1
|
Liu J, Sun J, Hu J, Xue H, Lei L, Pan X. Biomaterial-based drug delivery strategies for oral mucosa. Colloids Surf B Biointerfaces 2025; 251:114604. [PMID: 40081256 DOI: 10.1016/j.colsurfb.2025.114604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
Drug therapy is a key field in modern medicine, and the optimization of its delivery method is crucial. Traditional methods are inherently limited by first-pass effects, high-risk adverse reactions, and patient compliance challenges, which significantly restrict the effectiveness and application potential of drugs. Oral mucosal drug delivery has become a minimally invasive and effective drug delivery strategy. The unique anatomical structure of the oral mucosa facilitates the rapid absorption of drugs into the systemic circulation, thus producing rapid therapeutic effects. However, a complex oral microenvironment and mucosal barrier impede drug absorption. Biomaterials have become an important driving force for the innovative development of oral medicine, owing to their unique and excellent properties. They are widely used for preventing, diagnosing, treating, and rehabilitating oral diseases. This review explores recent advancements in biomaterial-enabled oral mucosal drug delivery systems, analyzing key physiological factors and absorption barriers that impact therapeutic outcomes. Focusing on innovative material engineering strategies highlights significant progress in extending drug residence time and improving delivery precision within the oral cavity. Furthermore, the study identifies critical challenges in translating these advancements from research to clinical practice, emphasizing the need for solutions to bridge this gap.
Collapse
Affiliation(s)
- Junhui Liu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jiao Sun
- Changsha Stomatological Hospital, Changsha 410000, China
| | - Jun Hu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Huaqian Xue
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Xiaoyi Pan
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| |
Collapse
|
2
|
Lacroix M, Moreau J, Zampaloni C, Bissantz C, Mirfendereski H, Shirvani H, Marchand S, Couet W, Chauzy A. In vitro pharmacokinetic/pharmacodynamic modeling of the effect of mucin on polymyxin B activity against Acinetobacter baumannii. Antimicrob Agents Chemother 2025; 69:e0153524. [PMID: 40135861 PMCID: PMC12057341 DOI: 10.1128/aac.01535-24] [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: 10/15/2024] [Accepted: 02/22/2025] [Indexed: 03/27/2025] Open
Abstract
The antibacterial efficacy of polymyxins in lungs may be impacted by mucin. The aim of this study was to characterize in vitro the effect of mucin on polymyxin B (PMB) activity against two multidrug-resistant Acinetobacter baumannii strains isolated from a patient before (AB121-D0) and after colistin treatment (AB122-D12), using a pharmacokinetic/pharmacodynamic (PK/PD) modeling approach. PMB binding to mucin was characterized by ultracentrifugation in cation-adjusted Mueller-Hinton broth (CAMHB) supplemented with 1% mucin. Time-kill (TK) experiments were performed in CAMHB, with 1% mucin or without as control, and with PMB total concentrations ranging from 0.25 to 512 mg/L based on the strain's minimum inhibitory concentration (MIC). For each strain, TK data were modeled based on unbound PMB concentrations. Bacterial resistance to PMB was investigated via MIC and whole genome sequencing from bacteria that regrew in the presence of antibiotics at the end of the TK experiments. PMB unbound fraction increased nonlinearly from 6% to 60% when total concentration increased from 0.5 to 512 mg/L. In addition to binding to PMB, mucin had an impact on PMB activity, which differed between the two strains. For AB121-D0, PMB activity increased in the presence of mucin resulting in a reduction of the bacterial regrowth, whereas for AB122-D12, a decrease in PMB activity was observed. Mutations in genes involved in PMB resistance appeared randomly and explained only partially the bacterial regrowth observed in TK with antibiotics. This study showed that PMB binding to mucin had a real and important impact but was not the only factor explaining the impaired PMB efficacy in the presence of mucin.
