1
|
Kaler L, Engle EM, Corkran M, Iverson E, Boboltz A, Ignacio MA, Yeruva T, Scull MA, Duncan GA. Mucus Physically Restricts Influenza A Viral Particle Access to the Epithelium. Adv Biol (Weinh) 2025; 9:e2400329. [PMID: 39936480 PMCID: PMC12001005 DOI: 10.1002/adbi.202400329] [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: 06/12/2024] [Revised: 12/20/2024] [Indexed: 02/13/2025]
Abstract
Prior work suggests influenza A virus (IAV) crosses the airway mucus barrier in a sialic acid-dependent manner through the actions of the viral envelope proteins, hemagglutinin, and neuraminidase. However, host and viral factors that influence how efficiently mucus traps IAV remain poorly defined. In this work, how the physicochemical properties of mucus influence its ability to effectively capture IAV is assessed using fluorescence video microscopy and multiple particle tracking. Our studies suggest an airway mucus gel layer must be produced with virus-sized pores to physically constrain IAV. While sialic acid binding by IAV may improve mucus trapping efficiency, sialic acid binding preference is found to have little impact on IAV mobility and the fraction of viral particles expected to penetrate the mucus barrier. Further, synthetic polymeric hydrogels engineered with mucus-like architecture are similarly protective against IAV infection despite their lack of sialic acid decoy receptors. Together, this work provides new insights on mucus barrier function toward IAV with important implications on innate host defense and transmission of respiratory viruses.
Collapse
Affiliation(s)
- Logan Kaler
- Biophysics Program, University of Maryland, College Park, MD, 20742, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Elizabeth M Engle
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Molecular and Cellular Biology Program, University of Maryland, College Park, MD, 20742, USA
| | - Maria Corkran
- Molecular and Cellular Biology Program, University of Maryland, College Park, MD, 20742, USA
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD, 20742, USA
| | - Ethan Iverson
- Molecular and Cellular Biology Program, University of Maryland, College Park, MD, 20742, USA
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD, 20742, USA
| | - Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Maxinne A Ignacio
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD, 20742, USA
| | - Taj Yeruva
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Margaret A Scull
- Molecular and Cellular Biology Program, University of Maryland, College Park, MD, 20742, USA
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD, 20742, USA
| | - Gregg A Duncan
- Biophysics Program, University of Maryland, College Park, MD, 20742, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Molecular and Cellular Biology Program, University of Maryland, College Park, MD, 20742, USA
| |
Collapse
|
2
|
Papamentzelopoulou M, Pitiriga VC. Unlocking the Interactions Between the Whole-Body Microbiome and HPV Infection: A Literature Review. Pathogens 2025; 14:293. [PMID: 40137778 PMCID: PMC11945791 DOI: 10.3390/pathogens14030293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
The human microbiome plays a vital role in maintaining human homeostasis, acting as a key regulator of host immunity and defense mechanisms. However, dysbiotic microbial communities may cause disruption of the symbiotic relationship between the host and the local microbiota, leading to the pathogenesis of various diseases, including viral infections and cancers. One of the most common infectious agents causing cancer is the human papilloma virus (HPV), which accounts for more than 90% of cervical cancers. In most cases, the host immune system is activated and clears HPV, whereas in some cases, the infection persists and can lead to precancerous lesions. Over the last two decades, the advent of next-generation sequencing (NGS) technology and bioinformatics has allowed a thorough and in-depth analysis of the microbial composition in various anatomical niches, allowing researchers to unveil the interactions and the underlying mechanisms through which the human microbiota could affect HPV infection establishment, persistence, and progression. Accordingly, the present narrative review aims to shed light on our understanding of the role of the human microbiome in the context of HPV infection and its progression, mainly to cervical cancer. Furthermore, we explore the mechanisms by which the composition and balance of microbial communities exert potential pathogenic or protective effects, leading to either HPV persistence and disease outcomes or clearance. Special interest is given to how the microbiome can modulate host immunity to HPV infection. Lastly, we summarize the latest findings on the therapeutic efficacy of probiotics and prebiotics in preventing and/or treating HPV infections and the potential of vaginal microbiota transplantation while highlighting the significance of personalized medicine approaches emerging from NGS-based microbiome profiling and artificial intelligence (AI) for the optimal management of HPV-related diseases.
Collapse
Affiliation(s)
- Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassiliki C. Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| |
Collapse
|
3
|
Chandna S, Povolotsky TL, Nie C, Schwartz S, Wedepohl S, Quaas E, Ludwig K, Boyakova Y, Bhatia S, Meyer K, Falkenhagen J, Haag R, Block S. Lignin-Based Mucus-Mimicking Antiviral Hydrogels with Enzyme Stability and Tunable Porosity. ACS APPLIED MATERIALS & INTERFACES 2025; 17:8962-8975. [PMID: 39876589 PMCID: PMC11826508 DOI: 10.1021/acsami.4c18519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Mucus is a complex hydrogel that acts as a defensive and protective barrier in various parts of the human body. The rise in the level of viral infections has underscored the importance of advancing research into mucus-mimicking hydrogels for the efficient design of antiviral agents. Herein, we demonstrate the gram-scale synthesis of biocompatible, lignin-based virus-binding inhibitors that reduce waste and ensure long-term availability. The lignin-based inhibitors are equipped with sulfate moieties, which are known binding partners for many viruses, including SARS-CoV-2 and herpes viruses. In addition, cross-linking the synthesized inhibitors yielded hydrogels that mimicked native mucus concerning surface functionality and rheology. The degree of sulfation exhibits a very strong impact on the mesh size distribution of the hydrogels, which provides a new means to fine-tune the steric and electrostatic contributions of the virus-hydrogel interaction. This feature strongly impacts the sequestration capability of the lignin-based hydrogels, which is demonstrated by infection inhibition assays involving human herpes simplex virus 1, influenza A viruses, and the bacterium Escherichia coli (E. coli). These measurements showed a reduction in plaque-forming units (HSV-1) and colony-forming units (E. coli) by more than 4 orders of magnitude, indicating the potent inhibition by the lignin-based hydrogels.
Collapse
Affiliation(s)
- Sanjam Chandna
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Tatyana L. Povolotsky
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Chuanxiong Nie
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Sophia Schwartz
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Stefanie Wedepohl
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Elisa Quaas
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Kai Ludwig
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Yulia Boyakova
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Sumati Bhatia
- Faculty
of
Science and Engineering, Department of Chemistry, Swansea University, Singleton Campus,
Swansea, Swansea SA28PP, U.K.
| | - Klas Meyer
- Federal
Institute for Materials Research and Testing (Bundesanstalt für
Materialforschung und -prüfung), Berlin 12489, Germany
| | - Jana Falkenhagen
- Federal
Institute for Materials Research and Testing (Bundesanstalt für
Materialforschung und -prüfung), Berlin 12489, Germany
| | - Rainer Haag
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| | - Stephan Block
- Institute
for Chemistry and Biochemistry, Freie Universität
Berlin, Berlin 14195, Germany
| |
Collapse
|
4
|
Powell DJ, Li D, Smith B, Chen WN. Cultivated meat microbiological safety considerations and practices. Compr Rev Food Sci Food Saf 2025; 24:e70077. [PMID: 39731713 DOI: 10.1111/1541-4337.70077] [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: 06/03/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 12/30/2024]
Abstract
Cultivated meat, produced using cell culture technology, is an alternative to conventional meat production that avoids the risks from enteric pathogens associated with animal slaughter and processing. Cultivated meat therefore has significant theoretical microbiological safety advantages, though limited information is available to validate this. This review discusses sources and vectors of microbial contamination throughout cultivated meat production, introduces industry survey data to evaluate current industry practices for monitoring and mitigating these hazards, and highlights future research needs. Industry survey respondents reported an average microbiological contamination batch failure rate of 11.2%. The most common vectors were related to personnel, equipment, and the production environment, while the most commonly reported type of microbiological contaminant was bacteria. These will likely remain prominent vectors and source organisms in commercial-scale production but can be addressed by a modified combination of existing commercial food and biopharmaceutical production safety systems such as Hazard Analysis and Critical Control Points (HACCP), Good Manufacturing Practices (GMP), and Good Cell Culture Practice (GCCP). As the sector matures and embeds these and other safety management systems, microbiological contamination issues should be surmountable. Data are also included to investigate whether the limited microbiome of cultivated products poses a novel food safety risk. However, further studies are needed to assess the growth potential of microorganisms in different cultivated meat products, taking into account factors such as their composition, pH, water activity, and background microflora.
