Evaluate Correlation between Microscopic and Macroscopic Properties. Macromol Biosci 2024; 24:e2400146. [PMID: 39374341 DOI: 10.1002/mabi.202400146] [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: 03/27/2024] [Revised: 08/06/2024] [Indexed: 10/09/2024]
Abstract
A major limitation in the development of mucosal drug delivery systems is the design of in vitro models that accurately reflect in vivo conditions. Traditionally, models seek to mimic characteristics of physiological mucus, often focusing on property-specific trial metrics such as rheological behavior or diffusion of a nanoparticle of interest. Despite the success of these models, translation from in vitro results to in vivo trials is limited. As a result, several authors have called for work to develop standardized testing methodologies and characterize the influence of model properties on drug delivery performance. To this end, a series of trials is performed on 12 mucomimetic hydrogels reproduced from literature. Experiments show that there is no consistent correlation between barrier performance and rheological or microstructural properties of the tested mucomimetic hydrogels. In addition, the permeability of both mucopenetrating and mucoadhesive nanoparticles is assessed, revealing non-obvious variations in barrier properties such as the relative contributions of electrostatic and hydrophobic interactions in different models. These results demonstrate the limitations of predicting mucomimetic behavior with common characterization techniques and highlight the importance of testing barrier performance with multiple nanoparticle formulations.
Collapse
Affiliation(s)
- Kristina L Faurschou
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Jeffrey Watchorn
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Acceleration Consortium, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| | - Jennifer Tram Su
- Department of Chemical Engineering, McGill University, Montreal, Quebec, H3A 0C5, Canada
| | - Nancy T Li
- Computational Biology, Ontario Institute for Cancer Research, Toronto, Ontario, M5G 0A3, Canada
| | - Alison P McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Frank X Gu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Acceleration Consortium, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
3
|
Yang X, Fang Z, Shang L. Functional PEGylated bacteria reinforce the intestinal mucosal barrier by effective mucus-penetration. Sci Bull (Beijing) 2024; 69:2964-2966. [PMID: 39138112 DOI: 10.1016/j.scib.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Affiliation(s)
- Xinyuan Yang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zhonglin Fang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Varricchio A, Presutti L, La Mantia I, Ciprandi G. Inter-societal Delphi Consensus on the topical nasal treatments in Italy. Multidiscip Respir Med 2024; 19. [PMID: 39229922 DOI: 10.5826/mrm.2024.991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Topical nasal therapy is widely used in clinical practice by different specialists. However, it is multifaceted and still controversial. Namely, there is no consensus about the many aspects, and there needs to be specific guidelines. Four independent experts involved 14 Italian scientific societies (concerning ENT, allergy, and pediatrics areas) to participate in generating an Intersocietal Delphi Consensus on this matter. Three iterative rounds collected experts (4 in the first round, 20 in the second round, and 45 in the third round) designed by the scientific societies based on their clinical expertise and documented scientific value. Thirty-four statements were discussed and voted on. At the second round, all statements accomplished a very high consensus grade (>95%). At the third round, many statements reached a high or very high grade of consensus (>70%). However, some statements did not obtain sufficient agreement. Consequently, there is a need to implement knowledge about this issue through educational initiatives and new studies conducted with a robust methodology. In conclusion, topical nasal therapy deserves adequate knowledge as it is widespread and fruitful in managing upper respiratory diseases.
Collapse
|
5
|
Kassaee SN, Richard D, Ayoko GA, Islam N. Lipid polymer hybrid nanoparticles against lung cancer and their application as inhalable formulation. Nanomedicine (Lond) 2024; 19:2113-2133. [PMID: 39143915 PMCID: PMC11486133 DOI: 10.1080/17435889.2024.2387530] [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: 03/14/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024] Open
Abstract
Lung cancer is a leading cause of global cancer mortality, often treated with chemotherapeutic agents. However, conventional approaches such as oral or intravenous administration of drugs yield low bioavailability and adverse effects. Nanotechnology has unlocked new gateways for delivering medicine to their target sites. Lipid-polymer hybrid nanoparticles (LPHNPs) are one of the nano-scaled delivery platforms that have been studied to exploit advantages of liposomes and polymers, enhancing stability, drug loading, biocompatibility and controlled release. Pulmonary administration of drug-loaded LPHNPs enables direct lung deposition, rapid onset of action and heightened efficacy at low doses of drugs. In this manuscript, we will review the potential of LPHNPs in management of lung cancer through pulmonary administration.
Collapse
Affiliation(s)
- Seyedeh Negin Kassaee
- School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLDQLD4001, Australia
| | - Derek Richard
- Centre for Genomics & Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLDQLD4001, Australia
| | - Godwin A. Ayoko
- School of Chemistry & Physics & Centre for Materials Science, Queensland University of Technology (QUT), Brisbane, QLDQLD4001, Australia
| | - Nazrul Islam
- School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLDQLD4001, Australia
| |
Collapse
|
6
|
Franzé S, Rama F, Scarpa E, Violatto MB, Peqini K, Gennari CGM, Anderluzzi G, Camastra R, Salmaso A, Moscatiello G, Pellegrino S, Rizzello L, Bigini P, Cilurzo F. Mucosa-penetrating liposomes for esophageal local drug delivery. Int J Pharm 2024; 661:124413. [PMID: 38960342 DOI: 10.1016/j.ijpharm.2024.124413] [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: 04/30/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
Local drug delivery to the esophagus is hampered by rapid transit time and poor permeability of the mucosa. If some strategies aimed to improve the residence time have been proposed, non-invasive approaches to increase the drug penetration in the mucosa have not been described so far. Herein, we designed mucosa-penetrating liposomes to favor the penetration and retention of curcumin (CURC) in the esophagus. A novel mucosa penetrating peptide (MPP), SLENKGP, was selected by Phage Display and conjugated to pegylated liposomes at different PEG and MPP's surface densities. Pegylation assured a long residence time of liposomes (at least 30 min) in the esophagus in vivo, but it did not favor the penetration of CURC in the mucosa. MPP-decorated liposomes instead delivered a significant higher amount of CURC in the mucosa compared to naked pegylated liposomes. Confocal microscopy studies showed that naked pegylated liposomes remain confined in the superficial layers of the mucosa whereas MPP-decorated liposomes penetrate the whole epithelium. In vitro, MPP reduced the interaction of PEG with mucin, meanwhile favoring the paracellular penetration of liposomes across epithelial cell multilayers. In conclusion, pegylated liposomes represent a valid approach to target the esophagus and the surface functionalization with MPP enhances their penetration in the mucosa.
Collapse
Affiliation(s)
- Silvia Franzé
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy.
| | - Francesco Rama
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| | - Edoardo Scarpa
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| | - Martina Bruna Violatto
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Kaliroi Peqini
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| | | | - Giulia Anderluzzi
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| | - Rebecca Camastra
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Anita Salmaso
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Giulia Moscatiello
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Sara Pellegrino
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| | - Loris Rizzello
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| | - Paolo Bigini
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Francesco Cilurzo
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133, Milan, Italy
| |
Collapse
|
7
|
Abdalla Y, McCoubrey LE, Ferraro F, Sonnleitner LM, Guinet Y, Siepmann F, Hédoux A, Siepmann J, Basit AW, Orlu M, Shorthouse D. Machine learning of Raman spectra predicts drug release from polysaccharide coatings for targeted colonic delivery. J Control Release 2024; 374:103-111. [PMID: 39127449 DOI: 10.1016/j.jconrel.2024.08.010] [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: 02/19/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Colonic drug delivery offers numerous pharmaceutical opportunities, including direct access to local therapeutic targets and drug bioavailability benefits arising from the colonic epithelium's reduced abundance of cytochrome P450 enzymes and particular efflux transporters. Current workflows for developing colonic drug delivery systems involve time-consuming, low throughput in vitro and in vivo screening methods, which hinder the identification of suitable enabling materials. Polysaccharides are useful materials for colonic targeting, as they can be utilised as dosage form coatings that are selectively digested by the colonic microbiota. However, polysaccharides are a heterogeneous family of molecules with varying suitability for this purpose. To address the need for high-throughput material selection tools for colonic drug delivery, we leveraged machine learning (ML) and publicly accessible experimental data to predict the release of the drug 5-aminosalicylic acid from polysaccharide-based coatings in simulated human, rat, and dog colonic environments. For the first time, Raman spectra alone were used to characterise polysaccharides for input as ML features. Models were validated on 8 unseen drug release profiles from new polysaccharide coatings, demonstrating the generalisability and reliability of the method. Further, model analysis facilitated an understanding of the chemical features that influence a polysaccharide's suitability for colonic drug delivery. This work represents a major step in employing spectral data for forecasting drug release from pharmaceutical formulations and marks a significant advancement in the field of colonic drug delivery. It offers a powerful tool for the efficient, sustainable, and successful development and pre-ranking of colon-targeted formulation coatings, paving the way for future more effective and targeted drug delivery strategies.
Collapse
Affiliation(s)
- Youssef Abdalla
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Fabiana Ferraro
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | | | - Yannick Guinet
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000 Lille, France
| | | | - Alain Hédoux
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000 Lille, France
| | | | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Mine Orlu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - David Shorthouse
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
8
|
Bozza A, Bordano V, Marengo A, Muntoni E, Marini E, Lazzarato L, Dianzani C, Monge C, Rosa AC, Cangemi L, Valsania MC, Colitti B, Camisassa E, Battaglia L. Green Solid Lipid Nanoparticles by Fatty Acid Coacervation: An Innovative Nasal Delivery Tool for Drugs Targeting Cerebrovascular and Neurological Diseases. Pharmaceutics 2024; 16:1051. [PMID: 39204396 PMCID: PMC11360092 DOI: 10.3390/pharmaceutics16081051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Cerebrovascular and neurological diseases are characterized by neuroinflammation, which alters the neurovascular unit, whose interaction with the choroid plexus is critical for maintaining brain homeostasis and producing cerebrospinal fluid. Dysfunctions in such process can lead to conditions such as idiopathic normal pressure hydrocephalus, a common disease in older adults. Potential pharmacological treatments, based upon intranasal administration, are worthy of investigation because they might improve symptoms and avoid surgery by overcoming the blood-brain barrier and avoiding hepatic metabolism. Nasal lipid nanocarriers, such as solid lipid nanoparticles, may increase the nasal retention and permeation of drugs. To this aim, green solid lipid nanoparticles, obtained by coacervation from natural soaps, are promising vehicles due to their specific lipid matrix composition and the unsaponifiable fraction, endowed with antioxidant and anti-inflammatory properties, and thus suitable for restoring the neurovascular unit function. In this experimental work, such green solid lipid nanoparticles, fully characterized from a physico-chemical standpoint, were loaded with a drug combination suitable for reverting hydrocephalus symptoms, allowing us to obtain a non-toxic formulation, a reduction in the production of the cerebrospinal fluid in vitro, and a vasoprotective effect on an isolated vessel model. The pharmacokinetics and biodistribution of fluorescently labelled nanoparticles were also tested in animal models.