Collapse
Affiliation(s)
- Mathilde Lacroix
- Université de Poitiers INSERM, PHAR2, Poitiers, Nouvelle-Aquitaine, France
- Institut Roche–Boulogne-Billancourt, Boulogne-Billancourt, France
| | - Jérémy Moreau
- Université de Poitiers INSERM, PHAR2, Poitiers, Nouvelle-Aquitaine, France
| | - Claudia Zampaloni
- Roche Pharma Research and Early Development, Immunology, Infectious Disease and Ophthalmology, Basel, Basel-Stadt, Switzerland
| | - Caterina Bissantz
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Basel-Stadt, Switzerland
| | - Hélène Mirfendereski
- Département de Pharmacocinétique et Toxicologie, CHU Poitiers, Poitiers, Nouvelle-Aquitaine, France
| | | | - Sandrine Marchand
- Université de Poitiers INSERM, PHAR2, Poitiers, Nouvelle-Aquitaine, France
- Département de Pharmacocinétique et Toxicologie, CHU Poitiers, Poitiers, Nouvelle-Aquitaine, France
| | - William Couet
- Université de Poitiers INSERM, PHAR2, Poitiers, Nouvelle-Aquitaine, France
- Département de Pharmacocinétique et Toxicologie, CHU Poitiers, Poitiers, Nouvelle-Aquitaine, France
| | - Alexia Chauzy
- Université de Poitiers INSERM, PHAR2, Poitiers, Nouvelle-Aquitaine, France
| |
Collapse
|
3
|
Sarkar P, Iyengar D, Mukhopadhyay K. Emergence of snail mucus as a multifunctional biogenic material for biomedical applications. Acta Biomater 2025:S1742-7061(25)00330-7. [PMID: 40319989 DOI: 10.1016/j.actbio.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/23/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Snails are mollusks or shelled gastropods found everywhere on Earth. Biologically, snail mucus can be described as a multifunctional natural polymeric gel with adhesive and antimicrobial properties, rendering it a promising ingredient in pharmaceutics and biomedical applications. These properties have been exploited in cosmetics and dermatology applications over the last few years. However, the exploration of snail mucus for other biomedical applications, e.g., wound healing and drug delivery, remains new and very promising. Against this backdrop, this review explores the potential of snail mucus for a wide spectrum of biomedical applications, ranging from wound healing to cancer treatment to regenerative engineering. It will be emphasized how its application in wound healing has gained traction owing to its antimicrobial and anti-inflammatory properties. Beyond wound care, snail mucus has been investigated as a drug delivery vehicle in treating diabetes and targeted cancer therapies. While further extensive research and clinical trials are needed to solidify the efficacy of snail mucus as a biomaterial, this review will shed light on the prospect of using snail mucus alone and in combination with other natural or synthetic biopolymers as soft materials for widespread biomedical applications. STATEMENT OF SIGNIFICANCE: Exploring snail mucus as a biomaterial across various fields, including oncology, drug delivery, cosmetics, antibacterial properties, and wound healing, presents a fascinating avenue for zootherapy research. This review provides an in-depth account of the recent developments in snail mucus' potential for a broad spectrum of biomedical applications, from wound healing to cancer treatment and regenerative engineering. It highlights the growing interest in mucus' use in wound healing, attributed to its antimicrobial and anti-inflammatory properties. It has also been investigated as a drug delivery vehicle for diabetes treatment and targeted cancer therapies. The impact of such research is significant, as it could lead to the creation of innovative biomaterials for a wide range of applications, revolutionizing the field of biomaterials.
Collapse
Affiliation(s)
- Pritha Sarkar
- Department of Materials Science and Engineering, University of Central Florida, Orlando, USA
| | - Disha Iyengar
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, USA
| | - Kausik Mukhopadhyay
- Department of Materials Science and Engineering, University of Central Florida, Orlando, USA; Department of Mechanical and Aerospace Engineering - Biomedical Engineering, University of Central Florida, Orlando, USA.
| |
Collapse
|
4
|
Butnarasu C, Safferthal M, Thomas J, Povolotsky TL, Diehn R, Fentker K, Mertins P, Pagel K, Lauster DC. Structural determinants of mucins in influenza virus inhibition: The role of sialylated glycans and molecular size. Int J Biol Macromol 2025; 307:142357. [PMID: 40120898 DOI: 10.1016/j.ijbiomac.2025.142357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/03/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Mucins are heavily glycosylated proteins that play a crucial role in protecting mucosal surfaces against pathogens, including influenza viruses. This study investigates the antiviral properties of bovine submaxillary mucins (BSM) as a model for oral mucins against the influenza virus (A/H3N2 subtype), focusing on glycan composition and mucin size. BSM was purified, and characterized by proteomic and glycomic analysis and its antiviral efficacy was assessed after selective removal of sialic acids, N-glycans, or all glycans via enzymatic and chemical treatments. We employed virus binding and inhibition assays, including microscale thermophoresis (MST) and hemagglutination inhibition (HAI), to characterize processed mucins for structure activity correlations. Removal of sialic acids reduced BSM's antiviral activity by over 10-fold, while complete glycan removal abolished it entirely, highlighting sialylated O-glycans as critical for viral inhibition. N-glycan removal had minimal impact on antiviral efficacy. A size-dependent antiviral effect was observed: smaller mucin fragments (∼50 and 330 kDa), which retained comparable O-glycosylation patterns, showed significantly reduced inhibition and viral binding affinity several orders of magnitude below intact BSM. These findings underscore the importance of mucin size and sialylated O-glycans in antiviral defense mechanisms against influenza.
Collapse
Affiliation(s)
- Cosmin Butnarasu
- Institute of Pharmacy, Biopharmaceuticals, Freie Universität, Berlin 14195, Germany
| | - Marc Safferthal
- Institute of Chemistry and Biochemistry, Freie Universität, Berlin 14195, Germany; Fritz Haber Institute of the Max Planck Society, 14195 Berlin, Germany
| | - Jolly Thomas
- Institute of Pharmacy, Biopharmaceuticals, Freie Universität, Berlin 14195, Germany
| | - Tatyana L Povolotsky
- Institute of Chemistry and Biochemistry, Freie Universität, Berlin 14195, Germany
| | - Robyn Diehn
- Institute of Pharmacy, Biopharmaceuticals, Freie Universität, Berlin 14195, Germany
| | - Kerstin Fentker
- Institute of Chemistry and Biochemistry, Freie Universität, Berlin 14195, Germany; Max Delbrück Center of Molecular Medicine, Berlin 13125, Germany
| | - Philipp Mertins
- Max Delbrück Center of Molecular Medicine, Berlin 13125, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Berlin, Berlin, Germany
| | - Kevin Pagel
- Institute of Chemistry and Biochemistry, Freie Universität, Berlin 14195, Germany; Fritz Haber Institute of the Max Planck Society, 14195 Berlin, Germany
| | - Daniel C Lauster
- Institute of Pharmacy, Biopharmaceuticals, Freie Universität, Berlin 14195, Germany.