Collapse
Affiliation(s)
- Dean Joel Powell
- The Good Food Institute Asia Pacific (GFI APAC), Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Dan Li
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
- Bezos Center for Sustainable Protein, National University of Singapore, Singapore, Singapore
| | - Ben Smith
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Wei Ning Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Future Ready Food Safety Hub (FRESH), Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
5
|
Yang N, Li Y, Cai Y, Liu Y, Zhang Y, Fu Y, Tan C, Willems L, Liu G. A mucus layer derived from porcine intestinal organoid air-liquid interface monolayer attenuates swine enteric coronavirus infection by antiviral activity of Muc2. BMC Biol 2024; 22:297. [PMID: 39716180 DOI: 10.1186/s12915-024-02094-7] [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: 05/08/2023] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND The mucus layer provides the first defense that keeps the epithelium free from microorganisms. However, the effect of the small intestinal mucus layer on pathogen invasion is still poorly understood, especially for swine enteric coronavirus. To better understand virus‒mucus layer‒intestinal epithelium interactions, here, we developed a porcine intestinal organoid mucus‒monolayer model under air‒liquid interface (ALI) conditions. RESULTS We successfully established a differentiated intestinal organoid monolayer model comprising various differentiated epithelial cell types and a mucus layer under ALI conditions. Mass spectrometry analysis revealed that the mucus derived from the ALI monolayer shared a similar composition to that of the native small intestinal mucus. Importantly, our results demonstrated that the ALI monolayer exhibited lower infectivity of both TGEV and PEDV than did the submerged monolayer. To further confirm the impact of ALI mucus on coronavirus infection, mucus was collected from the ALI monolayer culture system and incubated with the viruses. These results indicated that ALI mucus treatment effectively reduced the infectivity of TGEV and PEDV. Additionally, Mucin 2 (Muc2), a major component of native small intestinal mucus, was found to be abundant in the mucus derived from the ALI monolayer, as determined by mass spectrometry analysis. Our study confirmed the potent antiviral activity of Muc2 against TGEV and PEDV infection. Considering the sialylation of Muc2 and the known sialic acid-binding activity of coronavirus, further investigations revealed that the sialic acid residues of Muc2 play a potential role in inhibiting coronavirus infection. CONCLUSIONS We established the porcine intestinal organoid mucus monolayer as a novel and valuable model for confirming the pivotal role of the small intestinal mucus layer in combating pathogen invasion. In addition, our findings highlight the significance of sialic acid modification of Muc2 in blocking coronavirus infections. This discovery opens promising avenues for the development of tailor-made drugs aimed at preventing porcine enteric coronavirus invasion.
Collapse
Affiliation(s)
- Ning Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liège, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yifei Cai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- Nutritional Biology, Wageningen University and Research, Wageningen, The Netherlands
| | - Yuanyuan Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yunhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liège, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yuguang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chen Tan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liège, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Luc Willems
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liège, Belgium
| | - Guangliang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China.
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.
| |
Collapse
|
6
|
Ortega-Prieto AM, Jimenez-Guardeño JM. Interferon-stimulated genes and their antiviral activity against SARS-CoV-2. mBio 2024; 15:e0210024. [PMID: 39171921 PMCID: PMC11389394 DOI: 10.1128/mbio.02100-24] [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] [Indexed: 08/23/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic remains an international health problem caused by the recent emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As of May 2024, SARS-CoV-2 has caused more than 775 million cases and over 7 million deaths globally. Despite current vaccination programs, infections are still rapidly increasing, mainly due to the appearance and spread of new variants, variations in immunization rates, and limitations of current vaccines in preventing transmission. This underscores the need for pan-variant antivirals and treatments. The interferon (IFN) system is a critical element of the innate immune response and serves as a frontline defense against viruses. It induces a generalized antiviral state by transiently upregulating hundreds of IFN-stimulated genes (ISGs). To gain a deeper comprehension of the innate immune response to SARS-CoV-2, its connection to COVID-19 pathogenesis, and the potential therapeutic implications, this review provides a detailed overview of fundamental aspects of the diverse ISGs identified for their antiviral properties against SARS-CoV-2. It emphasizes the importance of these proteins in controlling viral replication and spread. Furthermore, we explore methodological approaches for the identification of ISGs and conduct a comparative analysis with other viruses. Deciphering the roles of ISGs and their interactions with viral pathogens can help identify novel targets for antiviral therapies and enhance our preparedness to confront current and future viral threats.
Collapse
Affiliation(s)
- Ana Maria Ortega-Prieto
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | - Jose M Jimenez-Guardeño
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
7
|
Giedraitis E, Neve RL, Phelan VV. Iron content of commercial mucin contributes to compositional stability of a cystic fibrosis airway synthetic microbiota community. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611695. [PMID: 39282275 PMCID: PMC11398496 DOI: 10.1101/2024.09.06.611695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
In vitro culture models of mucosal environments are used to elucidate the mechanistic roles of the microbiota in human health. These models often include commercial mucins to reflect the in-situ role of mucins as an attachment site and nutrient source for the microbiota. Two types of mucins are commercially available: porcine gastric mucin (PGM) and bovine submaxillary mucin (BSM). These commercial mucins have been shown to contain iron, an essential element required by the microbiota as a co-factor for a variety of metabolic functions. In these mucin preparations, the concentration of available iron can exceed physiological concentrations present in the native environment. This unexpected source of iron influences experimental outcomes, including shaping the interactions between co-existing microbes in synthetic microbial communities used to elucidate the multispecies interactions within native microbiota. In this work, we leveraged the well-characterized iron-dependent production of secondary metabolites by the opportunistic pathogen Pseudomonas aeruginosa to aid in the development of a simple, low-cost, reproducible workflow to remove iron from commercial mucins. Using the mucosal environment of the cystic fibrosis (CF) airway as a model system, we show that P. aeruginosa is canonically responsive to iron concentration in the chemically defined synthetic CF medium complemented with semi-purified PGM, and community composition of a clinically relevant, synthetic CF airway microbial community is modulated, in part, by iron concentration in PGM.
Collapse
Affiliation(s)
- Emily Giedraitis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rachel L. Neve
- Department of Immunology and Microbiology, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Vanessa V. Phelan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
8
|
Xue M, Deng A, Wang JN, Mi X, Lao Z, Yang Y. A Zanamivir-protein conjugate mimicking mucin for trapping influenza virion particles and inhibiting neuraminidase activity. Int J Biol Macromol 2024; 275:133564. [PMID: 38955298 DOI: 10.1016/j.ijbiomac.2024.133564] [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: 04/08/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Influenza viruses contribute significantly to the global health burden, necessitating the development of strategies against transmission as well as effective antiviral treatments. The present study reports a biomimetic strategy inspired by the natural antiviral properties of mucins. A bovine serum albumin (BSA) conjugate decorated with the multivalent neuraminidase inhibitor Zanamivir (ZA-BSA) was synthesized using copper-free click chemistry. This synthetic pseudo-mucin exhibited potent neuraminidase inhibitory activity against several influenza strains. Virus capture and growth inhibition assays demonstrated its effective absorption of virion particles and ability to prevent viral infection in nanomolar concentrations. Investigation of the underlying antiviral mechanism of ZA-BSA revealed a dual mode of action, involving disruption of the initial stages of host-cell binding and fusion by inducing viral aggregation, followed by blocking the release of newly assembled virions by targeting neuraminidase activity. Notably, the conjugate also exhibited potent inhibitory activity against Oseltamivir-resistant neuraminidase variant comparable to the monomeric Zanamivir. These findings highlight the application of multivalent drug presentation on protein scaffold to mimic mucin adsorption of viruses, together with counteracting drug resistance. This innovative approach has potential for the creation of antiviral agents against influenza and other viral infections.