Collapse
Affiliation(s)
- Annalisa Bozza
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Valentina Bordano
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Arianna Marengo
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Elisabetta Muntoni
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Chiara Monge
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Arianna Carolina Rosa
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Luigi Cangemi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
| | - Maria Carmen Valsania
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125 Torino, Italy;
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Turin, Via Quarello 15/a, 10135 Torino, Italy
| | - Barbara Colitti
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini, 2, 10095 Grugliasco, Italy;
| | | | - Luigi Battaglia
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (V.B.); (A.M.); (E.M.); (E.M.); (L.L.); (C.D.); (C.M.); (A.C.R.); (L.C.); (L.B.)
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Turin, Via Quarello 15/a, 10135 Torino, Italy
| |
Collapse
|
9
|
Elovitz M, Anton L, Cristancho A, Ferguson B, Joseph A, Ravel J. Vaginal microbes alter epithelial transcriptome and induce epigenomic modifications providing insight into mechanisms for susceptibility to adverse reproductive outcomes. RESEARCH SQUARE 2024:rs.3.rs-4385224. [PMID: 38854063 PMCID: PMC11160883 DOI: 10.21203/rs.3.rs-4385224/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The cervicovaginal microbiome is highly associated with women's health, with microbial communities dominated by Lactobacillus species considered optimal. Conversely, a lack of lactobacilli and a high abundance of strict and facultative anaerobes, including Gardnerella vaginalis, have been associated with adverse reproductive outcomes. However, how host-microbial interactions alter specific molecular pathways and impact cervical and vaginal epithelial function remains unclear. Using RNA-sequencing, we characterized the in vitro cervicovaginal epithelial transcriptional response to different vaginal bacteria and their culture supernatants. We showed that G. vaginalis upregulates genes associated with an activated innate immune response. Unexpectedly, G. vaginalis specifically induced inflammasome pathways through activation of NLRP3-mediated increases in caspase-1, IL-1β and cell death, while live L. crispatus had minimal transcriptomic changes on epithelial cells. L. crispatus culture supernatants resulted in a shift in the epigenomic landscape of cervical epithelial cells that was confirmed by ATAC-sequencing showing reduced chromatin accessibility. This study reveals new insights into host-microbe interactions in the lower reproductive tract and suggests potential therapeutic strategies leveraging the vaginal microbiome to improve reproductive health.
Collapse
|
10
|
Ma X, Liu Y, Wang J, Liu H, Wei G, Lu W, Liu Y. Combination of PEGylation and Cationization on Phospholipid-Coated Cyclosporine Nanosuspensions for Enhanced Ocular Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27040-27054. [PMID: 38743443 DOI: 10.1021/acsami.4c01732] [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: 05/16/2024]
Abstract
Strong precorneal clearance mechanisms including reflex blink, constant tear drainage, and rapid mucus turnover constitute great challenges for eye drops for effective drug delivery to the ocular epithelium. In this study, cyclosporine A (CsA) for the treatment of dry eye disease (DED) was selected as the model drug. Two strategies, PEGylation for mucus penetration and cationization for potent cellular uptake, were combined to construct a novel CsA nanosuspension (NS@lipid-PEG/CKC) by coating nanoscale drug particles with a mixture of lipids, DSPE-PEG2000, and a cationic surfactant, cetalkonium chloride (CKC). NS@lipid-PEG/CKC with the mean size ∼173 nm and positive zeta potential ∼+40 mV showed promoted mucus penetration, good cytocompatibility, more cellular uptake, and prolonged precorneal retention without obvious ocular irritation. More importantly, NS@lipid-PEG/CKC recovered tear production and goblet cell density more efficiently than the commercial cationic nanoemulsion on a dry eye disease rat model. All results indicated that a combination of PEGylation and cationization might provide a promising strategy to coordinate mucus penetration and cellular uptake for enhanced drug delivery to the ocular epithelium for nanomedicine-based eye drops.
Collapse
Affiliation(s)
- Xiaopei Ma
- Department of Pharmaceutics. School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yaodong Liu
- Department of Pharmaceutics. School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jun Wang
- Department of Pharmaceutics. School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Hui Liu
- Department of Breast Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Gang Wei
- Department of Pharmaceutics. School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Weiyue Lu
- Department of Pharmaceutics. School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yu Liu
- Department of Pharmaceutics. School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| |
Collapse
|
11
|
Dubashynskaya NV, Petrova VA, Skorik YA. Biopolymer Drug Delivery Systems for Oromucosal Application: Recent Trends in Pharmaceutical R&D. Int J Mol Sci 2024; 25:5359. [PMID: 38791397 PMCID: PMC11120705 DOI: 10.3390/ijms25105359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Oromucosal drug delivery, both local and transmucosal (buccal), is an effective alternative to traditional oral and parenteral dosage forms because it increases drug bioavailability and reduces systemic drug toxicity. The oral mucosa has a good blood supply, which ensures that drug molecules enter the systemic circulation directly, avoiding drug metabolism during the first passage through the liver. At the same time, the mucosa has a number of barriers, including mucus, epithelium, enzymes, and immunocompetent cells, that are designed to prevent the entry of foreign substances into the body, which also complicates the absorption of drugs. The development of oromucosal drug delivery systems based on mucoadhesive biopolymers and their derivatives (especially thiolated and catecholated derivatives) is a promising strategy for the pharmaceutical development of safe and effective dosage forms. Solid, semi-solid and liquid pharmaceutical formulations based on biopolymers have several advantageous properties, such as prolonged residence time on the mucosa due to high mucoadhesion, unidirectional and modified drug release capabilities, and enhanced drug permeability. Biopolymers are non-toxic, biocompatible, biodegradable and may possess intrinsic bioactivity. A rational approach to the design of oromucosal delivery systems requires an understanding of both the anatomy/physiology of the oral mucosa and the physicochemical and biopharmaceutical properties of the drug molecule/biopolymer, as presented in this review. This review summarizes the advances in the pharmaceutical development of mucoadhesive oromucosal dosage forms (e.g., patches, buccal tablets, and hydrogel systems), including nanotechnology-based biopolymer nanoparticle delivery systems (e.g., solid lipid particles, liposomes, biopolymer polyelectrolyte particles, hybrid nanoparticles, etc.).
Collapse
Affiliation(s)
| | | | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, 199004 St. Petersburg, Russia
| |
Collapse
|
12
|
Dishaw LJ, Litman GW, Liberti A. Tethering of soluble immune effectors to mucin and chitin reflects a convergent and dynamic role in gut immunity. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230078. [PMID: 38497268 PMCID: PMC10945408 DOI: 10.1098/rstb.2023.0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/04/2023] [Indexed: 03/19/2024] Open
Abstract
The immune system employs soluble effectors to shape luminal spaces. Antibodies are soluble molecules that effect immunological responses, including neutralization, opsonization, antibody-dependent cytotoxicity and complement activation. These molecules are comprised of immunoglobulin (Ig) domains. The N-terminal Ig domains recognize antigen, and the C-terminal domains facilitate their elimination through phagocytosis (opsonization). A less-recognized function mediated by the C-terminal Ig domains of the IgG class of antibodies (Fc region) involves the formation of multiple low-affinity bonds with the mucus matrix. This association anchors the antibody molecule to the matrix to entrap potential pathogens. Even though invertebrates are not known to have antibodies, protochordates have a class of secreted molecules containing Ig domains that can bind bacteria and potentially serve a similar purpose. The VCBPs (V region-containing chitin-binding proteins) possess a C-terminal chitin-binding domain that helps tether them to chitin-rich mucus gels, mimicking the IgG-mediated Fc trapping of microbes in mucus. The broad functional similarity of these structurally divergent, Ig-containing, secreted effectors makes a case for a unique form of convergent evolution within chordates. This opinion essay highlights emerging evidence that divergent secreted immune effectors with Ig-like domains evolved to manage immune recognition at mucosal surfaces in strikingly similar ways. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- L. J. Dishaw
- Morsani College of Medicine, Department of Pediatrics, University of South Florida, Children's Research Institute, St. Petersburg, FL 33701, USA
| | - G. W. Litman
- Morsani College of Medicine, Department of Pediatrics, University of South Florida, Children's Research Institute, St. Petersburg, FL 33701, USA
| | - A. Liberti
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, 80122 Naples, Italy
| |
Collapse
|
13
|
Barmpatsalou V, Tjakra M, Li L, Dubbelboer IR, Karlsson E, Pedersen Lomstein B, Bergström CAS. Development of a canine artificial colonic mucus model for drug diffusion studies. Eur J Pharm Sci 2024; 194:106702. [PMID: 38218203 DOI: 10.1016/j.ejps.2024.106702] [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] [Received: 09/27/2023] [Revised: 12/14/2023] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
Colonic mucus is a key factor in the colonic environment because it may affect drug absorption. Due to the similarity of human and canine gastrointestinal physiology, dogs are an established preclinical species for the assessment of controlled release formulations. Here we report the development of an artificial colonic mucus model to mimic the native canine one. In vitro models of the canine colonic environment can provide insights for early stages of drug development and contribute to the implementation of the 3Rs (refinement, reduction, and replacement) of animal usage in the drug development process. Our artificial colonic mucus could predict diffusion trends observed in native mucus and was successfully implemented in microscopic and macroscopic assays to study macromolecular permeation through the mucus. The traditional Transwell set up was optimized with the addition of a nylon filter to ensure homogenous representation of the mucus barrier in vitro. In conclusion, the canine artificial colonic mucus can be used to study drug permeation across the mucus and its flexibility allows its use in various set ups depending on the nature of the compound under investigation and equipment availability.