| |
Collapse
|
5
|
Degen GD, Stevens CA, Cárcamo-Oyarce G, Song J, Bej R, Tang P, Ribbeck K, Haag R, McKinley GH. Mussel-inspired cross-linking mechanisms enhance gelation and adhesion of multifunctional mucin-derived hydrogels. Proc Natl Acad Sci U S A 2025; 122:e2415927122. [PMID: 39969995 PMCID: PMC11874598 DOI: 10.1073/pnas.2415927122] [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: 08/07/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025] Open
Abstract
Mucus supports human health by hydrating, lubricating, and preventing infection of wet epithelial surfaces. The beneficial material properties and bioactivity of mucus stem from glycoproteins called mucins, motivating the development of mucin-derived hydrogels for wound dressings and antifouling coatings. However, these applications require robust gelation and adhesion to a wide range of substrates. Inspired by the chemical cross-linking and water-tolerant adhesion of marine mussel adhesive structures, we use catechol-thiol bonding to drive gelation of native mucin proteins and synthetic mucin-inspired polymers, forming soft, adhesive hydrogels that can be coated onto diverse surfaces. The gelation dynamics and adhesive properties can be systematically tuned by varying the hydrogel composition, polymer architecture, and thiol availability, with gelation timescales adjustable from seconds to hours, and values of elastic modulus, failure stress, and debonding work spanning orders of magnitude. We demonstrate the functionality of these gels in two applications: as tissue adhesives, using porcine skin as a proxy for human skin, and as bioactive surface coatings to prevent bacterial colonization. The results highlight the potential of catechol-thiol cross-linking as a versatile platform for engineering multifunctional glycoprotein hydrogels with applications in wound repair and antimicrobial surface engineering.
Collapse
Affiliation(s)
- George D. Degen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Corey A. Stevens
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Gerardo Cárcamo-Oyarce
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Jake Song
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Raju Bej
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Peng Tang
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin14195, Germany
| | - Gareth H. McKinley
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
6
|
Younas A, Asad M, Wan X, Zhang Y, Ma X, Wang L, Gu H, Shang H, Zhang N. Oregano essential oil-infused mucin microneedle patch for the treatment of hypertrophic scar. Int J Pharm 2024; 665:124748. [PMID: 39317245 DOI: 10.1016/j.ijpharm.2024.124748] [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: 07/04/2024] [Revised: 09/05/2024] [Accepted: 09/21/2024] [Indexed: 09/26/2024]
Abstract
Hypertrophic scar (HS) manifests as abnormal dermal myofibroblast proliferation and excessive collagen deposition, leading to raised scars and significant physical, psychological, and financial burdens for patients. HS is difficult to cure in the clinic and current therapies lead to recurrence, pain, and side effects. In this study, a natural amphiphilic polymer mucin is used to prepare a dissolving microneedle (muMN) that is loaded with oregano essential oil (OEO) for HS therapy. muMN exhibits sufficient skin/scar tissue penetration, quick skin recovery time after removal, good loading of natural essential oil, fast dissolution and detachment from the base layer, and good biocompatibility to applied skin. In the rabbit HS model, OEO@muMN shows a significant reduction in scar thickness, epidermal thickness index, and scar elevation index. OEO@muMN also attenuates the mean collagen area fraction and decreases the number of capillaries in scar tissues. Biochemical Assay reveals that OEO@muMN significantly inhibits the expression of transforming growth factor-β1 (TGF-β1) and hydroxyproline (HYP). In summary, this study demonstrates the feasibility and good efficacy of using the anti-proliferative and anti-oxidative OEO for HS treatment. OEO@muMN is an efficient formulation that holds the potential for clinical anti-HS application. muMN is an efficient platform to load and apply essential oils transdermally.
Collapse
Affiliation(s)
- Ayesha Younas
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China; Central Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People's Hospital, The First Affiliated Hospital of Lishui University, Lishui, Zhejiang 323000, PR China
| | - Muhammad Asad
- Henan International Joint Laboratory of Nano-Photoelectric Magnetic Material, School of Materials Science and Engineering, Henan University of Technology, Zhengzhou, Henan 450001, PR China
| | - Xiangling Wan
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Yuzhen Zhang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Xuejing Ma
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Lei Wang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Huan Gu
- Department of Chemistry and Chemical Engineering & Biomedical Engineering, Tagliatela College of Engineering, University of New Haven, West Haven, CT 06516, USA
| | - Hongtao Shang
- School of Sport Sciences (main campus), Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Nan Zhang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|
7
|
He S, Zheng Q, Ma L, Shen H, Zheng B, Zhang Y, Deng HH, Chen W, Fan K. Mucin-Triggered Osmium Nanoclusters as Protein-Corona-Like Nanozymes with Photothermal-Enhanced Peroxidase-Like Activity for Tumor-Specific Therapy. NANO LETTERS 2024; 24:14337-14345. [PMID: 39470470 DOI: 10.1021/acs.nanolett.4c04026] [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: 10/30/2024]
Abstract
Nanomaterials with peroxidase-like activity and photothermal conversion efficiency have garnered significant attention for their ability to generate cytotoxic hydroxyl radicals and provide synergistic therapeutic effects. Selecting nanozymes with suitable properties and carriers is crucial for maximizing efficacy. While the mucin family is known for its mucoadhesive, glycosylated structures that enhance drug bioavailability and targeting, its potential in nanozymes remains underexplored. Here, we utilize mucin-2 to facilitate osmium nanoclusters (Os@Mucin), creating protein-corona-like nanozymes. This configuration bestows Os@Mucin with excellent peroxidase-like activity (769 U/mg) and photothermal conversion efficiency (22.83%, 808 nm). Mucin-2 promotes Os uptake by cells, allowing Os@Mucin to exhibit tumor environment-responsive peroxidase-like activity, further enhanced under photothermal conditions for targeted cytotoxicity and synergistic effects. In vivo experiments demonstrate that this integration effectively treats triple-negative breast cancer. This study innovatively highlights the potential of the mucin family and underscores the promising role of Os nanozymes in tumor therapy.