Collapse
Affiliation(s)
- Mingming Xue
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Ang Deng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Jia-Ning Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xue Mi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Zhiqi Lao
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Yang Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China.
| |
Collapse
|
9
|
Tafech B, Rokhforouz MR, Leung J, Sung MM, Lin PJ, Sin DD, Lauster D, Block S, Quon BS, Tam Y, Cullis P, Feng JJ, Hedtrich S. Exploring Mechanisms of Lipid Nanoparticle-Mucus Interactions in Healthy and Cystic Fibrosis Conditions. Adv Healthc Mater 2024; 13:e2304525. [PMID: 38563726 DOI: 10.1002/adhm.202304525] [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: 12/19/2023] [Revised: 03/05/2024] [Indexed: 04/04/2024]
Abstract
Mucus forms the first defense line of human lungs, and as such hampers the efficient delivery of therapeutics to the underlying epithelium. This holds particularly true for genetic cargo such as CRISPR-based gene editing tools which cannot readily surmount the mucosal barrier. While lipid nanoparticles (LNPs) emerge as versatile non-viral gene delivery systems that can help overcome the delivery challenge, many knowledge gaps remain, especially for diseased states such as cystic fibrosis (CF). This study provides fundamental insights into Cas9 mRNA or ribonucleoprotein-loaded LNP-mucus interactions in healthy and diseased states by assessing the impact of the genetic cargo, mucin sialylation, mucin concentration, ionic strength, pH, and polyethylene glycol (PEG) concentration and nature on LNP diffusivity leveraging experimental approaches and Brownian dynamics (BD) simulations. Taken together, this study identifies key mucus and LNP characteristics that are critical to enabling a rational LNP design for transmucosal delivery.
Collapse
Affiliation(s)
- Belal Tafech
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Mohammad-Reza Rokhforouz
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Jerry Leung
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Molly Mh Sung
- Acuitas Therapeutics, Vancouver, BC, V6T 1Z3, Canada
| | - Paulo Jc Lin
- Acuitas Therapeutics, Vancouver, BC, V6T 1Z3, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Daniel Lauster
- Institute of Pharmacy, Biopharmaceuticals, Freie Universität Berlin, 12169, Berlin, Germany
| | - Stephan Block
- Institute of Organic Chemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Bradley S Quon
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
- Adult Cystic Fibrosis Clinic, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, V6T 1Z3, Canada
| | - Pieter Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - James J Feng
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Department of Mathematics, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
- Center of Biological Design, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| |
Collapse
|
10
|
Kaler L, Engle EM, Iverson E, Boboltz A, Ignacio MA, Rife M, Scull MA, Duncan G. Mucus physically restricts influenza A viral particle access to the epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.14.553271. [PMID: 37645821 PMCID: PMC10462089 DOI: 10.1101/2023.08.14.553271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Prior work suggests influenza A virus (IAV) crosses the airway mucus barrier in a sialic acid-dependent manner through the actions of the viral envelope proteins, hemagglutinin and neuraminidase. However, host and viral factors that influence how efficiently mucus traps IAV remain poorly defined. In this work, we assessed how the physicochemical properties of mucus influence its ability to effectively capture IAV with altered sialic acid preference using fluorescence video microscopy and multiple particle tracking. We found an airway mucus gel layer must be produced with pores on the order of size of the virus to physically constrain IAV. Sialic acid binding by IAV also improves mucus trapping efficiency, but interestingly, sialic acid preferences had little impact on the fraction of IAV particles expected to penetrate the mucus barrier. Together, this work provides new insights on mucus barrier function toward IAV with important implications on innate host defense and interspecies transmission.
Collapse
|
11
|
Al Kindi A, Courelli NS, Ogbonna K, Urueña JM, Chau AL, Pitenis AA. Bioinspired Lubricity from Surface Gel Layers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:9926-9933. [PMID: 38683632 PMCID: PMC11100014 DOI: 10.1021/acs.langmuir.3c03686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
Surface gel layers on commercially available contact lenses have been shown to reduce frictional shear stresses and mitigate damage during sliding contact with fragile epithelial cell layers in vitro. Spencer and co-workers recently demonstrated that surface gel layers could arise from oxygen-inhibited free-radical polymerization. In this study, polyacrylamide hydrogel shell probes (7.5 wt % acrylamide, 0.3 wt % N,N'-methylenebisacrylamide) were polymerized in three hemispherical molds listed in order of decreasing surface energy and increasing oxygen permeability: borosilicate glass, polyether ether ketone (PEEK), and polytetrafluoroethylene (PTFE). Hydrogel probes polymerized in PEEK and PTFE molds exhibited 100× lower elastic moduli at the surface (E PEEK * = 80 ± 31 and E PTFE * = 106 ± 26 Pa, respectively) than those polymerized in glass molds (E glass * = 31,560 ± 1,570 Pa), in agreement with previous investigations by Spencer and co-workers. Biotribological experiments revealed that hydrogel probes with surface gel layers reduced frictional shear stresses against cells (τPEEK = 35 ± 15 and τPTFE = 22 ± 16 Pa) more than those without (τglass = 68 ± 15 Pa) and offered greater protection against cell damage when sliding against human telomerase-immortalized corneal epithelial (hTCEpi) cell monolayers. Our work demonstrates that the "mold effect" resulting in oxygen-inhibition polymerization creates hydrogels with surface gel layers that reduce shear stresses in sliding contact with cell monolayers, similar to the protection offered by gradient mucin gel networks across epithelial cell layers.
Collapse
Affiliation(s)
- Ahmed Al Kindi
- Department
of Mechanical Engineering, University of
California, Santa
Barbara, California 93106, United States
| | - Nemea S. Courelli
- Department
of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Kevin Ogbonna
- College
of Creative Studies, Biological Sciences, University of California, Santa
Barbara, California 93106, United States
| | - Juan Manuel Urueña
- NSF
BioPACIFIC Materials Innovation Platform, University of California, Santa
Barbara, California 93106, United States
| | - Allison L. Chau
- Materials
Department, University of California, Santa Barbara, California 93106, United States
| | - Angela A. Pitenis
- Materials
Department, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
12
|
Walsh D, Bevan J, Harrison F. How Does Airway Surface Liquid Composition Vary in Different Pulmonary Diseases, and How Can We Use This Knowledge to Model Microbial Infections? Microorganisms 2024; 12:732. [PMID: 38674677 PMCID: PMC11052052 DOI: 10.3390/microorganisms12040732] [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: 03/11/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Growth environment greatly alters many facets of pathogen physiology, including pathogenesis and antimicrobial tolerance. The importance of host-mimicking environments for attaining an accurate picture of pathogen behaviour is widely recognised. Whilst this recognition has translated into the extensive development of artificial cystic fibrosis (CF) sputum medium, attempts to mimic the growth environment in other respiratory disease states have been completely neglected. The composition of the airway surface liquid (ASL) in different pulmonary diseases is far less well characterised than CF sputum, making it very difficult for researchers to model these infection environments. In this review, we discuss the components of human ASL, how different lung pathologies affect ASL composition, and how different pathogens interact with these components. This will provide researchers interested in mimicking different respiratory environments with the information necessary to design a host-mimicking medium, allowing for better understanding of how to treat pathogens causing infection in these environments.
Collapse
Affiliation(s)
- Dean Walsh
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK (F.H.)
| | | | | |
Collapse
|
13
|
Henkel M, Kimna C, Lieleg O. DNA Crosslinked Mucin Hydrogels Allow for On-Demand Gel Disintegration and Triggered Particle Release. Macromol Biosci 2024; 24:e2300427. [PMID: 38217373 DOI: 10.1002/mabi.202300427] [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: 09/19/2023] [Revised: 12/04/2023] [Indexed: 01/15/2024]
Abstract
Whereas hydrogels created from synthetic polymers offer a high level of control over their stability and mechanical properties, their biomedical activity is typically limited. In contrast, biopolymers have evolved over billions of years to integrate a broad range of functionalities into a single design. Thus, biopolymeric hydrogels can show remarkable capabilities such as regulatory behavior, selective barrier properties, or antimicrobial effects. Still, despite their widespread use in numerous biomedical applications, achieving a meticulous control over the physical properties of macroscopic biopolymeric networks remains a challenge. Here, a macroscopic, DNA-crosslinked mucin hydrogel with tunable viscoelastic properties that responds to two types of triggers: temperature alterations and DNA displacement strands, is presented. As confirmed with bulk rheology and single particle tracking, the hybridized base pairs governing the stability of the hydrogel can be opened, thus allowing for a precise control over the hydrogel stiffness and even enabling a full gel-to-sol transition. As those DNA-crosslinked mucin hydrogels possess tunable mechanical properties and can be disintegrated on demand, they can not only be considered for controlled cargo release but may also serve as a role model for the development of smart biomedical materials in applications such as tissue engineering and wound healing.