Collapse
Affiliation(s)
- V Barmpatsalou
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Box 580, SE-751 23, Uppsala, Sweden
| | - M Tjakra
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Box 580, SE-751 23, Uppsala, Sweden
| | - L Li
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Box 580, SE-751 23, Uppsala, Sweden
| | - I R Dubbelboer
- The Swedish Drug Delivery Center, Department of Pharmaceutical Biosciences, Uppsala University, Box 574, SE-751 23, Uppsala, Sweden
| | - E Karlsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - B Pedersen Lomstein
- Product Development & Drug Delivery, Global Pharmaceutical R&D, Ferring Pharmaceuticals A/S, Amager Strandvej 405, 2770, Kastrup, Denmark
| | - C A S Bergström
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Box 580, SE-751 23, Uppsala, Sweden.
| |
Collapse
|
14
|
Semenova MG, Antipova AS, Martirosova EI, Palmina NP, Zelikina DV, Chebotarev SA, Bogdanova NG, Anokhina MS, Kasparov VV. Key structural factors and intermolecular interactions underlying the formation, functional properties and behaviour in the gastrointestinal tract in vitro of the liposomal form of nutraceuticals coated with whey proteins and chitosan. Food Funct 2024; 15:2008-2021. [PMID: 38289251 DOI: 10.1039/d3fo04285e] [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: 02/20/2024]
Abstract
The aim of this study was to gain a better understanding of the key structural factors and intermolecular interactions underlying the formation, functionality, and in vitro gastrointestinal behaviour of the liposomal form of nutraceuticals coated with whey proteins (WPI) and chitosan (CHIT). Phosphatidylcholine (PC) liposomes were used to encapsulate a combination of hydrophobic and hydrophilic nutraceuticals. The hydrophobic constituents were long-chain (LC) n-3 PUFAs (DHA and EPA) from fish oil (FO), vitamin D3, and clove essential oil (CEO), while the hydrophilic component was γ-aminobutyric acid (GABA). A combination of physicochemical methods was used to achieve this goal, including electron paramagnetic resonance spectroscopy (EPRS), laser light scattering in dynamic, static, and electrophoretic modes, transmission electron microscopy, spectrophotometry and tensiometry. The efficiency of encapsulating the nutraceuticals in PC liposomes simultaneously was as follows: 100 ± 1% for both FO triglycerides and CEO, 82 ± 2% for vitamin D3, and 50 ± 1% for GABA. According to EPRS data, encapsulating LC PUFA reduced microviscosity at a depth of 20 Å in the PC bilayer. The co-encapsulation of other nutraceuticals in PC liposomes at selected concentrations did not alter this effect. The upper part (8 Å) of PC liposome bilayers showed an increase in rigidity parameter S, indicating the presence of D3, CEO, and partially GABA. The liposome layer-by-layer encapsulation efficiency (EE%) was achieved by using WPI to form the binary complex [WPI-(PC-FO-D3-GABA-CEO)] (EE = 50% at pH 7.0 and I = 0.001 M), followed by coating with chitosan to form the ternary complex [WPI-(PC-FO-D3-GABA-CEO)]-CHIT (EE = 80% at pH 5.1 and I = 0.001 M). The biopolymer-coated liposomes displayed high water solubility owing to their submicron sizes, thermodynamic affinity for the aqueous medium, and 20 mV ζ-potential values. The chitosan shell regulated the release of liposomes from the ternary complex during in vitro gastrointestinal digestion. In the stomach, the hydrolysis of chitosan by pepsin resulted in a 40% release of liposomes. In the small intestine, chitosan was separated from the WPI-liposome core, facilitatig its hydrolysis and resulting in a 60% release of liposomes. The bioavailability of nutraceuticals encapsulated in PC liposomes in the small intestine may be enhanced by the interactions of both non-hydrolysed and hydrolysed liposomes with bile salts and mucin.
Collapse
Affiliation(s)
- Maria G Semenova
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Anna S Antipova
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Elena I Martirosova
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Nadezhda P Palmina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Daria V Zelikina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Sergey A Chebotarev
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Natalya G Bogdanova
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Maria S Anokhina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| | - Valery V Kasparov
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Russian Federation.
| |
Collapse
|
15
|
Huang Q, Chen Y, Zhang W, Xia X, Li H, Qin M, Gao H. Nanotechnology for enhanced nose-to-brain drug delivery in treating neurological diseases. J Control Release 2024; 366:519-534. [PMID: 38182059 DOI: 10.1016/j.jconrel.2023.12.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/07/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Despite the increasing global incidence of brain disorders, achieving sufficient delivery towards the central nervous system (CNS) remains a formidable challenge in terms of translating into improved clinical outcomes. The brain is highly safeguarded by physiological barriers, primarily the blood-brain barrier (BBB), which routinely excludes most therapeutics from entering the brain following systemic administration. Among various strategies investigated to circumvent this challenge, intranasal administration, a noninvasive method that bypasses the BBB to allow direct access of drugs to the CNS, has been showing promising results. Nanotechnology-based drug delivery systems, in particular, have demonstrated remarkable capacities in overcoming the challenges posed by nose-to-brain drug delivery and facilitating targeted drug accumulation within the brain while minimizing side effects of systemic distribution. This review comprehensively summarizes the barriers of nose-to-brain drug delivery, aiming to enhance our understanding of potential physiological obstacles and improve the efficacy of nasal delivery in future trials. We then highlight cutting-edge nanotechnology-based studies that enhance nose-to-brain drug delivery in three key aspects, demonstrating substantial potential for improved treatment of brain diseases. Furthermore, the attention towards clinical studies will ease the regulatory approval process for nasal administration of nanomedicines targeting brain disease.
Collapse
Affiliation(s)
- Qianqian Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yongke Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Weiwei Zhang
- Department of Public Health, Chengdu Medical College, 783 Xindu Avenue, Xindu, Chengdu, Sichuan 610500, China
| | - Xue Xia
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Meng Qin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China.
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
16
|
Wang G, Ma F, Xie K, Li X, Tan X, Xia Y, Wang Y, Dong J. Liensinine alleviates mouse intestinal injury induced by sepsis through inhibition of oxidative stress, inflammation, and cell apoptosis. Int Immunopharmacol 2024; 127:111335. [PMID: 38101222 DOI: 10.1016/j.intimp.2023.111335] [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: 07/25/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023]
Abstract
Sepsis is a clinical syndrome triggered by an imbalanced host response to pathogens that can lead to multiple organ dysfunction. The immune response and barrier function of the gut play an important role in the pathogenesis and progression of sepsis. This study aimed to explore the potential role of natural alkaloid Liensinine in the treatment of intestinal injury caused by sepsis and its possible molecular mechanism. In this study, a mouse model of sepsis was established by injecting LPS to explore the protective effect of Liensinine on intestinal injury in sepsis. The results showed that Liensinine could reduce the intestinal damage caused by LPS and increase the number of goblet cells. Furthermore, it decreased the release of inflammatory cytokines by inhibiting NF-kB phosphorylation and NLRP3 inflammasome synthesis. Liensinine also reduced the oxidative stress and ROS accumulation caused by LPS, and played an anti-oxidative stress role by regulating the Nrf2/keap1 signaling pathway. In addition, Liensinine alleviated the inhibition of intestinal autophagy caused by LPS by inhibiting the PI3K/Akt/mTOR pathway. And then it reduced the excessive apoptosis of intestinal cells. This study provides valuable insights for sepsis prevention and treatment, offering a potential therapeutic candidate to protect against intestinal injury and regulate the inflammatory response in sepsis.
Collapse
Affiliation(s)
- Guanglu Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China; Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China
| | - Fenfen Ma
- Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China
| | - Kunmei Xie
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China; Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China
| | - Xueqing Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xuelian Tan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yan Xia
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yan Wang
- Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
17
|
Kamankesh M, Yadegar A, Llopis-Lorente A, Liu C, Haririan I, Aghdaei HA, Shokrgozar MA, Zali MR, Miri AH, Rad-Malekshahi M, Hamblin MR, Wacker MG. Future Nanotechnology-Based Strategies for Improved Management of Helicobacter pylori Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302532. [PMID: 37697021 DOI: 10.1002/smll.202302532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/25/2023] [Indexed: 09/13/2023]
Abstract
Helicobacter pylori (H. pylori) is a recalcitrant pathogen, which can cause gastric disorders. During the past decades, polypharmacy-based regimens, such as triple and quadruple therapies have been widely used against H. pylori. However, polyantibiotic therapies can disturb the host gastric/gut microbiota and lead to antibiotic resistance. Thus, simpler but more effective approaches should be developed. Here, some recent advances in nanostructured drug delivery systems to treat H. pylori infection are summarized. Also, for the first time, a drug release paradigm is proposed to prevent H. pylori antibiotic resistance along with an IVIVC model in order to connect the drug release profile with a reduction in bacterial colony counts. Then, local delivery systems including mucoadhesive, mucopenetrating, and cytoadhesive nanobiomaterials are discussed in the battle against H. pylori infection. Afterward, engineered delivery platforms including polymer-coated nanoemulsions and polymer-coated nanoliposomes are poposed. These bioinspired platforms can contain an antimicrobial agent enclosed within smart multifunctional nanoformulations. These bioplatforms can prevent the development of antibiotic resistance, as well as specifically killing H. pylori with no or only slight negative effects on the host gastrointestinal microbiota. Finally, the essential checkpoints that should be passed to confirm the potential effectiveness of anti-H. pylori nanosystems are discussed.