Collapse
Affiliation(s)
- Shaobin He
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou 350004, China
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qionghua Zheng
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou 350004, China
| | - Long Ma
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Huanran Shen
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou 350004, China
| | - Bohang Zheng
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Yin Zhang
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Hao-Hua Deng
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou 350004, China
| | - Wei Chen
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou 350004, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan 451163, China
| |
Collapse
|
8
|
Zhao Z, Yuwen W, Duan Z, Zhu C, Fan D. Novel Collagen Analogs with Multicopy Mucin-Type Sequences for Multifunctional Enhancement Properties Using SUMO Fusion Tags. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:22173-22185. [PMID: 39318025 DOI: 10.1021/acs.jafc.4c07179] [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: 09/26/2024]
Abstract
Multifunctional enhanced collagen materials in green biomanufacturing are highly desired yet challenging due to the poor comprehensive performance caused by the adoption of targeting monofunctional peptides. Herein, novel collagen analog design strategy using multicopy tandem of mucin-type sequence (GAPGAPGSQGAPGLQ) derived from human COL1α1 to construct basic building blocks is reported, in which SUMO tag is added to the N-terminal of the protein as a stabilizing core. In particular, novel collagen analogs (named S1506, S1511, S1523, and S1552) with multicopy mucin-type sequences (repeated 6, 11, 23, and 52 times), which were constructed in Escherichia coli, have distinct orientation preferences of functional enhancement (including cell proliferation, differentiation, migration, antioxidant activity, and anti-inflammatory property) compared to COL1α1 in HaCaT and THP-1 cell experiments due to variant three-dimensional structures (the different-length mucin-type polypeptide chains wind around central SUMO tag). Our findings suggest that the innovative protein design and synthesis approaches employed in the construction of these novel S15 proteins have the potential to advance the development of new types of recombinant collagen analogs.
Collapse
Affiliation(s)
- Zilong Zhao
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Weigang Yuwen
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| |
Collapse
|
9
|
Stie MB, Cunha C, Huang Z, Kirkensgaard JJK, Tuelung PS, Wan F, Nielsen HM, Foderà V, Rønholt S. A head-to-head comparison of polymer interaction with mucin from porcine stomach and bovine submaxillary glands. Sci Rep 2024; 14:21350. [PMID: 39266622 PMCID: PMC11393313 DOI: 10.1038/s41598-024-72233-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
Native mucus is heterogeneous, displays high inter-individual variation and is prone to changes during harvesting and storage. To overcome the lack of reproducibility and availability of native mucus, commercially available purified mucins, porcine gastric mucin (PGM) and mucin from bovine submaxillary gland (BSM), have been widely used. However, the question is to which extent the choice of mucin matters in studies of their interaction with polymers as their composition, structure and hence physicochemical properties differ. Accordingly, the interactions between PGM or BSM with two widely used polymers in drug delivery, polyethylene oxide and chitosan, was studied with orthogonal methods: turbidity, dynamic light scattering, and quartz crystal microbalance with dissipation monitoring. Polymer binding and adsorption to the two commercially available and purified mucins, PGM and BSM, is different depending on the mucin type. PEO, known to interact weakly with mucin, only displayed limited interaction with both mucins as confirmed by all employed methods. In contrast, chitosan was able to bind to both PGM and BSM. Interestingly, the results suggest that chitosan interacts with BSM to a greater extent than with PGM indicating that the choice of mucin, PGM or BSM, can affect the outcome of studies of mucin interactions with polymers.
Collapse
Affiliation(s)
- Mai Bay Stie
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| | - Cristiana Cunha
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Zheng Huang
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Jacob Judas Kain Kirkensgaard
- Department of Food Science, Rolighedsvej 26, 1958, Frederiksberg, Denmark
- Niels Bohr Institute, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Pernille Sønderby Tuelung
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Feng Wan
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Vito Foderà
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Stine Rønholt
- LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| |
Collapse
|
10
|
Sasikumar SC, Goswami U, Raichur AM. 3D Bioprinting with Visible Light Cross-Linkable Mucin-Hyaluronic Acid Composite Bioink for Lung Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5411-5422. [PMID: 38996006 DOI: 10.1021/acsabm.4c00579] [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: 07/14/2024]
Abstract
3D printing can revolutionize personalized medicine by allowing cost-effective, customized tissue-engineering constructs. However, the limited availability and diversity of biopolymeric hydrogels restrict the variety and applications of bioinks. In this study, we introduce a composite bioink for 3D bioprinting, combining a photo-cross-linkable derivative of Mucin (Mu) called Methacrylated Mucin (MuMA) and Hyaluronic acid (HA). The less explored Mucin is responsible for the hydrogel nature of mucus and holds the potential to be used as a bioink material because of its plethora of features. HA, a crucial extracellular matrix component, is mucoadhesive and enhances ink viscosity and printability. Photo-cross-linking with 405 nm light stabilizes the printed scaffolds without damaging cells. Rheological tests reveal shear-thinning behavior, aiding cell protection during printing and improved MuMA bioink viscosity by adding HA. The printed structures exhibited porous behavior conducive to nutrient transport and cell migration. After 4 weeks in phosphate-buffered saline, the scaffolds retain 70% of their mass, highlighting stability. Biocompatibility tests with lung epithelial cells (L-132) confirm cell attachment and growth, suggesting suitability for lung tissue engineering. It is envisioned that the versatility of bioink could lead to significant advancements in lung tissue engineering and various other biomedical applications.