Collapse
Affiliation(s)
- Manuel Henkel
- School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Boltzmannstraße 15, 85748, Garching, Germany
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering, Technical University of Munich, Ernst-Otto-Fischer Straße 8, 85748, Garching, Germany
| | - Ceren Kimna
- School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Boltzmannstraße 15, 85748, Garching, Germany
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering, Technical University of Munich, Ernst-Otto-Fischer Straße 8, 85748, Garching, Germany
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Boltzmannstraße 15, 85748, Garching, Germany
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering, Technical University of Munich, Ernst-Otto-Fischer Straße 8, 85748, Garching, Germany
| |
Collapse
|
14
|
McCoy R, Oldroyd S, Yang W, Wang K, Hoven D, Bulmer D, Zilbauer M, Owens RM. In Vitro Models for Investigating Intestinal Host-Pathogen Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306727. [PMID: 38155358 PMCID: PMC10885678 DOI: 10.1002/advs.202306727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Indexed: 12/30/2023]
Abstract
Infectious diseases are increasingly recognized as a major threat worldwide due to the rise of antimicrobial resistance and the emergence of novel pathogens. In vitro models that can adequately mimic in vivo gastrointestinal physiology are in high demand to elucidate mechanisms behind pathogen infectivity, and to aid the design of effective preventive and therapeutic interventions. There exists a trade-off between simple and high throughput models and those that are more complex and physiologically relevant. The complexity of the model used shall be guided by the biological question to be addressed. This review provides an overview of the structure and function of the intestine and the models that are developed to emulate this. Conventional models are discussed in addition to emerging models which employ engineering principles to equip them with necessary advanced monitoring capabilities for intestinal host-pathogen interrogation. Limitations of current models and future perspectives on the field are presented.
Collapse
Affiliation(s)
- Reece McCoy
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sophie Oldroyd
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Woojin Yang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Kaixin Wang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Darius Hoven
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - David Bulmer
- Department of PharmacologyUniversity of CambridgeCambridgeCB2 1PDUK
| | - Matthias Zilbauer
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| |
Collapse
|
15
|
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
|
16
|
Lao Z, Li Y, Mi X, Tang Q, Li J, Chen Y, Yang Y. Synthetic pentatrideca-valent triazolylsialoside inhibits influenza virus hemagglutinin/neuraminidase and aggregates virion particles. Eur J Med Chem 2023; 259:115578. [PMID: 37467617 DOI: 10.1016/j.ejmech.2023.115578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
A synthetic multivalent hemagglutinin and neuraminidase inhibitor was developed by the conjugation of a septa-valent triazolylsialoside to bovine serum albumin using di-(N-succinimidyl) adipate. Matrixassisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) confirmed the attachment of five septa-valent sialyl lactosides to the protein backbone, resulting in a pentatrideca-valent sialyl conjugate. This pseudo-glycoprotein demonstrated a high affinity for hemagglutinin/neuraminidase as well as for the drug-resistant NA mutation on the influenza virus surface due to the cluster effect. The conjugate also exhibited potent antiviral activity against a wide range of virus strains without cytotoxicity at high concentrations. Mechanistic studies revealed that the pentatrideca-valent sialyl conjugate bound strongly to the influenza virion particles through interactions with HA/NA on the virion surfaces. The KD of the interaction was approximately 1 μM, as determined by isothermal calorimetric titration, allowing the capture and trapping of the influenza virions and preventing their further infection of host cells. These findings provide insight into the development of new antiviral agents using multivalent sialic acid clusters.
Collapse
Affiliation(s)
- Zhiqi Lao
- Department of Medical Laboratory, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yang Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Xue Mi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Qi Tang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Jie Li
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200011, China.
| | - Yue Chen
- Department of Medical Laboratory, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, China.
| | - Yang Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China.
| |
Collapse
|
17
|
Rickert CA, Mansi S, Fan D, Mela P, Lieleg O. A Mucin-Based Bio-Ink for 3D Printing of Objects with Anti-Biofouling Properties. Macromol Biosci 2023; 23:e2300198. [PMID: 37466113 DOI: 10.1002/mabi.202300198] [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: 05/08/2023] [Revised: 07/10/2023] [Accepted: 07/16/2023] [Indexed: 07/20/2023]
Abstract
With its potential to revolutionize the field of personalized medicine by producing customized medical devices and constructs for tissue engineering at low costs, 3D printing has emerged as a highly promising technology. Recent advancements have sparked increasing interest in the printing of biopolymeric hydrogels. However, owing to the limited printability of those soft materials, the lack of variability in available bio-inks remains a major challenge. In this study, a novel bio-ink is developed based on functionalized mucin-a glycoprotein that exhibits a multitude of biomedically interesting properties such as immunomodulating activity and strong anti-biofouling behavior. To achieve sufficient printability of the mucin-based ink, its rheological properties are tuned by incorporating Laponite XLG as a stabilizing agent. It is shown that cured objects generated from this novel bio-ink exhibit mechanical properties partially similar to that of soft tissue, show strong anti-biofouling properties, good biocompatibility, tunable cell adhesion, and immunomodulating behavior. The presented findings suggest that this 3D printable bio-ink has a great potential for a wide range of biomedical applications, including tissue engineering, wound healing, and soft robotics.
Collapse
Affiliation(s)
- Carolin A Rickert
- TUM School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Boltzmannstr. 15, 85748, Garching b. München, Germany
- Center for Functional Protein Assemblies (CPA), Technical University of Munich, Ernst-Otto-Fischer Str. 8, 85748, Garching b. München, Germany
| | - Salma Mansi
- TUM School of Engineering and Design, Department of Mechanical Engineering, Chair of Medical Materials and Implants, Technical University of Munich, Boltzmannstr. 15, 85748, Garching b. München, Germany
- Munich Institute of Biomedical Engineering and Munich Institute of Integrated Materials, Energy and Process Engineering, Technical University of Munich, Boltzmannstr. 15, 85748, Garching, Germany
| | - Di Fan
- TUM School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Boltzmannstr. 15, 85748, Garching b. München, Germany
- Center for Functional Protein Assemblies (CPA), Technical University of Munich, Ernst-Otto-Fischer Str. 8, 85748, Garching b. München, Germany
| | - Petra Mela
- TUM School of Engineering and Design, Department of Mechanical Engineering, Chair of Medical Materials and Implants, Technical University of Munich, Boltzmannstr. 15, 85748, Garching b. München, Germany
- Munich Institute of Biomedical Engineering and Munich Institute of Integrated Materials, Energy and Process Engineering, Technical University of Munich, Boltzmannstr. 15, 85748, Garching, Germany
| | - Oliver Lieleg
- TUM School of Engineering and Design, Department of Materials Engineering, Technical University of Munich, Boltzmannstr. 15, 85748, Garching b. München, Germany
- Center for Functional Protein Assemblies (CPA), Technical University of Munich, Ernst-Otto-Fischer Str. 8, 85748, Garching b. München, Germany
| |
Collapse
|
18
|
Martínez-Oca P, Alba C, Sánchez-Roncero A, Fernández-Marcelo T, Martín MÁ, Escrivá F, Rodríguez JM, Álvarez C, Fernández-Millán E. Maternal Diet Determines Milk Microbiome Composition and Offspring Gut Colonization in Wistar Rats. Nutrients 2023; 15:4322. [PMID: 37892398 PMCID: PMC10609248 DOI: 10.3390/nu15204322] [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: 09/14/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Mother's milk contains a unique microbiome that plays a relevant role in offspring health. We hypothesize that maternal malnutrition during lactation might impact the microbial composition of milk and affect adequate offspring gut colonization, increasing the risk for later onset diseases. Then, Wistar rats were fed ad libitum (Control, C) food restriction (Undernourished, U) during gestation and lactation. After birth, offspring feces and milk stomach content were collected at lactating day (L)4, L14 and L18. The V3-V4 region of the bacterial 16S rRNA gene was sequenced to characterize bacterial communities. An analysis of beta diversity revealed significant disparities in microbial composition between groups of diet at L4 and L18 in both milk, and fecal samples. In total, 24 phyla were identified in milk and 18 were identified in feces, with Firmicutes, Proteobacteria, Actinobacteroidota and Bacteroidota collectively representing 96.1% and 97.4% of those identified, respectively. A higher abundance of Pasteurellaceae and Porphyromonas at L4, and of Gemella and Enterococcus at L18 were registered in milk samples from the U group. Lactobacillus was also significantly more abundant in fecal samples of the U group at L4. These microbial changes compromised the number and variety of milk-feces or feces-feces bacterial correlations. Moreover, increased offspring gut permeability and an altered expression of goblet cell markers TFF3 and KLF3 were observed in U pups. Our results suggest that altered microbial communication between mother and offspring through breastfeeding may explain, in part, the detrimental consequences of maternal malnutrition on offspring programming.