Collapse
Affiliation(s)
- Mojtaba Kamankesh
- Polymer Chemistry Department, School of Science, University of Tehran, PO Box 14155-6455, Tehran, 14144-6455, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | - Antoni Llopis-Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Insituto de Salud Carlos III, Valencia, 46022, Spain
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | | | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | - Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, 4 Science Drive 2, Singapore, 117545, Singapore
| |
Collapse
|
18
|
Liu W, Tu Z, Liu J, Wu T, Li D, Zhang N, Cui Y. Therapeutic effect of yinchenhao decoction on cholelithiasis via mucin in the gallbladder and intestine. Fitoterapia 2024; 172:105746. [PMID: 37967772 DOI: 10.1016/j.fitote.2023.105746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 11/07/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
Cholelithiasis is a common and frequently occurring disease worldwide that belongs to the category of jaundice in traditional Chinese medicine. Yinchenhao decoction (YD) consists of Artemisia capillaris Thunb., Gardenia jasminoides J.Ellis, and Rheum palmatum L., and is traditionally used to treat jaundice, which has a significant therapeutic effect on cholelithiasis. Our study aimed to investigate the pathological mechanism of cholelithiasis and the therapeutic mechanism of YD via mucin in the gallbladder and intestine. YD was prepared and analyzed using HPLC. The supersaturation stability experiment was designed by the solvent-shift method. The cell transport experiment was conducted by coculture monolayers. The animal experiment was performed using a cholelithiasis model with a high-cholesterol diet. The related indicators were detected by automatic biochemical analyzer, PCR, western blot, or ELISA. Statistics were analyzed using χ2-tests and t-tests. As the results, in cholelithiasis, MUC5AC highly expressed in the gallbladder shortened cholesterol supersaturation and promoted cholesterol crystallization via the inflammatory cytokine signaling pathway; MUC2 highly expressed in the small intestine prolonged cholesterol supersaturation and promoted cholesterol absorption via the inflammatory cytokine signaling pathway. YD inhibited mucin expression in the gallbladder and intestine in a concentration-dependent manner for cholelithiasis treatment by inhibiting the inflammatory cytokine signaling pathway, which was attributed to the active components, including chlorogenic acid, geniposide, and rhein.
Collapse
Affiliation(s)
- Weijun Liu
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China.
| | - Zhengwei Tu
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China
| | - Jinjin Liu
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China
| | - Teng Wu
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China
| | - Donghua Li
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China
| | - Nan Zhang
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China.
| | - Yunfeng Cui
- Tianjin NanKai Hospital, Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, No. 6 Changjiang Road, Nankai District, Tianjin 300100, China.
| |
Collapse
|
19
|
Yang S, Cai J, Su Q, Li Q, Meng X. Human milk oligosaccharides combine with Bifidobacterium longum to form the "golden shield" of the infant intestine: metabolic strategies, health effects, and mechanisms of action. Gut Microbes 2024; 16:2430418. [PMID: 39572856 PMCID: PMC11587862 DOI: 10.1080/19490976.2024.2430418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/04/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are the third most important nutrient in human milk and are the gold standard for infant nutrition. Due to the lack of an enzyme system capable of utilizing HMOs in the infant intestine, HMOs cannot be directly utilized. Instead, they function as natural prebiotics, participating in the establishment of the intestinal microbiota as a "bifidus factor." A crucial colonizer of the early intestine is Bifidobacterium longum (B. longum), particularly its subspecies B. longum subsp. infantis, which is the most active consumer of HMOs. However, due to the structural diversity of HMOs and the specificity of B. longum strains, studies on their synergy are limited. An in-depth investigation into the mechanisms of HMO utilization by B. longum is essential for applying both as synbiotics to promote early intestinal development in infants. This review describes the colonization advantages of B. longum in the infant intestinal tract and its metabolic strategies for HMOs. It also summarizes recent studies on the effect and mechanism of B. longum and HMOs in infant intestinal development directly or indirectly through the action of metabolites. In conclusion, further structural analysis of HMOs and a deeper understanding of the interactions between B. longum and HMOs, as well as clinical trials, are necessary to lay the foundation for future practical applications as synbiotics.
Collapse
Affiliation(s)
- Shuo Yang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Junwu Cai
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Qian Su
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Qiaohui Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| |
Collapse
|
20
|
Elovitz M, Anton L, Cristancho A, Ferguson B, Joseph A, Ravel J. Vaginal microbes alter epithelial transcriptomic and epigenomic modifications providing insight into the molecular mechanisms for susceptibility to adverse reproductive outcomes. RESEARCH SQUARE 2023:rs.3.rs-3580132. [PMID: 38014044 PMCID: PMC10680926 DOI: 10.21203/rs.3.rs-3580132/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The cervicovaginal microbiome is highly associated with women's health with microbial communities dominated by Lactobacillus spp. being considered optimal. Conversely, a lack of lactobacilli and a high abundance of strict and facultative anaerobes including Gardnerella vaginalis , have been associated with adverse reproductive outcomes. However, the molecular pathways modulated by microbe interactions with the cervicovaginal epithelia remain unclear. Using RNA-sequencing, we characterize the in vitro cervicovaginal epithelial transcriptional response to different vaginal bacteria and their culture supernatants. We showed that G. vaginalis upregulated genes were associated with an activated innate immune response including anti-microbial peptides and inflammasome pathways, represented by NLRP3-mediated increases in caspase-1, IL-1β and cell death. Cervicovaginal epithelial cells exposed to L. crispatus showed limited transcriptomic changes, while exposure to L. crispatus culture supernatants resulted in a shift in the epigenomic landscape of cervical epithelial cells. ATAC-sequencing confirmed epigenetic changes with reduced chromatin accessibility. This study reveals new insight into host-microbe interactions in the lower reproductive tract and suggest potential therapeutic strategies leveraging the vaginal microbiome to improve reproductive health.
Collapse
|
21
|
Ye J, Qi X. Vaginal microecology and its role in human papillomavirus infection and human papillomavirus associated cervical lesions. APMIS 2023. [PMID: 37941500 DOI: 10.1111/apm.13356] [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: 06/25/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023]
Abstract
The vaginal microecology comprises the vaginal microbiome, immune microenvironment, vaginal anatomy, and the cervicovaginal fluid, which is rich in metabolites, enzymes, and cytokines. Investigating its role in the female reproductive system holds paramount significance. The advent of next-generation sequencing enabled a more profound investigation into the structure of the vaginal microbial community in relation to the female reproductive system. Human papillomavirus infection is prevalent among women of reproductive age, and persistent oncogenic HPV infection is widely recognized as a factor associated with cervical cancer. Extensive previous research has demonstrated that dysbiosis of vaginal microbiota characterized by a reduction in Lactobacillus species, heightens susceptivity to HPV infection, consequently contributing to persistent HPV infection and the progression of cervical lesion. Likewise, HPV infection can exacerbate dysbiosis. This review aims to provide a comprehensive summary of current literatures and to elucidate potential mechanisms underlying the interaction between vaginal microecology and HPV infection, with the intention of offering valuable insights for future clinical interventions.
Collapse
Affiliation(s)
- Jiatian Ye
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Bartlett BA, Feng Y, Fromen CA, Ford Versypt AN. Computational fluid dynamics modeling of aerosol particle transport through lung airway mucosa. Comput Chem Eng 2023; 179:108458. [PMID: 37946856 PMCID: PMC10634618 DOI: 10.1016/j.compchemeng.2023.108458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Delivery of aerosols to the lung can treat various lung diseases. However, the conducting airways are coated by a protective mucus layer with complex properties that make this form of delivery difficult. Mucus is a non-Newtonian fluid and is cleared from the lungs over time by ciliated cells. Further, its gel-like structure hinders the diffusion of particles through it. Any aerosolized treatment of lung diseases must penetrate the mucosal barrier. Using computational fluid dynamics, a model of the airway mucus and periciliary layer was constructed to simulate the transport of impacted aerosol particles. The model predicts the dosage fraction of particles of a certain size that penetrate the mucus and reach the underlying tissue, as well as the distance downstream of the dosage site where tissue concentration is maximized. Reactions that may occur in the mucus are also considered, with simulated data for the interaction of a model virus and an antibody.
Collapse
Affiliation(s)
- Blake A. Bartlett
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, USA
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Yu Feng
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, USA
| | - Catherine A. Fromen
- Department of Chemical & Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Ashlee N. Ford Versypt
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, USA
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
23
|
Ruiz-Durán S, Tenorio CM, Vico-Zúñiga I, Manzanares S, Puertas-Prieto A, Altmäe S, Vargas E. Microenvironment of the Lower Reproductive Tract: Focus on the Cervical Mucus Plug. Semin Reprod Med 2023; 41:200-208. [PMID: 38262442 DOI: 10.1055/s-0043-1778661] [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: 01/25/2024]
Abstract
The female lower reproductive tract microbiota is a complex ecosystem comprising various microorganisms that play a pivotal role in maintaining women's reproductive well-being. During pregnancy, the vaginal microbiota undergoes dynamic changes that are important for a successful gestation. This review summarizes the implications of the cervical mucus plug microenvironment and its profound impact on reproductive health. Further, the symbiotic relationship between the vaginal microbiome and the cervical mucus plug is highlighted, with a special emphasis on how this natural barrier serves as a guardian against ascending infections. Understanding this complex host-microbes interplay could pave the way for innovative approaches to improve women's reproductive health and fertility.