Collapse
Affiliation(s)
- Sruthi C Sasikumar
- Department of Materials Engineering, Indian Institute of Science Bangalore, Karnataka 560012, India
| | - Upashi Goswami
- Department of Materials Engineering, Indian Institute of Science Bangalore, Karnataka 560012, India
| | - Ashok M Raichur
- Department of Materials Engineering, Indian Institute of Science Bangalore, Karnataka 560012, India
- Institute for Nanoscience and Water Sustainability, University of South Africa, the Science Campus, Florida Park, 1710 Roodepoort, Johannesburg 1735, South Africa
| |
Collapse
|
11
|
Einhorn V, Haase H, Maares M. Interaction and competition for intestinal absorption by zinc, iron, copper, and manganese at the intestinal mucus layer. J Trace Elem Med Biol 2024; 84:127459. [PMID: 38640745 DOI: 10.1016/j.jtemb.2024.127459] [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: 02/09/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Trace elements such as zinc, manganese, copper, or iron are essential for a wide range of physiological functions. It is therefore crucial to ensure an adequate supply of these elements to the body. Many previous investigations have dealt with the role of transport proteins, in particular their selectivity for, and competition between, different ions. Another so far less well investigated major factor influencing the absorption of trace elements seems to be the intestinal mucus layer. This gel-like substance covers the entire gastrointestinal tract and its physiochemical properties can be mainly assigned to the glycoproteins it contains, so-called mucins. Interaction with mucins has already been demonstrated for some metals. However, knowledge about the impact on the respective bioavailability and competition between those metals is still sketchy. This review therefore aims to summarize the findings and knowledge gaps about potential effects regarding the interaction between gastrointestinal mucins and the trace elements iron, zinc, manganese, and copper. Mucins play an indispensable role in the absorption of these trace elements in the neutral to slightly alkaline environment of the intestine, by keeping them in a soluble form that can be absorbed by enterocytes. Furthermore, the studies so far indicate that the competition between these trace elements for uptake already starts at the intestinal mucus layer, yet further research is required to completely understand this interaction.
Collapse
Affiliation(s)
- Vincent Einhorn
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany
| | - Hajo Haase
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany
| | - Maria Maares
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany; Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal 14558, Germany.
| |
Collapse
|
12
|
Kishimoto H, Ridley C, Inoue K, Thornton DJ. Assessment of polymeric mucin-drug interactions. PLoS One 2024; 19:e0306058. [PMID: 38935605 PMCID: PMC11210812 DOI: 10.1371/journal.pone.0306058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Mucosal-delivered drugs have to pass through the mucus layer before absorption through the epithelial cell membrane. Although there has been increasing interest in polymeric mucins, a major structural component of mucus, potentially acting as important physiological regulators of mucosal drug absorption, there are no reports that have systematically evaluated the interaction between mucins and drugs. In this study, we assessed the potential interaction between human polymeric mucins (MUC2, MUC5B, and MUC5AC) and various drugs with different chemical profiles by simple centrifugal method and fluorescence analysis. We found that paclitaxel, rifampicin, and theophylline likely induce the aggregation of MUC5B and/or MUC2. In addition, we showed that the binding affinity of drugs for polymeric mucins varied, not only between individual drugs but also among mucin subtypes. Furthermore, we demonstrated that deletion of MUC5AC and MUC5B in A549 cells increased the cytotoxic effects of cyclosporin A and paclitaxel, likely due to loss of mucin-drug interaction. In conclusion, our results indicate the necessity to determine the binding of drugs to mucins and their potential impact on the mucin network property.
Collapse
Affiliation(s)
- Hisanao Kishimoto
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| | - Caroline Ridley
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - David J. Thornton
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
13
|
Kimna C, Lutz TM, Lieleg O. Fabrication and Characterization of Mucin Nanoparticles for Drug Delivery Applications. Methods Mol Biol 2024; 2763:383-394. [PMID: 38347428 DOI: 10.1007/978-1-0716-3670-1_33] [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: 02/15/2024]
Abstract
Mucin glycoproteins are ideal biomacromolecules for drug delivery applications since they naturally offer a plethora of different functional groups that can engage in specific and unspecific binding interactions with cargo molecules. However, to fabricate drug carrier objects from mucins, suitable stabilization mechanisms have to be implemented into the nanoparticle preparation procedure that allow for drug release profiles that match the requirements of the selected cargo molecule and its particular mode of action. Here, we describe two different methods to prepare crosslinked mucin nanoparticles that can release their cargo either on-demand or in a sustained manner. This method chapter includes a description of the preparation and characterization of mucin nanoparticles (stabilized either with synthetic DNA strands or with covalent crosslinks generated by free radical polymerization), as well as protocols to quantify the release of a model drug from those nanoparticles.