Collapse
Affiliation(s)
- Paula Martínez-Oca
- Instituto de Investigación en Ciencias de la Alimentación (CIAL), Campus de Excelencia Científica, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
| | - Claudio Alba
- Department of Nutrition and Food Science, Faculty of Veterinary Sciences, University Complutense of Madrid, 28040 Madrid, Spain; (C.A.); (J.M.R.)
| | - Alicia Sánchez-Roncero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.S.-R.); (F.E.); (C.Á.)
| | - Tamara Fernández-Marcelo
- Centro de Investigación Biomédica en Red (CIBERDEM), ISCIII, 28029 Madrid, Spain; (T.F.-M.); (M.Á.M.)
| | - María Ángeles Martín
- Centro de Investigación Biomédica en Red (CIBERDEM), ISCIII, 28029 Madrid, Spain; (T.F.-M.); (M.Á.M.)
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Fernando Escrivá
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.S.-R.); (F.E.); (C.Á.)
- Centro de Investigación Biomédica en Red (CIBERDEM), ISCIII, 28029 Madrid, Spain; (T.F.-M.); (M.Á.M.)
| | - Juan Miguel Rodríguez
- Department of Nutrition and Food Science, Faculty of Veterinary Sciences, University Complutense of Madrid, 28040 Madrid, Spain; (C.A.); (J.M.R.)
| | - Carmen Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.S.-R.); (F.E.); (C.Á.)
- Centro de Investigación Biomédica en Red (CIBERDEM), ISCIII, 28029 Madrid, Spain; (T.F.-M.); (M.Á.M.)
| | - Elisa Fernández-Millán
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.S.-R.); (F.E.); (C.Á.)
- Centro de Investigación Biomédica en Red (CIBERDEM), ISCIII, 28029 Madrid, Spain; (T.F.-M.); (M.Á.M.)
| |
Collapse
|
19
|
Yang S, Duncan GA. Synthetic mucus biomaterials for antimicrobial peptide delivery. J Biomed Mater Res A 2023; 111:1616-1626. [PMID: 37199137 PMCID: PMC10524183 DOI: 10.1002/jbm.a.37559] [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: 03/07/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
Despite the promise of antimicrobial peptides (AMPs) as treatments for antibiotic-resistant infections, their therapeutic efficacy is limited due to the rapid degradation and low bioavailability of AMPs. To address this, we have developed and characterized a synthetic mucus (SM) biomaterial capable of delivering LL37 AMPs and enhancing their therapeutic effect. LL37 is an AMP that exhibits a wide range of antimicrobial activity against bacteria, including Pseudomonas aeruginosa. LL37 loaded SM hydrogels demonstrated controlled release with 70%-95% of loaded LL37 over 8 h due to charge-mediated interactions between mucins and LL37 AMPs. Compared to treatment with LL37 alone where antimicrobial activity was reduced after 3 h, LL37-SM hydrogels inhibited P. aeruginosa (PAO1) growth over 12 h. LL37-SM hydrogel treatment reduced PAO1 viability over 6 h whereas a rebound in bacterial growth was observed when treated with LL37 only. These data demonstrate LL37-SM hydrogels enhance antimicrobial activity by preserving LL37 AMP activity and bioavailability. Overall, this work establishes SM biomaterials as a platform for enhanced AMP delivery for antimicrobial applications.
Collapse
Affiliation(s)
- Sydney Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
20
|
Bustos NA, Ribbeck K, Wagner CE. The role of mucosal barriers in disease progression and transmission. Adv Drug Deliv Rev 2023; 200:115008. [PMID: 37442240 DOI: 10.1016/j.addr.2023.115008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/22/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Mucus is a biological hydrogel that coats and protects all non-keratinized wet epithelial surfaces. Mucins, the primary structural components of mucus, are critical components of the gel layer that protect against invading pathogens. For communicable diseases, pathogen-mucin interactions contribute to the pathogen's fate and the potential for disease progression in-host, as well as the potential for onward transmission. We begin by reviewing in-host mucus filtering mechanisms, including size filtering and interaction filtering, which regulate the permeability of mucus barriers to all molecules including pathogens. Next, we discuss the role of mucins in communicable diseases at the point of transmission (i.e. how the encapsulation of pathogens in emitted mucosal droplets externally to hosts may modulate pathogen infectivity and viability). Overall, mucosal barriers modulate both host susceptibility as well as the dynamics of population-level disease transmission. The study of mucins and their use in models and experimental systems are therefore crucial for understanding the mechanistic biophysical principles underlying disease transmission and the early stages of host infection.
Collapse
Affiliation(s)
- Nicole A Bustos
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline E Wagner
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
21
|
Arruda BL, Kanefsky RA, Hau S, Janzen GM, Anderson TK, Vincent Baker AL. Mucin 4 is a cellular biomarker of necrotizing bronchiolitis in influenza A virus infection. Microbes Infect 2023; 25:105169. [PMID: 37295769 DOI: 10.1016/j.micinf.2023.105169] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Influenza A virus (IAV) in the human and swine host infects epithelial cells lining the respiratory tract causing a necrotizing bronchitis and bronchiolitis. These epithelial surfaces are protected by large glycoproteins called mucins. Mucin 4 (MUC4) is a transmembrane mucin that consists of an alpha subunit responsible for surface protection and intracellular beta subunit involved in signal transduction which repress apoptosis and stimulate epithelial proliferation. This study was designed to determine the expression and potential role of MUC4 during IAV infection. We used immunohistochemistry in combination with machine learning image analysis to quantify differential protein expression of MUC4 subunits in IAV-infected and uninfected lung in a porcine model. MUC4 protein basal expression in control animals varied significantly by litter. MUC4 protein expression was significantly increased in bronchioles with necrotizing bronchiolitis compared to histologically normal bronchioles, likely representing a regenerative response to restore mucosal integrity of conducting airways. Understanding the impact of differential MUC4 expression among healthy individuals and during IAV infection will facilitate control strategies by elucidating mechanisms associated with susceptibility to IAV that can be therapeutically or genetically regulated and may be extended to other respiratory diseases.
Collapse
Affiliation(s)
- Bailey L Arruda
- Virus and Prion Research Unit, National Animal Disease Center, USDA Agricultural Research Service, 1920 Dayton Ave, Ames, IA 50010, USA.
| | - Rachel A Kanefsky
- Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd, North Grafton, MA 01536, USA
| | - Samantha Hau
- Virus and Prion Research Unit, National Animal Disease Center, USDA Agricultural Research Service, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Garrett M Janzen
- Virus and Prion Research Unit, National Animal Disease Center, USDA Agricultural Research Service, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Tavis K Anderson
- Virus and Prion Research Unit, National Animal Disease Center, USDA Agricultural Research Service, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Amy L Vincent Baker
- Virus and Prion Research Unit, National Animal Disease Center, USDA Agricultural Research Service, 1920 Dayton Ave, Ames, IA 50010, USA
| |
Collapse
|
22
|
Kohout VR, Wardzala CL, Kramer JR. Mirror Image Mucins and Thio Mucins with Tunable Biodegradation. J Am Chem Soc 2023; 145:16573-16583. [PMID: 37473442 PMCID: PMC11080933 DOI: 10.1021/jacs.3c03659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Mucin glycoproteins are the major component of mucus and are integral to the cellular glycocalyx. Mucins play diverse roles in health and disease, are an important element in epithelial tissue models, and have broad therapeutic potential. All mucin applications are currently challenged by their inherent structural heterogeneity and degradation by proteases. In this study, we describe the synthesis and study of chemically defined mucin analogues bearing native glycans. We utilized combinations of enantiomer amino acids and glycan thioether linkages to achieve tunable proteolysis while maintaining cytocompatibility and binding activity. Structural characterization revealed a previously unknown mirror-image helix and sheds light on the molecular drivers of glycoprotein conformation. This work represents an important step toward the development of artificial mucins for biomedical applications.