Collapse
Affiliation(s)
- Susana Ruiz-Durán
- Department of Obstetrics and Gynaecology, Virgen de las Nieves University Hospital, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Celia M Tenorio
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Irene Vico-Zúñiga
- Department of Obstetrics and Gynaecology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Sebastián Manzanares
- Department of Obstetrics and Gynaecology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Alberto Puertas-Prieto
- Department of Obstetrics and Gynaecology, Virgen de las Nieves University Hospital, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Signe Altmäe
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Eva Vargas
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| |
Collapse
|
24
|
Zhang H, Zhang J, Wang C, Wu D, Shi G. Extract of Sophorae flavescentis radix-Cnidii fructus couplet medicines treats vulvovaginal candidiasis by affecting the vaginal mucosal barrier. Future Microbiol 2023; 18:809-824. [PMID: 37668465 DOI: 10.2217/fmb-2023-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023] Open
Abstract
Aim: This study investigated the inhibition of extract of Sophorae flavescentis radix-Cnidii fructus couplet medicines (ESCC) on Candida albicans (C. albicans) in vitro and the effect of ESCC on the vaginal mucosal barrier in vivo. Materials & methods: Susceptibility testing was performed with C. albicans SC5314. A vulvovaginal candidiasis mouse model was successfully established. The plate method, Gram staining, hematoxylin and eosin staining and ELISA were used to detect relevant inflammatory indexes: IFN-γ, IL-1 and TNF-α. Quantitative real-time PCR and western blot were used to detect mucosal immune-related factors: MUC1, MUC4, DEFB1 and DEFB2. Results: ESCC was able to inhibit the proliferative activity of C. albicans, and it affected inflammation-related factors and indicators of vaginal mucosal immunity. Conclusion: ESCC showed potential value in the treatment of vulvovaginal candidiasis.
Collapse
Affiliation(s)
- Haitao Zhang
- Research Institute of Integrated Traditional Chinese & Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
| | - Jiaping Zhang
- Research Institute of Integrated Traditional Chinese & Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
| | - Changzhong Wang
- Research Institute of Integrated Traditional Chinese & Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Department of Pathogenic Biology & Immunology, College of Integrated Chinese & Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Daqiang Wu
- Research Institute of Integrated Traditional Chinese & Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Department of Pathogenic Biology & Immunology, College of Integrated Chinese & Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Gaoxiang Shi
- Research Institute of Integrated Traditional Chinese & Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Department of Pathogenic Biology & Immunology, College of Integrated Chinese & Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| |
Collapse
|
25
|
Zhang S, Zhu C, Huang W, Liu H, Yang M, Zeng X, Zhang Z, Liu J, Shi J, Hu Y, Shi X, Wang ZH. Recent progress of micro/nanomotors to overcome physiological barriers in the gastrointestinal tract. J Control Release 2023; 360:514-527. [PMID: 37429360 DOI: 10.1016/j.jconrel.2023.07.005] [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: 03/28/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023]
Abstract
Oral administration is a convenient administration route for gastrointestinal disease therapy with good patient compliance. But the nonspecific distribution of the oral drugs may cause serious side effects. In recent years, oral drug delivery systems (ODDS) have been applied to deliver the drugs to the gastrointestinal disease sites with decreased side effects. However, the delivery efficiency of ODDS is tremendously limited by physiological barriers in the gastrointestinal sites, such as the long and complex gastrointestinal tract, mucus layer, and epithelial barrier. Micro/nanomotors (MNMs) are micro/nanoscale devices that transfer various energy sources into autonomous motion. The outstanding motion characteristics of MNMs inspired the development of targeted drug delivery, especially the oral drug delivery. However, a comprehensive review of oral MNMs for the gastrointestinal diseases therapy is still lacking. Herein, the physiological barriers of ODDS were comprehensively reviewed. Afterward, the applications of MNMs in ODDS for overcoming the physiological barriers in the past 5 years were highlighted. Finally, future perspectives and challenges of MNMs in ODDS are discussed as well. This review will provide inspiration and direction of MNMs for the therapy of gastrointestinal diseases, pushing forward the clinical application of MNMs in oral drug delivery.
Collapse
Affiliation(s)
- Shuhao Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Chaoran Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Wanting Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Hua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Mingzhu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Xuejiao Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| | - Xiufang Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| | - Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| |
Collapse
|
26
|
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
|
27
|
Taherali F, Chouhan N, Wang F, Lavielle S, Baran M, McCoubrey LE, Basit AW, Yadav V. Impact of Peptide Structure on Colonic Stability and Tissue Permeability. Pharmaceutics 2023; 15:1956. [PMID: 37514143 PMCID: PMC10384666 DOI: 10.3390/pharmaceutics15071956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Most marketed peptide drugs are administered parenterally due to their inherent gastrointestinal (GI) instability and poor permeability across the GI epithelium. Several molecular design techniques, such as cyclisation and D-amino acid (D-AA) substitution, have been proposed to improve oral peptide drug bioavailability. However, very few of these techniques have been translated to the clinic. In addition, little is known about how synthetic peptide design may improve stability and permeability in the colon, a key site for the treatment of inflammatory bowel disease and colorectal cancer. In this study, we investigated the impact of various cyclisation modifications and D-AA substitutions on the enzymatic stability and colonic tissue permeability of native oxytocin and 11 oxytocin-based peptides. Results showed that the disulfide bond cyclisation present in native oxytocin provided an improved stability in a human colon model compared to a linear oxytocin derivative. Chloroacetyl cyclisation increased native oxytocin stability in the colonic model at 1.5 h by 30.0%, whereas thioether and N-terminal acetylated cyclisations offered no additional protection at 1.5 h. The site and number of D-AA substitutions were found to be critical for stability, with three D-AAs at Tyr, Ile and Leu, improving native oxytocin stability at 1.5 h in both linear and cyclic structures by 58.2% and 79.1%, respectively. Substitution of three D-AAs into native cyclic oxytocin significantly increased peptide permeability across rat colonic tissue; this may be because D-AA substitution favourably altered the peptide's secondary structure. This study is the first to show how the strategic design of peptide therapeutics could enable their delivery to the colon via the oral route.
Collapse
Affiliation(s)
- Farhan Taherali
- Intract Pharma Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
- Sygnature Discovery, Bio City, Pennyfoot Street, Nottingham NG1 1GR, UK
| | - Nerisha Chouhan
- Intract Pharma Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | - Fanjin Wang
- Intract Pharma Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | | | - Maryana Baran
- Orbit Discovery, Schrodinger Building, Heatley Rd, Oxford OX4 4GE, UK
| | - Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Vipul Yadav
- Intract Pharma Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| |
Collapse
|
28
|
CIPRANDI G. Chitosan-based (Captomucil® complex) medical device nasal spray for relieving symptoms in patients with rhinitis: a primary care study. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2023; 182. [DOI: 10.23736/s0393-3660.23.05042-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
|
29
|
Ferreira LMB, Cardoso VMO, Dos Santos Pedriz I, Souza MPC, Ferreira NN, Chorilli M, Gremião MPD, Zucolotto V. Understanding mucus modulation behavior of chitosan oligomers and dextran sulfate combining light scattering and calorimetric observations. Carbohydr Polym 2023; 306:120613. [PMID: 36746564 DOI: 10.1016/j.carbpol.2023.120613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
This study reports the fundamental understanding of mucus-modulatory strategies combining charged biopolymers with distinct molecular weights and surface charges. Here, key biophysical evidence supports that low-molecular-weight (Mw) polycation chitosan oligosaccharides (COSs) and high-Mw polyanion dextran sulfate (DS) exhibit distinct thermodynamic signatures upon interaction with mucin (MUC), the main protein of mucus. While the COS → MUC microcalorimetric titrations released ~14 kcal/mol and ~60 kcal/mol, the DS → MUC titrations released ~1200 and ~1450 kcal/mol at pH of 4.5 and 6.8, respectively. The MPT-2 titrations of COS → MUC and DS → MUC indicated a greater zeta potential variation at pH = 4.5 (relative variation = 815 % and 351 %, respectively) than at pH = 6.8 (relative variation = 282 % and 136 %, respectively). Further, the resultant binary (COS-MUC) and ternary (COS-DS-MUC) complexes showed opposite behavior (aggregation and charge inversion events) according to the pH environment. Most importantly, the results indicate that electrostatics could not be the driving force that governs COS-MUC interactions. To account for this finding, we proposed a two-level abstraction model. Macro features emerge collectively from individual interactions occurring at the molecular level. Therefore, to understand the outcomes of mucus modulatory strategy based on charged biopolymers it is necessary to integrate both visions into the same picture.
Collapse
Affiliation(s)
- Leonardo M B Ferreira
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil.
| | - Valéria M O Cardoso
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| | - Igor Dos Santos Pedriz
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| | - Maurício P C Souza
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Natália N Ferreira
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Maria P D Gremião
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| |
Collapse
|
30
|
Zhang C, Gao X, Ren X, Xu T, Peng Q, Zhang Y, Chao Z, Jiang W, Jia L, Han L. Bacteria-Induced Colloidal Encapsulation for Probiotic Oral Delivery. ACS NANO 2023; 17:6886-6898. [PMID: 36947056 DOI: 10.1021/acsnano.3c00600] [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: 06/18/2023]
Abstract
Probiotic oral delivery has crucial implications in biomedical engineering, but its oral bioavailability remains unsatisfactory because of the limited survival and colonization of probiotics in the harsh gastrointestinal tract. Here, a bacteria-induced encapsulation strategy is achieved by assembling metastable colloids to enhance the oral bioavailability of probiotics. The colloids (NTc) composed of amino-modified poly-β-cyclodextrin and tannic acid are formed based on the balance of host-guest interaction-driven attraction and electrostatic repulsion between colloids. Negatively charged probiotics electrostatically attract positively charged NTc to break the balance and induce further assembly surrounding the probiotics. Through a facile one-step mixing, 97% of bacteria are rapidly encapsulated into NTc shells within 10 s, with a high utilization rate of feeding colloids of 91%. More importantly, we show that the compact, thick, and positively charged NTc shells synergistically endow the encapsulated probiotics with strong resistance against simulated gastric fluid with an excellent survival rate of up to 19%, 7500 times superior to the commercial enteric material L100. Moreover, owing to the dynamically noncovalent and self-adaptive nature of host-guest interactions, NTc shells support the proliferation of the encapsulated EcN comparable with that of the naked EcN. In vitro and in vivo experiments also confirm that the NTc-encapsulated probiotics possess durable intestinal adhesion, continuous proliferation activity, enhanced oral bioavailability, good oral biosafety, and excellent therapeutic efficacy in a colitis mouse model. This facile bacteria-induced colloidal encapsulation strategy may extend to various microbes as oral bioagents for treating various diseases.