Collapse
Affiliation(s)
- Ceren Kimna
- School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Garching, Germany
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering, Technical University of Munich, Garching, Germany
| | - Theresa M Lutz
- School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Garching, Germany
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering, Technical University of Munich, Garching, Germany
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Garching, Germany.
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering, Technical University of Munich, Garching, Germany.
| |
Collapse
|
14
|
Wang D, Jiang Q, Dong Z, Meng T, Hu F, Wang J, Yuan H. Nanocarriers transport across the gastrointestinal barriers: The contribution to oral bioavailability via blood circulation and lymphatic pathway. Adv Drug Deliv Rev 2023; 203:115130. [PMID: 37913890 DOI: 10.1016/j.addr.2023.115130] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
Oral administration is the preferred route of drug delivery in clinical practice due to its noninvasiveness, safety, convenience, and high patient compliance. The gastrointestinal tract (GIT) plays a crucial role in facilitating the targeted delivery of oral drugs. However, the GIT presents multiple barriers that impede drug absorption, including the gastric barrier in the stomach and the mucus and epithelial barriers in the intestine. In recent decades, nanotechnology has emerged as a promising approach for overcoming these challenges by utilizing nanocarrier-based drug delivery systems such as liposomes, micelles, polymeric nanoparticles, solid lipid nanoparticles, and inorganic nanoparticles. Encapsulating drugs within nanocarriers not only protects them from degradation but also enhances their transport and absorption across the GIT, ultimately improving oral bioavailability. The aim of this review is to elucidate the mechanisms underlying nanocarrier-mediated transportation across the GIT into systemic circulation via both the blood circulation and lymphatic pathway.
Collapse
Affiliation(s)
- Ding Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Zhefan Dong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Jianwei Wang
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China; China Jinhua Institute of Zhejiang University, Jinhua 321299, PR China.
| |
Collapse
|
15
|
Watchorn J, Stuart S, Clasky AJ, Oliveira MH, Burns DC, Gu FX. Transfer-based nuclear magnetic resonance uncovers unique mechanisms for protein-polymer and protein-nanoparticle binding behavior. J Mater Chem B 2023; 11:10121-10130. [PMID: 37824091 DOI: 10.1039/d3tb01668d] [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: 10/13/2023]
Abstract
Nanoparticle-based drug delivery systems have shown increasing popularity as a means to improve patient outcomes by improving the effectiveness of active pharmaceutical ingredients (APIs). Similarly, nanoparticles have shown success in targeting alternative routes of API administration, such as applying mucoadhesion or mucopenetration to mucosal drug delivery to enhance uptake. While there are many promising examples of mucoadhesive nanomedicines in literature, there are also many examples of contradictory mucoadhesive binding behavior, most prominently in cases using the same nanoparticle materials. We have uncovered mechanistic insights in polymer-protein binding systems using nOe transfer-based NMR and sought to leverage them to explore nanoparticle-protein interactions. We tested several polymer-coated nanoparticles and micellar polymer nanoparticles and evaluated their binding with mucin proteins. We uncovered that the composition and interaction intimacy of polymer moieties that promote mucin binding change when the polymers are incorporated onto nanoparticle surfaces compared to polymer in solution. This change from solution state to nanoparticle coating can enable switching of behavior of these materials from inert to binding, as we observed in polyvinyl pyrrolidone. We also found the nanoparticle core was influential in determining the binding fate of polymer materials, whereas the nanoparticle size did not possess a clear correlation in the ranges we tested (60-270 nm). These experiments demonstrate that identical polymers may switch their binding behavior to mucin as a function of conformational changes that are induced by incorporating the polymers onto the surface of nanoparticles. These NMR-derived insights could be further leveraged to optimize nanoparticle formulations and guide polymer-mediated mucoadhesion.
Collapse
Affiliation(s)
- Jeffrey Watchorn
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.
| | - Samantha Stuart
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.
| | - Matthew H Oliveira
- Division of Engineering Science, University of Toronto, Toronto, Ontario, M5S 2E4, Canada
| | - Darcy C Burns
- Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| | - Frank X Gu
- 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
- Acceleration Consortium, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| |
Collapse
|
16
|
Donahue R, Sahoo JK, Rudolph S, Chen Y, Kaplan DL. Mucosa-Mimetic Materials for the Study of Intestinal Homeostasis and Disease. Adv Healthc Mater 2023; 12:e2300301. [PMID: 37329337 DOI: 10.1002/adhm.202300301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Mucus is a viscoelastic hydrogel that lines and protects the epithelial surfaces of the body that houses commensal microbiota and functions in host defense against pathogen invasion. As a first-line physical and biochemical barrier, intestinal mucus is involved in immune surveillance and spatial organization of the microbiome, while dysfunction of the gut mucus barrier is implicated in several diseases. Mucus can be collected from a variety of mammalian sources for study, however, established methods are challenging in terms of scale and efficiency, as well as with regard to rheological similarity to native human mucus. Therefore, there is a need for mucus-mimetic hydrogels that more accurately reflect the physical and chemical profile of the in vivo human epithelial environment to enable the investigation of the role of mucus in human disease and interactions with the intestinal microbiome. This review will evaluate the material properties of synthetic mucus mimics to date designed to address the above need, with a focus toward an improved understanding of the biochemical and immunological functions of these biopolymers related to utility for research and therapeutic applications.