Collapse
Affiliation(s)
- Victoria R Kohout
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Casia L Wardzala
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jessica R Kramer
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
23
|
Ejazi SA, Louisthelmy R, Maisel K. Mechanisms of Nanoparticle Transport across Intestinal Tissue: An Oral Delivery Perspective. ACS NANO 2023. [PMID: 37410891 DOI: 10.1021/acsnano.3c02403] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Oral drug administration has been a popular choice due to patient compliance and limited clinical resources. Orally delivered drugs must circumvent the harsh gastrointestinal (GI) environment to effectively enter the systemic circulation. The GI tract has a number of structural and physiological barriers that limit drug bioavailability including mucus, the tightly regulated epithelial layer, immune cells, and associated vasculature. Nanoparticles have been used to enhance oral bioavailability of drugs, as they can act as a shield to the harsh GI environment and prevent early degradation while also increasing uptake and transport of drugs across the intestinal epithelium. Evidence suggests that different nanoparticle formulations may be transported via different intracellular mechanisms to cross the intestinal epithelium. Despite the existence of a significant body of work on intestinal transport of nanoparticles, many key questions remain: What causes the poor bioavailability of the oral drugs? What factors contribute to the ability of a nanoparticle to cross different intestinal barriers? Do nanoparticle properties such as size and charge influence the type of endocytic pathways taken? In this Review, we summarize the different components of intestinal barriers and the types of nanoparticles developed for oral delivery. In particular, we focus on the various intracellular pathways used in nanoparticle internalization and nanoparticle or cargo translocation across the epithelium. Understanding the gut barrier, nanoparticle characteristics, and transport pathways may lead to the development of more therapeutically useful nanoparticles as drug carriers.
Collapse
Affiliation(s)
- Sarfaraz Ahmad Ejazi
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| | - Rebecca Louisthelmy
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| |
Collapse
|
24
|
Mei X, Li J, Wang Z, Zhu D, Huang K, Hu S, Popowski KD, Cheng K. An inhaled bioadhesive hydrogel to shield non-human primates from SARS-CoV-2 infection. NATURE MATERIALS 2023; 22:903-912. [PMID: 36759564 PMCID: PMC10615614 DOI: 10.1038/s41563-023-01475-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
The surge of fast-spreading SARS-CoV-2 mutated variants highlights the need for fast, broad-spectrum strategies to counteract viral infections. In this work, we report a physical barrier against SARS-CoV-2 infection based on an inhalable bioadhesive hydrogel, named spherical hydrogel inhalation for enhanced lung defence (SHIELD). Conveniently delivered via a dry powder inhaler, SHIELD particles form a dense hydrogel network that coats the airway, enhancing the diffusional barrier properties and restricting virus penetration. SHIELD's protective effect is first demonstrated in mice against two SARS-CoV-2 pseudo-viruses with different mutated spike proteins. Strikingly, in African green monkeys, a single SHIELD inhalation provides protection for up to 8 hours, efficiently reducing infection by the SARS-CoV-2 WA1 and B.1.617.2 (Delta) variants. Notably, SHIELD is made with food-grade materials and does not affect normal respiratory functions. This approach could offer additional protection to the population against SARS-CoV-2 and other respiratory pathogens.
Collapse
Affiliation(s)
- Xuan Mei
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Junlang Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Zhenzhen Wang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA
| | - Kristen D Popowski
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill & Raleigh, NC, USA.
| |
Collapse
|
25
|
Arnold DP, Xu Y, Takatori SC. Antibody binding reports spatial heterogeneities in cell membrane organization. Nat Commun 2023; 14:2884. [PMID: 37208326 DOI: 10.1038/s41467-023-38525-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/05/2023] [Indexed: 05/21/2023] Open
Abstract
The spatial organization of cell membrane glycoproteins and glycolipids is critical for mediating the binding of ligands, receptors, and macromolecules on the plasma membrane. However, we currently do not have the methods to quantify the spatial heterogeneities of macromolecular crowding on live cell surfaces. In this work, we combine experiment and simulation to report crowding heterogeneities on reconstituted membranes and live cell membranes with nanometer spatial resolution. By quantifying the effective binding affinity of IgG monoclonal antibodies to engineered antigen sensors, we discover sharp gradients in crowding within a few nanometers of the crowded membrane surface. Our measurements on human cancer cells support the hypothesis that raft-like membrane domains exclude bulky membrane proteins and glycoproteins. Our facile and high-throughput method to quantify spatial crowding heterogeneities on live cell membranes may facilitate monoclonal antibody design and provide a mechanistic understanding of plasma membrane biophysical organization.
Collapse
Affiliation(s)
- Daniel P Arnold
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Yaxin Xu
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Sho C Takatori
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
26
|
Kwiatkowska A, Granicka LH. Anti-Viral Surfaces in the Fight against the Spread of Coronaviruses. MEMBRANES 2023; 13:464. [PMID: 37233525 PMCID: PMC10223398 DOI: 10.3390/membranes13050464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023]
Abstract
This review is conducted against the background of nanotechnology, which provides us with a chance to effectively combat the spread of coronaviruses, and which primarily concerns polyelectrolytes and their usability for obtaining protective function against viruses and as carriers for anti-viral agents, vaccine adjuvants, and, in particular, direct anti-viral activity. This review covers nanomembranes in the form of nano-coatings or nanoparticles built of natural or synthetic polyelectrolytes--either alone or else as nanocomposites for creating an interface with viruses. There are not a wide variety of polyelectrolytes with direct activity against SARS-CoV-2, but materials that are effective in virucidal evaluations against HIV, SARS-CoV, and MERS-CoV are taken into account as potentially active against SARS-CoV-2. Developing new approaches to materials as interfaces with viruses will continue to be relevant in the future.
Collapse
Affiliation(s)
| | - Ludomira H. Granicka
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland;
| |
Collapse
|
27
|
Wu CM, Wheeler KM, Cárcamo-Oyarce G, Aoki K, McShane A, Datta SS, Mark Welch JL, Tiemeyer M, Griffen AL, Ribbeck K. Mucin glycans drive oral microbial community composition and function. NPJ Biofilms Microbiomes 2023; 9:11. [PMID: 36959210 PMCID: PMC10036478 DOI: 10.1038/s41522-023-00378-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/20/2023] [Indexed: 03/25/2023] Open
Abstract
Human microbiome composition is closely tied to health, but how the host manages its microbial inhabitants remains unclear. One important, but understudied, factor is the natural host environment: mucus, which contains gel-forming glycoproteins (mucins) that display hundreds of glycan structures with potential regulatory function. Leveraging a tractable culture-based system to study how mucins influence oral microbial communities, we found that mucin glycans enable the coexistence of diverse microbes, while resisting disease-associated compositional shifts. Mucins from tissues with unique glycosylation differentially tuned microbial composition, as did isolated mucin glycan libraries, uncovering the importance of specific glycan patterns in microbiome modulation. We found that mucins shape microbial communities in several ways: serving as nutrients to support metabolic diversity, organizing spatial structure through reduced aggregation, and possibly limiting antagonism between competing taxa. Overall, this work identifies mucin glycans as a natural host mechanism and potential therapeutic intervention to maintain healthy microbial communities.
Collapse
Affiliation(s)
- Chloe M Wu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kelsey M Wheeler
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gerardo Cárcamo-Oyarce
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Abigail McShane
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sujit S Datta
- Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | | | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Ann L Griffen
- Department of Dentistry, Nationwide Children's Hospital, Columbus, OH, USA
- Divisions of Biosciences and Pediatric Dentistry, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
28
|
Wang BX, Takagi J, McShane A, Park JH, Aoki K, Griffin C, Teschler J, Kitts G, Minzer G, Tiemeyer M, Hevey R, Yildiz F, Ribbeck K. Host-derived O-glycans inhibit toxigenic conversion by a virulence-encoding phage in Vibrio cholerae. EMBO J 2023; 42:e111562. [PMID: 36504455 PMCID: PMC9890226 DOI: 10.15252/embj.2022111562] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 12/14/2022] Open
Abstract
Pandemic and endemic strains of Vibrio cholerae arise from toxigenic conversion by the CTXφ bacteriophage, a process by which CTXφ infects nontoxigenic strains of V. cholerae. CTXφ encodes the cholera toxin, an enterotoxin responsible for the watery diarrhea associated with cholera infections. Despite the critical role of CTXφ during infections, signals that affect CTXφ-driven toxigenic conversion or expression of the CTXφ-encoded cholera toxin remain poorly characterized, particularly in the context of the gut mucosa. Here, we identify mucin polymers as potent regulators of CTXφ-driven pathogenicity in V. cholerae. Our results indicate that mucin-associated O-glycans block toxigenic conversion by CTXφ and suppress the expression of CTXφ-related virulence factors, including the toxin co-regulated pilus and cholera toxin, by interfering with the TcpP/ToxR/ToxT virulence pathway. By synthesizing individual mucin glycan structures de novo, we identify the Core 2 motif as the critical structure governing this virulence attenuation. Overall, our results highlight a novel mechanism by which mucins and their associated O-glycan structures affect CTXφ-mediated evolution and pathogenicity of V. cholerae, underscoring the potential regulatory power housed within mucus.