Collapse
Affiliation(s)
- Chong Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Xiaorong Gao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Xinxiu Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Ting Xu
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Qiang Peng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Yixin Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Zhenhua Chao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Wenning Jiang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| |
Collapse
|
31
|
Arzi RS, Davidovich-Pinhas M, Cohen N, Sosnik A. An experimental and theoretical approach to understand the interaction between particles and mucosal tissues. Acta Biomater 2023; 158:449-462. [PMID: 36596435 DOI: 10.1016/j.actbio.2022.12.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/24/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023]
Abstract
Nanonization of poorly water-soluble drugs has shown great potential in improving their oral bioavailability by increasing drug dissolution rate and adhesion to the gastrointestinal mucus. However, the fundamental features that govern the particle-mucus interactions have not been investigated in a systematic way before. In this work, we synthesize mucin hydrogels that mimic those of freshly excised porcine mucin. By using fluorescent pure curcumin particles, we characterize the effect of particle size (200 nm, and 1.2 and 1.3 μm), concentration (18, 35, and 71 μg mL-1), and hydrogel crosslinking density on the diffusion-driven particle penetration in vitro. Next, we derive a phenomenological model that describes the physics behind the diffusion-derived penetration and considers the contributions of the key parameters assessed in vitro. Finally, we challenge our model by assessing the oral pharmacokinetics of an anti-cancer model drug, namely dasatinib, in pristine and nanonized forms and two clinically relevant doses in rats. For a dose of 10 mg kg-1, drug nanonization leads to a significant ∼8- and ∼21-fold increase of the drug oral bioavailability and half-life, respectively, with respect to the unprocessed drug. When the dose of the nanoparticles was increased to 15 mg kg-1, the oral bioavailability increased though not significantly, suggesting the saturation of the mucus penetration sites, as demonstrated by the in vitro model. Our overall results reveal the potential of this approach to pave the way for the development of tools that enable a more rational design of nano-drug delivery systems for mucosal administration. STATEMENT OF SIGNIFICANCE: The development of experimental-theoretical tools to understand and predict the diffusion-driven penetration of particles into mucus is crucial not only to rationalize the design of nanomedicines for mucosal administration but also to anticipate the risks of the exposure of the body to nano-pollutants. However, a systematic study of such tools is still lacking. Here we introduce an experimental-theoretical approach to predict the diffusion-driven penetration of particles into mucus and investigate the effect of three key parameters on this interaction. Then, we challenge the model in a preliminary oral pharmacokinetics study in rats which shows a very good correlation with in vitro results. Overall, this work represents a robust platform for the modelling of the interaction of particles with mucosae under dynamic conditions.
Collapse
Affiliation(s)
- Roni Sverdlov Arzi
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Maya Davidovich-Pinhas
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Noy Cohen
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
32
|
Perez VL, Mah FS, Willcox M, Pflugfelder S. Anti-Inflammatories in the Treatment of Dry Eye Disease: A Review. J Ocul Pharmacol Ther 2023; 39:89-101. [PMID: 36796014 DOI: 10.1089/jop.2022.0133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Inflammation is an important driver of dry eye disease (DED) pathogenesis. An initial insult that results in the loss of tear film homeostasis can initiate a nonspecific innate immune response that leads to a chronic and self-sustaining inflammation of the ocular surface, which results in classic symptoms of dry eye. This initial response is followed by a more prolonged adaptive immune response, which can perpetuate and aggravate inflammation and result in a vicious cycle of chronic inflammatory DED. Effective anti-inflammatory therapies can help patients exit this cycle, and effective diagnosis of inflammatory DED and selection of the most appropriate treatment are therefore key to successful DED management and treatment. This review explores the cellular and molecular mechanisms of the immune and inflammatory components of DED, and examines the evidence base for the use of currently available topical treatment options. These agents include topical steroid therapy, calcineurin inhibitors, T cell integrin antagonists, antibiotics, autologous serum/plasma therapy, and omega-3 fatty acid dietary supplements.
Collapse
Affiliation(s)
- Victor L Perez
- Department of Ophthalmology, Foster Center for Ocular Immunology at Duke Eye Center, Duke University School of Medicine, Durham, North Carolina. USA
| | - Francis S Mah
- Scripps Clinic Torrey Pines, La Jolla, California, USA
| | - Mark Willcox
- School of Optometry and Vision Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Stephen Pflugfelder
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
33
|
Zheng L, Luo M, Zhou H, Chen J. Natural products from plants and microorganisms: Novel therapeutics for chronic kidney disease via gut microbiota regulation. Front Pharmacol 2023; 13:1068613. [PMID: 36733377 PMCID: PMC9887141 DOI: 10.3389/fphar.2022.1068613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
Abstract
Dysbiosis of gut microbiota plays a fundamental role in the pathogenesis and development of chronic kidney disease (CKD) and its complications. Natural products from plants and microorganisms can achieve recognizable improvement in renal function and serve as an alternative treatment for chronic kidney disease patients with a long history, yet less is known on its beneficial effects on kidney injury by targeting the intestinal microbiota. In this review, we summarize studies on the effects of natural products from plants and microorganisms, including herbal medicines and their bioactive extracts, polysaccharides from plants and microorganisms, and phytochemicals, on the prevention and treatment of chronic kidney disease through targeting gut microflora. We describe the strategies of these anti-CKD effects in animal experiments including remodulation of gut microbiota structure, reduction of uremic toxins, enhancement of short-chain fatty acid (SCFA) production, regulation of intestinal inflammatory signaling, and improvement in intestinal integrity. Meanwhile, the clinical trials of different natural products in chronic kidney disease clinical practice were also analyzed and discussed. These provide information to enable a better understanding of the renoprotective effects of these effective natural products from plants and microorganisms in the treatment of chronic kidney disease. Finally, we propose the steps to prove the causal role of the intestinal microflora in the treatment of chronic kidney disease by natural products from plants and microorganisms. We also assess the future perspective that natural active products from plants and microorganisms can beneficially delay the onset and progression of kidney disease by targeting the gut flora and highlight the remaining challenges in this area. With the continuous deepening of studies in recent years, it has been proved that gut microbiota is a potential target of natural active products derived from plants and microorganisms for chronic kidney disease treatment. Fully understanding the functions and mechanisms of gut microbiota in these natural active products from plants and microorganisms is conducive to their application as an alternative therapeutic in the treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Lin Zheng
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Mingjing Luo
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Haokui Zhou
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Jianping Chen
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
34
|
McCoubrey LE, Favaron A, Awad A, Orlu M, Gaisford S, Basit AW. Colonic drug delivery: Formulating the next generation of colon-targeted therapeutics. J Control Release 2023; 353:1107-1126. [PMID: 36528195 DOI: 10.1016/j.jconrel.2022.12.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/26/2022]
Abstract
Colonic drug delivery can facilitate access to unique therapeutic targets and has the potential to enhance drug bioavailability whilst reducing off-target effects. Delivering drugs to the colon requires considered formulation development, as both oral and rectal dosage forms can encounter challenges if the colon's distinct physiological environment is not appreciated. As the therapeutic opportunities surrounding colonic drug delivery multiply, the success of novel pharmaceuticals lies in their design. This review provides a modern insight into the key parameters determining the effective design and development of colon-targeted medicines. Influential physiological features governing the release, dissolution, stability, and absorption of drugs in the colon are first discussed, followed by an overview of the most reliable colon-targeted formulation strategies. Finally, the most appropriate in vitro, in vivo, and in silico preclinical investigations are presented, with the goal of inspiring strategic development of new colon-targeted therapeutics.
Collapse
Affiliation(s)
- Laura E McCoubrey
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alessia Favaron
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Atheer Awad
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Mine Orlu
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Simon Gaisford
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Abdul W Basit
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK.
| |
Collapse
|
35
|
Ma N, Guo P, Chen J, Qi Z, Liu C, Shen J, Sun Y, Chen X, Chen GQ, Ma X. Poly-β-hydroxybutyrate alleviated diarrhea and colitis via Lactobacillus johnsonii biofilm-mediated maturation of sulfomucin. SCIENCE CHINA. LIFE SCIENCES 2022:10.1007/s11427-022-2213-6. [PMID: 36580163 DOI: 10.1007/s11427-022-2213-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/04/2022] [Indexed: 12/30/2022]
Abstract
Maintainance of sulfomucin is a key end point in the treatment of diarrhea and inflammatory bowel disease (IBD). However, the mechanisms underlying the microbial sense to sulfomucin are poorly understood, and to date, there are no therapies targeting the secretion and maturation of sulfomucin in IBD. Herein, we biosynthesized poly-β-hydroxybutyrate (PHB) and found that PHB could alleviate inflammation caused by diarrhea and colitis by enhancing the differentiation of sulfomucin. Microbiota transplantation and clearance together demonstrate that PHB promoting sulfomucin is mediated by Lactobacillus johnsonii (L. johnsonii). Further studies revealed that PHB provides a favorable niche for L. johnsonii biofilm formation to resist disturbance and support its growth. L. johnsonii-biofilm alleviates colitis by regulating fucose residues to promote goblet cell differentiation and subsequent sulfomucin maturation. Importantly, PHB alleviates colitis by enhancing sulfomucin secretion and maturation in a L. johnsonii-dependent manner. PHB represents a class of guardians, acting as a safe probiotic-biofilm delivery system that significantly promotes probiotic proliferation. Altogether, this study adds weight to the possible role of probiotics and functional materials in the treatment of intestinal inflammation. The application of PHB and biofilm self-coating L. johnsonii carries high translational potential and may be of clinical relevance.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Pingting Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jingyu Chen
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Zengkai Qi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chunchen Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiakun Shen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yiwei Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiyue Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guo-Qiang Chen
- Center for Synthetic and Systems Biology, MOE Key Lab for Industrial Biocatalysis, School of Life Sciences and Dept Chemical Engineering, Tsinghua University, Beijing, 100084, China.