Collapse
Affiliation(s)
- Rebecca Donahue
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|
17
|
Wang CM, Fernez MT, Woolston BM, Carrier RL. Native gastrointestinal mucus: Critical features and techniques for studying interactions with drugs, drug carriers, and bacteria. Adv Drug Deliv Rev 2023; 200:114966. [PMID: 37329985 PMCID: PMC11184232 DOI: 10.1016/j.addr.2023.114966] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Gastrointestinal mucus plays essential roles in modulating interactions between intestinal lumen contents, including orally delivered drug carriers and the gut microbiome, and underlying epithelial and immune tissues and cells. This review is focused on the properties of and methods for studying native gastrointestinal mucus and its interactions with intestinal lumen contents, including drug delivery systems, drugs, and bacteria. The properties of gastrointestinal mucus important to consider in its analysis are first presented, followed by a discussion of different experimental setups used to study gastrointestinal mucus. Applications of native intestinal mucus are then described, including experimental methods used to study mucus as a barrier to drug delivery and interactions with intestinal lumen contents that impact barrier properties. Given the significance of the microbiota in health and disease, its impact on drug delivery and drug metabolism, and the use of probiotics and microbe-based delivery systems, analysis of interactions of bacteria with native intestinal mucus is then reviewed. Specifically, bacteria adhesion to, motility within, and degradation of mucus is discussed. Literature noted is focused largely on applications of native intestinal mucus models as opposed to isolated mucins or reconstituted mucin gels.
Collapse
Affiliation(s)
- Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Matthew T Fernez
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Benjamin M Woolston
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, USA; Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
18
|
Yang Z, Sarkar AK, Amdursky N. Glycoproteins as a Platform for Making Proton-Conductive Free-Standing Biopolymers. Biomacromolecules 2023; 24:1111-1120. [PMID: 36787188 DOI: 10.1021/acs.biomac.2c01007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Biopolymers are an attractive environmentally friendly alternative to common synthetic polymers, whereas primarily proteins and polysaccharides are the biomacromolecules that are used for making the biopolymer. Due to the breadth of side chains of such biomacromolecules capable of participating in hydrogen bonding, proteins and polysaccharide biopolymers were also used for the making of proton-conductive biopolymers. Here, we introduce a new platform for combining the merits of both proteins and polysaccharides while using a glycosylated protein for making the biopolymer. We use mucin as our starting point, whereas being a waste of the food industry, it is a highly available and low-cost glycoprotein. We show how we can use different chemical strategies to target either the glycan part or specific amino acids for both crosslinking between the different glycoproteins, thus making a free-standing biopolymer, as well as for introducing superior proton conductivity properties to the formed biopolymer. The resultant proton-conductive soft biopolymer is an appealing candidate for any soft bioelectronic application.
Collapse
Affiliation(s)
- Ziyu Yang
- Schulich Faculty of Chemistry, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Amit Kumar Sarkar
- Schulich Faculty of Chemistry, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Nadav Amdursky
- Schulich Faculty of Chemistry, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
19
|
Park S, Chin-Hun Kuo J, Reesink HL, Paszek MJ. Recombinant mucin biotechnology and engineering. Adv Drug Deliv Rev 2023; 193:114618. [PMID: 36375719 PMCID: PMC10253230 DOI: 10.1016/j.addr.2022.114618] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/14/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Mucins represent a largely untapped class of polymeric building block for biomaterials, therapeutics, and other biotechnology. Because the mucin polymer backbone is genetically encoded, sequence-specific mucins with defined physical and biochemical properties can be fabricated using recombinant technologies. The pendent O-glycans of mucins are increasingly implicated in immunomodulation, suppression of pathogen virulence, and other biochemical activities. Recent advances in engineered cell production systems are enabling the scalable synthesis of recombinant mucins with precisely tuned glycan side chains, offering exciting possibilities to tune the biological functionality of mucin-based products. New metabolic and chemoenzymatic strategies enable further tuning and functionalization of mucin O-glycans, opening new possibilities to expand the chemical diversity and functionality of mucin building blocks. In this review, we discuss these advances, and the opportunities for engineered mucins in biomedical applications ranging from in vitro models to therapeutics.
Collapse
Affiliation(s)
- Sangwoo Park
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Joe Chin-Hun Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Heidi L Reesink
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Matthew J Paszek
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA; Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
20
|
Bej R, Haag R. Mucus-Inspired Dynamic Hydrogels: Synthesis and Future Perspectives. J Am Chem Soc 2022; 144:20137-20152. [PMID: 36074739 PMCID: PMC9650700 DOI: 10.1021/jacs.1c13547] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Indexed: 11/30/2022]
Abstract
Mucus hydrogels at biointerfaces are crucial for protecting against foreign pathogens and for the biological functions of the underlying cells. Since mucus can bind to and host both viruses and bacteria, establishing a synthetic model system that can emulate the properties and functions of native mucus and can be synthesized at large scale would revolutionize the mucus-related research that is essential for understanding the pathways of many infectious diseases. The synthesis of such biofunctional hydrogels in the laboratory is highly challenging, owing to their complex chemical compositions and the specific chemical interactions that occur throughout the gel network. In this perspective, we discuss the basic chemical structures and diverse physicochemical interactions responsible for the unique properties and functions of mucus hydrogels. We scrutinize the different approaches for preparing mucus-inspired hydrogels, with specific examples. We also discuss recent research and what it reveals about the challenges that must be addressed and the opportunities to be considered to achieve desirable de novo synthetic mucus hydrogels.