Collapse
Affiliation(s)
- Benjamin X Wang
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Microbiology and ImmunologyStanford UniversityStanfordCAUSA
| | - Julie Takagi
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Abigail McShane
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Jin Hwan Park
- Department of Microbiology and Environmental ToxicologyUniversity of CaliforniaSanta CruzCAUSA
| | - Kazuhiro Aoki
- Complex Carbohydrate Research CenterUniversity of GeorgiaAthensGAUSA
| | - Catherine Griffin
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Jennifer Teschler
- Department of Microbiology and Environmental ToxicologyUniversity of CaliforniaSanta CruzCAUSA
| | - Giordan Kitts
- Department of Microbiology and Environmental ToxicologyUniversity of CaliforniaSanta CruzCAUSA
| | - Giulietta Minzer
- Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
| | - Michael Tiemeyer
- Complex Carbohydrate Research CenterUniversity of GeorgiaAthensGAUSA
| | - Rachel Hevey
- Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
| | - Fitnat Yildiz
- Department of Microbiology and Environmental ToxicologyUniversity of CaliforniaSanta CruzCAUSA
| | - Katharina Ribbeck
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| |
Collapse
|
29
|
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
|
30
|
Multifunctional glycoprotein coatings improve the surface properties of highly oxygen permeable contact lenses. BIOMATERIALS ADVANCES 2023; 145:213233. [PMID: 36521413 DOI: 10.1016/j.bioadv.2022.213233] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/03/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022]
Abstract
To achieve and maintain good operability of medical devices while reducing putative side effects for the patient, a promising strategy is to tailor the surface properties of such devices as they critically dictate the tissue compatibility and the biofouling behavior. Indeed, those properties can be strongly improved by generating mucin coatings on such medical devices. However, using coatings on optical systems, e.g., contact lenses, comes with various challenges: here, the geometrical and optical characteristics of the lens may not be compromised by either the coating process or the coating itself. In this study, we show how mucin macromolecules can be attached onto the surfaces of rigid, gas permeable contact lenses while maintaining all critical lens parameters. We demonstrate that the generated coatings improve the surface wettability (contact angles are reduced from 105° to 40° and liquid film break-up times are increased from <1 s to 31 s) and prevent tribological damage to corneal tissue. Additionally, such coatings are highly transparent (transmission values above 98 % compared to an uncoated sample are reached) and efficiently reduce lipid deposition to the lens surface by 90 % but fully maintain the geometrical and mechanical properties of the lenses. Thus, such mucin coatings could also be highly beneficial for other optical systems that are used in direct contact with tissues or body fluids.
Collapse
|
31
|
Watson MM, van der Giezen M, Søreide K. Gut Microbiome Influence on Human Epigenetics, Health, and Disease. HANDBOOK OF EPIGENETICS 2023:669-686. [DOI: 10.1016/b978-0-323-91909-8.00012-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
32
|
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: 27] [Impact Index Per Article: 9.0] [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
|
33
|
Kretschmer M, Ceña‐Diez R, Butnarasu C, Silveira V, Dobryden I, Visentin S, Berglund P, Sönnerborg A, Lieleg O, Crouzier T, Yan H. Synthetic Mucin Gels with Self-Healing Properties Augment Lubricity and Inhibit HIV-1 and HSV-2 Transmission. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203898. [PMID: 36104216 PMCID: PMC9661867 DOI: 10.1002/advs.202203898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/14/2022] [Indexed: 05/02/2023]
Abstract
Mucus is a self-healing gel that lubricates the moist epithelium and provides protection against viruses by binding to viruses smaller than the gel's mesh size and removing them from the mucosal surface by active mucus turnover. As the primary nonaqueous components of mucus (≈0.2%-5%, wt/v), mucins are critical to this function because the dense arrangement of mucin glycans allows multivalence of binding. Following nature's example, bovine submaxillary mucins (BSMs) are assembled into "mucus-like" gels (5%, wt/v) by dynamic covalent crosslinking reactions. The gels exhibit transient liquefaction under high shear strain and immediate self-healing behavior. This study shows that these material properties are essential to provide lubricity. The gels efficiently reduce human immunodeficiency virus type 1 (HIV-1) and genital herpes virus type 2 (HSV-2) infectivity for various types of cells. In contrast, simple mucin solutions, which lack the structural makeup, inhibit HIV-1 significantly less and do not inhibit HSV-2. Mechanistically, the prophylaxis of HIV-1 infection by BSM gels is found to be that the gels trap HIV-1 by binding to the envelope glycoprotein gp120 and suppress cytokine production during viral exposure. Therefore, the authors believe the gels are promising for further development as personal lubricants that can limit viral transmission.
Collapse
Affiliation(s)
- Martin Kretschmer
- School of Engineering and Design, Department of Materials EngineeringTechnical University of MunichBoltzmannstrasse 1585748GarchingGermany
- Center for Protein AssembliesTechnical University of MunichErnst‐Otto‐Fischer Str. 885748GarchingGermany
| | - Rafael Ceña‐Diez
- Department of Medicine HuddingeDivision of Infectious DiseasesKarolinska University HospitalKarolinska Institutet, I73Stockholm141 86Sweden
| | - Cosmin Butnarasu
- Department of Molecular Biotechnology and Health ScienceUniversity of TurinTurin10135Italy
| | - Valentin Silveira
- Division of GlycoscienceDepartment of ChemistrySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
| | - Illia Dobryden
- Division of Bioeconomy and HealthDepartment of Material and Surface DesignRISE Research Institutes of SwedenMalvinas väg 3StockholmSE‐114 86Sweden
| | - Sonja Visentin
- Department of Molecular Biotechnology and Health ScienceUniversity of TurinTurin10135Italy
| | - Per Berglund
- Department of Industrial BiotechnologySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
| | - Anders Sönnerborg
- Department of Medicine HuddingeDivision of Infectious DiseasesKarolinska University HospitalKarolinska Institutet, I73Stockholm141 86Sweden
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials EngineeringTechnical University of MunichBoltzmannstrasse 1585748GarchingGermany
- Center for Protein AssembliesTechnical University of MunichErnst‐Otto‐Fischer Str. 885748GarchingGermany
| | - Thomas Crouzier
- Division of GlycoscienceDepartment of ChemistrySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of TechnologyStockholmSweden
- Department of NeuroscienceKarolinska InstitutetStockholmSE‐171 77Sweden
| | - Hongji Yan
- Division of GlycoscienceDepartment of ChemistrySchool of Engineering Sciences in ChemistryBiotechnology and HealthKTH Royal Institute of TechnologyAlbaNova University CenterStockholm106 91Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of TechnologyStockholmSweden
- Department of NeuroscienceKarolinska InstitutetStockholmSE‐171 77Sweden
| |
Collapse
|
34
|
Crnčević N, Rifatbegović Z, Hukić M, Deumić S, Pramenković E, Selimagić A, Gavrankapetanović I, Avdić M. Atypical Viral Infections in Gastroenterology. Diseases 2022; 10:diseases10040087. [PMID: 36278586 PMCID: PMC9590025 DOI: 10.3390/diseases10040087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/29/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
Enteric viruses are commonly found obligate parasites in the gastrointestinal (GI) tract. These viruses usually follow a fecal-oral route of transmission and are characterized by their extraordinary stability as well as resistance in high-stress environments. Most of them cause similar symptoms including vomiting, diarrhea, and abdominal pain. In order to come in contract with mucosal surfaces, these viruses need to pass the three main lines of defense: mucus layer, innate immune defenses, and adaptive immune defenses. The following atypical gastrointestinal infections are discussed: SARS-CoV2, hantavirus, herpes simplex virus I, cytomegalovirus, and calicivirus. Dysbiosis represents any modification to the makeup of resident commensal communities from those found in healthy individuals and can cause a patient to become more susceptible to bacterial and viral infections. The interaction between bacteria, viruses, and host physiology is still not completely understood. However, with growing research on viral infections, dysbiosis, and new methods of detection, we are getting closer to understanding the nature of these viruses, their typical and atypical characteristics, long-term effects, and mechanisms of action in different organ systems.