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
36
|
Shapiro RL, DeLong K, Zulfiqar F, Carter D, Better M, Ensign LM. In vitro and ex vivo models for evaluating vaginal drug delivery systems. Adv Drug Deliv Rev 2022; 191:114543. [PMID: 36208729 PMCID: PMC9940824 DOI: 10.1016/j.addr.2022.114543] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/26/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
Vaginal drug delivery systems are often preferred for treating a variety of diseases and conditions of the female reproductive tract (FRT), as delivery can be more targeted with less systemic side effects. However, there are many anatomical and biological barriers to effective treatment via the vaginal route. Further, biocompatibility with the local tissue and microbial microenvironment is desired. A variety of in vitro and ex vivo models are described herein for evaluating the physicochemical properties and toxicity profile of vaginal drug delivery systems. Deciding whether to utilize organoids in vitro or fresh human cervicovaginal mucus ex vivo requires careful consideration of the intended use and the formulation characteristics. Optimally, in vitro and ex vivo experimentation will inform or predict in vivo performance, and examples are given that describe utilization of a range of methods from in vitro to in vivo. Lastly, we highlight more advanced model systems for other mucosa as inspiration for the future in model development for the FRT.
Collapse
Affiliation(s)
- Rachel L Shapiro
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, 3400 N Charles St., Baltimore, MD 21218, USA.
| | - Kevin DeLong
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA.
| | - Fareeha Zulfiqar
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA.
| | - Davell Carter
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA.
| | - Marina Better
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA.
| | - Laura M Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA; Departments of Gynecology and Obstetrics, Infectious Diseases, and Oncology, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA; Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA.
| |
Collapse
|
37
|
Ueda Y, Mogami H, Kawamura Y, Takakura M, Inohaya A, Yasuda E, Matsuzaka Y, Chigusa Y, Ito S, Mandai M, Kondoh E. Cervical MUC5B and MUC5AC are Barriers to Ascending Pathogens During Pregnancy. J Clin Endocrinol Metab 2022; 107:3010-3021. [PMID: 36112402 DOI: 10.1210/clinem/dgac545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Indexed: 12/15/2022]
Abstract
CONTEXT Cervical excision is a risk factor for preterm birth. This suggests that the cervix plays an essential role in the maintenance of pregnancy. OBJECTIVE We investigated the role of the cervix through proteomic analysis of cervicovaginal fluid (CVF) from pregnant women after trachelectomy surgery, the natural model of a lack of cervix. METHODS The proteome compositions of CVF in pregnant women after trachelectomy were compared with those in control pregnant women by liquid chromatography-tandem mass spectrometry and label-free relative quantification. MUC5B/AC expression in the human and murine cervices was analyzed by immunohistochemistry. Regulation of MUC5B/AC expression by sex steroids was assessed in primary human cervical epithelial cells. In a pregnant mouse model of ascending infection, Escherichia coli or phosphate-buffered saline was inoculated into the vagina at 16.5 dpc, and the cervices were collected at 17.5 dpc. RESULTS The expression of MUC5B/5AC in cervicovaginal fluid was decreased in pregnant women after trachelectomy concomitant with the anatomical loss of cervical glands. Post-trachelectomy women delivered at term when MUC5B/AC abundance was greater than the mean normalized abundance of the control. MUC5B levels in the cervix were increased during pregnancy in both humans and mice. MUC5B mRNA was increased by addition of estradiol in human cervical epithelial cells, whereas MUC5AC was not. In a pregnant mouse model of ascending infection, E. coli was trapped in the MUC5B/AC-expressing mucin of the cervix, and neutrophils were colocalized there. CONCLUSION Endocervical MUC5B and MUC5AC may be barriers to ascending pathogens during pregnancy.
Collapse
Affiliation(s)
- Yusuke Ueda
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Haruta Mogami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yosuke Kawamura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masahito Takakura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Asako Inohaya
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Eriko Yasuda
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yu Matsuzaka
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Eiji Kondoh
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
38
|
Li B, Ding M, Liu X, Zhao J, Ross RP, Stanton C, Yang B, Chen W. Bifidobacterium breve CCFM1078 Alleviates Collagen-Induced Arthritis in Rats via Modulating the Gut Microbiota and Repairing the Intestinal Barrier Damage. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14665-14678. [PMID: 36377740 DOI: 10.1021/acs.jafc.2c04602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
This study focused on the effects of Bifidobacterium breve CCFM1078 on the intestinal barrier and systemic inflammation of collagen-induced arthritis (CIA) rats. Female rats were divided into three groups with daily intragastric administration of either saline (control group and model group) or B. breve CCFM1078 (CCFM1078 group, 3 × 109cfu/rat per day) for 5 weeks. In the Model and CCFM1078 groups, arthritis was induced by subcutaneous collagen injection. We found that B. breve CCFM1078 can repair the intestinal barrier, reduce LPS translocation, regulate gut microbiota composition, and increase short-chain fatty acids in the intestine. Then, it can reduce pro-inflammatory cytokines release, adjust immune dysfunction, and inhibit TLR4-MyD88-dependent pathways and downstream inflammatory pathways to alleviate joint inflammation in CIA rats. These findings suggest that B. breve CCFM1078 may alleviate joint inflammation by adjusting the profile of gut microbiota and enhancing the intestinal barrier.
Collapse
Affiliation(s)
- Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mengfan Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - R Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 102401, China
| |
Collapse
|
39
|
Liu W, Liu J, Wu T, Smyth H, Cui Y. The effect of mucin on supersaturation of poorly water-soluble drugs with different crystallization behavior and in vitro-in vivo correlation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
40
|
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
|
41
|
Lundquist P, Khodus G, Niu Z, Thwala LN, McCartney F, Simoff I, Andersson E, Beloqui A, Mabondzo A, Robla S, Webb DL, Hellström PM, Keita ÅV, Sima E, Csaba N, Sundbom M, Preat V, Brayden DJ, Alonso MJ, Artursson P. Barriers to the Intestinal Absorption of Four Insulin-Loaded Arginine-Rich Nanoparticles in Human and Rat. ACS NANO 2022; 16:14210-14229. [PMID: 35998570 PMCID: PMC9527806 DOI: 10.1021/acsnano.2c04330] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Peptide drugs and biologics provide opportunities for treatments of many diseases. However, due to their poor stability and permeability in the gastrointestinal tract, the oral bioavailability of peptide drugs is negligible. Nanoparticle formulations have been proposed to circumvent these hurdles, but systemic exposure of orally administered peptide drugs has remained elusive. In this study, we investigated the absorption mechanisms of four insulin-loaded arginine-rich nanoparticles displaying differing composition and surface characteristics, developed within the pan-European consortium TRANS-INT. The transport mechanisms and major barriers to nanoparticle permeability were investigated in freshly isolated human jejunal tissue. Cytokine release profiles and standard toxicity markers indicated that the nanoparticles were nontoxic. Three out of four nanoparticles displayed pronounced binding to the mucus layer and did not reach the epithelium. One nanoparticle composed of a mucus inert shell and cell-penetrating octarginine (ENCP), showed significant uptake by the intestinal epithelium corresponding to 28 ± 9% of the administered nanoparticle dose, as determined by super-resolution microscopy. Only a small fraction of nanoparticles taken up by epithelia went on to be transcytosed via a dynamin-dependent process. In situ studies in intact rat jejunal loops confirmed the results from human tissue regarding mucus binding, epithelial uptake, and negligible insulin bioavailability. In conclusion, while none of the four arginine-rich nanoparticles supported systemic insulin delivery, ENCP displayed a consistently high uptake along the intestinal villi. It is proposed that ENCP should be further investigated for local delivery of therapeutics to the intestinal mucosa.
Collapse
Affiliation(s)
- Patrik Lundquist
- Department
of Pharmacy, Uppsala University, SE-751 43 Uppsala, Sweden
| | - Georgiy Khodus
- Department
of Pharmacy, Uppsala University, SE-751 43 Uppsala, Sweden
| | - Zhigao Niu
- Department
of Pharmacy and Pharmaceutical Technology, CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela ES 15782, Spain
| | - Lungile Nomcebo Thwala
- Department
of Pharmacy and Pharmaceutical Technology, CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela ES 15782, Spain
- Université
catholique de Louvain, UCLouvain, Louvain Drug Research Institute,
Advanced Drug Delivery and Biomaterials, BE 1200 Brussels, Belgium
| | - Fiona McCartney
- UCD
School of Veterinary Medicine, University
College Dublin, Belfield D04 V1W8, Ireland
| | - Ivailo Simoff
- Department
of Pharmacy, Uppsala University, SE-751 43 Uppsala, Sweden
| | - Ellen Andersson
- Department
of Surgery in Norrköping, Linköping
University, SE-581 83 Norrköping, Sweden
- Department
of Biomedical and Clinical Sciences, Linköping
University, SE-581 83 Linköping, Sweden
| | - Ana Beloqui
- Université
catholique de Louvain, UCLouvain, Louvain Drug Research Institute,
Advanced Drug Delivery and Biomaterials, BE 1200 Brussels, Belgium
| | - Aloise Mabondzo
- CEA,
Institute of Biology and Technology of Saclay, Department of Pharmacology
and Immunoanalysis, Gif sur Yvette FR 91191, France
| | - Sandra Robla
- Department
of Pharmacy and Pharmaceutical Technology, CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela ES 15782, Spain
| | - Dominic-Luc Webb
- Department
of Medical Sciences, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Per M. Hellström
- Department
of Medical Sciences, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Åsa V Keita
- Department
of Biomedical and Clinical Sciences, Linköping
University, SE-581 83 Linköping, Sweden
| | - Eduardo Sima
- Department
of Surgical Sciences−Upper Abdominal Surgery, Uppsala University, SE-751
85 Uppsala, Sweden
| | - Noemi Csaba
- Department
of Pharmacy and Pharmaceutical Technology, CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela ES 15782, Spain
| | - Magnus Sundbom
- Department
of Surgical Sciences−Upper Abdominal Surgery, Uppsala University, SE-751
85 Uppsala, Sweden
| | - Veronique Preat
- Université
catholique de Louvain, UCLouvain, Louvain Drug Research Institute,
Advanced Drug Delivery and Biomaterials, BE 1200 Brussels, Belgium
| | - David J. Brayden
- UCD
School of Veterinary Medicine, University
College Dublin, Belfield D04 V1W8, Ireland
| | - Maria Jose Alonso
- Department
of Pharmacy and Pharmaceutical Technology, CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela ES 15782, Spain
| | - Per Artursson
- Department
of Pharmacy, Uppsala University, SE-751 43 Uppsala, Sweden
| |
Collapse
|
42
|
Puri V, Kaur VP, Singh A, Singh C. Recent advances on drug delivery applications of mucopenetrative/mucoadhesive particles: A review. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Loktionov A. Colon mucus in colorectal neoplasia and beyond. World J Gastroenterol 2022; 28:4475-4492. [PMID: 36157924 PMCID: PMC9476883 DOI: 10.3748/wjg.v28.i32.4475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/23/2022] [Accepted: 08/06/2022] [Indexed: 02/06/2023] Open
Abstract
Little was known about mammalian colon mucus (CM) until the beginning of the 21st century. Since that time considerable progress has been made in basic research addressing CM structure and functions. Human CM is formed by two distinct layers composed of gel-forming glycosylated mucins that are permanently secreted by goblet cells of the colonic epithelium. The inner layer is dense and impenetrable for bacteria, whereas the loose outer layer provides a habitat for abundant commensal microbiota. Mucus barrier integrity is essential for preventing bacterial contact with the mucosal epithelium and maintaining homeostasis in the gut, but it can be impaired by a variety of factors, including CM-damaging switch of commensal bacteria to mucin glycan consumption due to dietary fiber deficiency. It is proven that impairments in CM structure and function can lead to colonic barrier deterioration that opens direct bacterial access to the epithelium. Bacteria-induced damage dysregulates epithelial proliferation and causes mucosal inflammatory responses that may expand to the loosened CM and eventually result in severe disorders, including colitis and neoplastic growth. Recently described formation of bacterial biofilms within the inner CM layer was shown to be associated with both inflammation and cancer. Although obvious gaps in our knowledge of human CM remain, its importance for the pathogenesis of major colorectal diseases, comprising inflammatory bowel disease and colorectal cancer, is already recognized. Continuing progress in CM exploration is likely to result in the development of a range of new useful clinical applications addressing colorectal disease diagnosis, prevention and therapy.