Collapse
Affiliation(s)
- Raju Bej
- Institute for Chemistry and
Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Rainer Haag
- Institute for Chemistry and
Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| |
Collapse
|
21
|
Xiao X, Teng F, Shi C, Chen J, Wu S, Wang B, Meng X, Essiet Imeh A, Li W. Polymeric nanoparticles—Promising carriers for cancer therapy. Front Bioeng Biotechnol 2022; 10:1024143. [PMID: 36277396 PMCID: PMC9585261 DOI: 10.3389/fbioe.2022.1024143] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
Polymeric nanoparticles (NPs) play an important role in controlled cancer drug delivery. Anticancer drugs can be conjugated or encapsulated by polymeric nanocarriers, which are known as polymeric nanomedicine. Polymeric nanomedicine has shown its potential in providing sustained release of drugs with reduced cytotoxicity and modified tumor retention, but until now, few delivery systems loading drugs have been able to meet clinical demands, so more efforts are needed. This research reviews the current state of the cancer drug-loading system by exhibiting a series of published articles that highlight the novelty and functions from a variety of different architectures including micelles, liposomes, dendrimers, polymersomes, hydrogels, and metal–organic frameworks. These may contribute to the development of useful polymeric NPs to achieve different therapeutic purposes.
Collapse
Affiliation(s)
- Xiao Xiao
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Fei Teng
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Changkuo Shi
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Junyu Chen
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Shuqing Wu
- School of Pharmacy, Jilin Medical University, Jilin, China
| | - Bao Wang
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, China
| | - Xiang Meng
- School of Pharmacy, Jilin Medical University, Jilin, China
| | | | - Wenliang Li
- School of Pharmacy, Jilin Medical University, Jilin, China
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
- *Correspondence: Wenliang Li,
| |
Collapse
|
22
|
Gyarmati B, Stankovits G, Szilágyi BÁ, Galata DL, Gordon P, Szilágyi A. A robust mucin-containing poly(vinyl alcohol) hydrogel model for the in vitro characterization of mucoadhesion of solid dosage forms. Colloids Surf B Biointerfaces 2022; 213:112406. [PMID: 35219220 DOI: 10.1016/j.colsurfb.2022.112406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 12/27/2022]
Abstract
Mucoadhesion testing at macroscopic scale needs a robust, convenient in vitro method as ex vivo methods suffer from poor reproducibility and ethical problems. Here we synthesized mucin-free poly(vinyl alcohol) (PVA) and mucin-containing PVA hydrogel substrates (Muc/PVA) to measure adhesion of polymer tablets. Freezing-thawing method was used for gelation to avoid chemical cross-linking and to preserve the functionality of mucin. The adhesion of first generation mucoadhesive polymers, poly(acrylic acid) (PAA) and hydroxypropylmethylcellulose (HPMC) was tested with outstanding reproducibility on individual batches of hydrogels and qualitative agreement with ex vivo literature data. Negatively charged PAA was less adhesive on Muc/PVA surface than on mucin-free PVA whereas HPMC as a neutral polymer displayed similar adhesion strength on both surfaces. Chitosan as a positively charged polymer showed enhanced adhesion on Muc/PVA substrate compared to mucin-free PVA. These results are corroborated by turbidimetric titration which indicated attractive electrostatic interactions between chitosan and mucin in contrast to the lack of attractive interactions for PAA and HPMC. These results prove the role of electronic theory in macroscopic mucoadhesion.
Collapse
Affiliation(s)
- Benjámin Gyarmati
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary.
| | - Gergely Stankovits
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Barnabás Áron Szilágyi
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Dorián László Galata
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Péter Gordon
- Department of Electronics Technology, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - András Szilágyi
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| |
Collapse
|
23
|
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: 40] [Impact Index Per Article: 13.3] [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
|
24
|
Filatova L, Emelianov G, Balabushevich N, Klyachko N. Supramolecular assemblies of mucin and lysozyme: Formation and physicochemical characterization. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
25
|
Hunt NJ, McCourt PAG, Kuncic Z, Le Couteur DG, Cogger VC. Opportunities and Challenges for Nanotherapeutics for the Aging Population. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.832524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nanotherapeutics utilize the properties of nanomaterials to alter the pharmacology of the drugs and therapies being transported, leading to changes in their biological disposition (absorption, distribution, cellular uptake, metabolism and elimination) and ultimately, their pharmacological effect. This provides an opportunity to optimize the pharmacology of drugs, particularly for those that are dependent on hepatic action. Old age is associated with changes in many pharmacokinetic processes which tend to impair drug efficacy and increase risk of toxicity. While these age-related changes are drug-specific they could be directly addressed using nanotechnology and precision targeting. The benefits of nanotherapeutics needs to be balanced against toxicity, with future use in humans dependent upon the gathering of information about the clearance and long-term safety of nanomaterials.
Collapse
|