Collapse
|
44
|
Marczynski M, Rickert CA, Fuhrmann T, Lieleg O. An improved, filtration-based process to purify functional mucins from mucosal tissues with high yields. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.121209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
45
|
Mai Y, Ouyang Y, Qin Y, Jia C, McCoubrey LE, Basit AW, Nie Y, Jia Y, Yu L, Dou L, Deng W, Deng Y, Liu Y. Poly(lactic acid)-hyperbranched polyglycerol nanoparticles enhance bioadhesive treatment of esophageal disease and reduce systemic drug exposure. NANOSCALE 2022; 14:8418-8428. [PMID: 35639565 DOI: 10.1039/d2nr01846b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The effective treatment of esophageal disease represents a significant unmet clinical need, as existing treatments often lead to unnecessary systemic drug exposure and suboptimal concentrations at the disease site. Here, surface-modified bioadhesive poly(lactic acid)-hyperbranched polyglycerol nanoparticles (BNPs), with an average 100-200 nm diameter, were developed for local and sustained esophageal drug delivery. BNPs showed significantly higher adhesion and permeation into ex vivo human and rat esophageal tissue than non-adhesive nanoparticles (NNPs) and had longer residence times within the rat esophagus in vivo. Incubation with human esophagus (Het-1A) cells confirmed BNPs' biocompatibility at clinically relevant concentrations. In a rat model of achalasia, nifedipine-loaded BNPs significantly enhanced esophageal drug exposure, increased therapeutic efficacy, and reduced systemic drug exposure compared to NNPs and free drug. The safety of BNPs was demonstrated by an absence of intestinal, hepatic, and splenic toxicity following administration. This study is the first to demonstrate the efficacy of BNPs for esophageal drug delivery and highlight their potential for improving the lives of patients suffering with esophageal conditions.
Collapse
Affiliation(s)
- Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yaqi Ouyang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yujia Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Changchang Jia
- Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
| | - Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-sen University Foshan Hospital, Foshan, 528000, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Liu Dou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
46
|
Aldossary AM, Ekweremadu CS, Offe IM, Alfassam HA, Han S, Onyali VC, Ozoude CH, Ayeni EA, Nwagwu CS, Halwani AA, Almozain NH, Tawfik EA. A guide to oral vaccination: Highlighting electrospraying as a promising manufacturing technique toward a successful oral vaccine development. Saudi Pharm J 2022; 30:655-668. [PMID: 35812139 PMCID: PMC9257926 DOI: 10.1016/j.jsps.2022.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022] Open
Abstract
Most vaccines approved by regulatory bodies are administered via intramuscular or subcutaneous injections and have shortcomings, such as the risk of needle-associated blood infections, pain and swelling at the injection site. Orally administered vaccines are of interest, as they elicit both systemic and mucosal immunities, in which mucosal immunity would neutralize the mucosa invading pathogen before the onset of an infection. Hence, oral vaccination can eliminate the injection associated adverse effects and enhance the person's compliance. Conventional approaches to manufacturing oral vaccines, such as coacervation, spray drying, and membrane emulsification, tend to alter the structural proteins in vaccines that result from high temperature, organic and toxic solvents during production. Electrohydrodynamic processes, specifically electrospraying, could solve these challenges, as it also modulates antigen release and has a high loading efficiency. This review will highlight the mucosal immunity and biological basis of the gastrointestinal immune system, different oral vaccine delivery approaches, and the application of electrospraying in vaccines development.
Collapse
Key Words
- APCs, Antigen-presenting cells
- BALT, Bronchus-associated lymphoid tissue
- DCs, Dendritic cells
- Electrospraying
- FAE, Follicle-associated epithelium
- GALT, Gut-associated lymphoid tissue
- GIT, Gastro-intestinal tract
- HIV, Human immune virus
- IL, Interleukin
- Ig, Immunoglobulin
- Infectious diseases
- MALT, Mucosa-associated lymphoid tissue
- MLN, Mesenteric lymph nodes
- MNPs, Micro/Nanoparticles
- Mucosal immunity
- Mucosal pathogen
- NALT, Nasopharynx-associated lymphoid tissue
- Oral vaccines
- PLGA, Polylactide-co-glycolide acid
- PP, Peyer’s patches
- Secretory, (SIgA1 and SIgA2)
- TGF-β, Transforming growth factor-β
- TLRs, Toll-like receptors
- WHO, World Health Organization
Collapse
Affiliation(s)
- Ahmad M. Aldossary
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Chinedu S.M. Ekweremadu
- Department of Pharmaceutics and Pharmaceutical Technology, Enugu State University of Science and Technology, Agbani, Enugu State, Nigeria
| | - Ifunanya M. Offe
- Department of Biological Sciences, Faculty of Natural Sciences and Environmental Studies, Godfrey Okoye University, Enugu, Nigeria
| | - Haya A. Alfassam
- KACST-BWH Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Sooyeon Han
- UCL Medical School, University College London, London, United Kingdom
| | - Vivian C. Onyali
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, United State
| | - Chukwuebuka H. Ozoude
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Lagos, College of Medicine Campus, Surulere, Lagos, Nigeria
| | - Emmanuel A. Ayeni
- The Research Unit, New Being Foundation, Abuja, Nigeria
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Chinekwu S. Nwagwu
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria Nsukka, Nigeria
| | - Abdulrahman A. Halwani
- Pharmaceutics Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nada H. Almozain
- Pharmaceutical Services Department, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Essam A. Tawfik
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| |
Collapse
|
47
|
Yan J, Chen Q, Tian L, Li K, Lai W, Bian L, Han J, Jia R, Liu X, Xi Z. Intestinal toxicity of micro- and nano-particles of foodborne titanium dioxide in juvenile mice: Disorders of gut microbiota-host co-metabolites and intestinal barrier damage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:153279. [PMID: 35074372 DOI: 10.1016/j.scitotenv.2022.153279] [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] [Received: 10/24/2021] [Revised: 01/09/2022] [Accepted: 01/16/2022] [Indexed: 05/28/2023]
Abstract
The wide use of TiO2 particles in food and the high exposure risk to children have prompted research into the health risks of TiO2. We used the microbiome and targeted metabolomics to explore the potential mechanism of intestinal toxicity of foodborne TiO2 micro-/nanoparticles after oral exposure for 28 days in juvenile mice. Results showed that the gut microbiota-including the abundance of Bacteroides, Bifidobacterium, Lactobacillus, and Prevotella-changed dynamically during exposure. The organic inflammatory response was activated, and lipopolysaccharide levels increased. Intestinal toxicity manifested as increased mucosal permeability, impaired intestinal barrier, immune damage, and pathological changes. The expression of antimicrobial peptides, occludin, and ZO-1 significantly reduced, while that of JNK2 and Src/pSrc increased. Compared with micro-TiO2 particles, the nano-TiO2 particles had strong toxicity. Fecal microbiota transplant confirmed the key role of gut microbiota in intestinal toxicity. The levels of gut microbiota-host co-metabolites, including pyroglutamic acid, L-glutamic acid, phenylacetic acid, and 3-hydroxyphenylacetic acid, changed significantly. Significant changes were observed in the glutathione and propanoate metabolic pathways. There was a significant correlation between the changes in gut microbiota, metabolites, and intestinal cytokine levels. These, together with the intestinal barrier damage signaling pathway, constitute the network mechanism of the intestinal toxicity of TiO2 particles.
Collapse
Affiliation(s)
- Jun Yan
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Qi Chen
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Lei Tian
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Wenqing Lai
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Liping Bian
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Jie Han
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Rui Jia
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Xiaohua Liu
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China.
| |
Collapse
|
48
|
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
|
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
|
Engineering drug delivery systems to overcome the vaginal mucosal barrier: Current understanding and research agenda of mucoadhesive formulations of vaginal delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|