Collapse
Affiliation(s)
- Neira Crnčević
- Department of Genetics and Bioengineering, International Burch University, Francuske revolucije bb, 71210 Ilidža, Bosnia and Herzegovina
- Correspondence: ; Tel.: +387-(61)-034487
| | - Zijah Rifatbegović
- Department of Abdominal Surgery, Clinic for Surgery, University Clinical Centre Tuzla, 75000 Tuzla, Bosnia and Herzegovina
| | - Mirsada Hukić
- Center for Disease Control and Geohealth Studies, Academy of Sciences and Arts of Bosnia and Herzegovina, Bistrik 7, 71000 Sarajevo, Bosnia and Herzegovina
- Institute for Biomedical Diagnostics and Research Nalaz, Čekaluša 69, 71000 Sarajevo, Bosnia and Herzegovina
| | - Sara Deumić
- Department of Genetics and Bioengineering, International Burch University, Francuske revolucije bb, 71210 Ilidža, Bosnia and Herzegovina
| | - Emina Pramenković
- Department of Genetics and Bioengineering, International Burch University, Francuske revolucije bb, 71210 Ilidža, Bosnia and Herzegovina
| | - Amir Selimagić
- Department of Gastroenterohepatology, General Hospital “Prim. dr. Abdulah Nakas”, 71000 Sarajevo, Bosnia and Herzegovina
| | - Ismet Gavrankapetanović
- Clinic of Orthopedics and Traumatology, University Clinical Center Sarajevo, Bolnička 25, 71000 Sarajevo, Bosnia and Herzegovina
| | - Monia Avdić
- Department of Genetics and Bioengineering, International Burch University, Francuske revolucije bb, 71210 Ilidža, Bosnia and Herzegovina
- Center for Disease Control and Geohealth Studies, Academy of Sciences and Arts of Bosnia and Herzegovina, Bistrik 7, 71000 Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
35
|
Abstract
The thick mucus layer covering of the intestinal epithelium has received increasing attention, owing to its protective role in intestinal infection. However, the exact mechanisms by which the mucus increases intestinal resistance against viral infection remain largely unclear. Here, we identify prominent antiviral activity of the small intestinal mucus and extracted total mucus proteins, as evidenced by their inhibitory effects against porcine epidemic diarrhea virus (PEDV) infection. Of all the extracted mucus proteins, mucin 2 and fraction III (~70 kDa) exhibited potent antiviral activity. We further evaluated the antiviral effects of three candidate factors in fraction III and found that calpain-1 contributed substantially to its antiviral activity. In vivo studies demonstrated that oral administration of calpain-1 provided effective protection against intestinal PEDV infection. As a calcium-activated cysteine protease, calpain-1 inhibited viral invasion by binding to and hydrolyzing the S1 domain of the viral spike protein. The region between amino acids 297 and 337 in the b domain of PEDV S1 protein was critical for calpain-1-mediated hydrolysis. Further investigation indicated that calpain-1 could be produced by goblet cells between intestinal epithelia. Taken together, the results of our study revealed calpain-1 to be a novel antiviral protein in porcine small intestinal mucus, suggesting that calpain-1 has potential for defending against intestinal infections.
Collapse
|
36
|
Zhou J, Rong XL, Cao X, Tang Q, Liu D, Jin YH, Shi XX, Zhong M, Zhao Y, Yang Y. Assembly of Poly(ethylene glycol)ylated Oleanolic Acid on a Linear Polymer as a Pseudomucin for Influenza Virus Inhibition and Adsorption. Biomacromolecules 2022; 23:3213-3221. [PMID: 35797332 DOI: 10.1021/acs.biomac.2c00314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Biomimicry of the mucin barrier function is an efficient strategy to counteract influenza. We report the simple aminolyzation of poly(methyl vinyl ether-alt-maleic anhydride) (PM) using amine-terminated poly(ethylene glycol)ylated oleanolic acid (OAPEG) to mimic the mucin structure and its adsorption of the influenza virus. Direct interactions between influenza hemagglutinin (HA) and the prepared macromolecule evaluated by surface plasmon resonance and isothermal titration calorimetry demonstrated that the multivalent presentation of OAPEG on PM enhanced the binding affinity to HA with a decrease in KD of approximately three orders of magnitude compared with monomeric OAPEG. Moreover, hemagglutination inhibition assay, viral growth inhibition assay, and cytopathic effect reduction assay indicated that the nonglycosylated polymer could mimic natural heavily glycosylated mucin and thus promote the attachment of the virus in a subnanomolar range. Further investigation of the antiviral effects via time-of-addition assay, dynamic light scattering experiments, and transmission electron microscopy photographs indicated that the pseudomucin could adsorb the virion particles and synergistically inhibit the early attachment and final release steps of the influenza infection cycle. These findings demonstrate the effectiveness of the macromolecule in the physical sequestration and prevention of viral infection. Notably, due to its structural similarities with mucin, the biomacropolymer also has the potential for the rational design of antiviral drugs, influenza adsorbents, or filtration materials and the construction of model systems to explore protection against other pathogenic viruses.
Collapse
Affiliation(s)
- JiaPing Zhou
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China.,Research Centre of Modern Analytical Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xue-Lin Rong
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xuan Cao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Qi Tang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Dong Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Yin-Hua Jin
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Xiao-Xiao Shi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Ming Zhong
- Medical College of Shaoguan University, Shaoguan, Guangdong Province 512026, China
| | - YueTao Zhao
- School of Life Sciences, Central South University, Changsha, Hunan Province 410013, China
| | - Yang Yang
- Research Centre of Modern Analytical Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China.,China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| |
Collapse
|
37
|
Takagi J, Aoki K, Turner BS, Lamont S, Lehoux S, Kavanaugh N, Gulati M, Valle Arevalo A, Lawrence TJ, Kim CY, Bakshi B, Ishihara M, Nobile CJ, Cummings RD, Wozniak DJ, Tiemeyer M, Hevey R, Ribbeck K. Mucin O-glycans are natural inhibitors of Candida albicans pathogenicity. Nat Chem Biol 2022; 18:762-773. [PMID: 35668191 PMCID: PMC7613833 DOI: 10.1038/s41589-022-01035-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Mucins are large gel-forming polymers inside the mucus barrier that inhibit the yeast-to-hyphal transition of Candida albicans, a key virulence trait of this important human fungal pathogen. However, the molecular motifs in mucins that inhibit filamentation remain unclear despite their potential for therapeutic interventions. Here, we determined that mucins display an abundance of virulence-attenuating molecules in the form of mucin O-glycans. We isolated and cataloged >100 mucin O-glycans from three major mucosal surfaces and established that they suppress filamentation and related phenotypes relevant to infection, including surface adhesion, biofilm formation and cross-kingdom competition between C. albicans and the bacterium Pseudomonas aeruginosa. Using synthetic O-glycans, we identified three structures (core 1, core 1 + fucose and core 2 + galactose) that are sufficient to inhibit filamentation with potency comparable to the complex O-glycan pool. Overall, this work identifies mucin O-glycans as host molecules with untapped therapeutic potential to manage fungal pathogens.
Collapse
Affiliation(s)
- Julie Takagi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Bradley S Turner
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sabrina Lamont
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH, USA
| | - Sylvain Lehoux
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, National Center for Functional Glycomics, Boston, MA, USA
| | - Nicole Kavanaugh
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Megha Gulati
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Molecular Cell, Cell Press, Cambridge, MA, USA
| | - Ashley Valle Arevalo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, CA, USA
| | - Travis J Lawrence
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, CA, USA
- Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Colin Y Kim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bhavya Bakshi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mayumi Ishihara
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Health Sciences Research Institute, University of California Merced, Merced, CA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, National Center for Functional Glycomics, Boston, MA, USA
| | - Daniel J Wozniak
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH, USA
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Rachel Hevey
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
38
|
Pednekar DD, Liguori MA, Marques CNH, Zhang T, Zhang N, Zhou Z, Amoako K, Gu H. From Static to Dynamic: A Review on the Role of Mucus Heterogeneity in Particle and Microbial Transport. ACS Biomater Sci Eng 2022; 8:2825-2848. [PMID: 35696291 DOI: 10.1021/acsbiomaterials.2c00182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mucus layers (McLs) are on the front line of the human defense system that protect us from foreign abiotic/biotic particles (e.g., airborne virus SARS-CoV-2) and lubricates our organs. Recently, the impact of McLs on human health (e.g., nutrient absorption and drug delivery) and diseases (e.g., infections and cancers) has been studied extensively, yet their mechanisms are still not fully understood due to their high variety among organs and individuals. We characterize these variances as the heterogeneity of McLs, which lies in the thickness, composition, and physiology, making the systematic research on the roles of McLs in human health and diseases very challenging. To advance mucosal organoids and develop effective drug delivery systems, a comprehensive understanding of McLs' heterogeneity and how it impacts mucus physiology is urgently needed. When the role of airway mucus in the penetration and transmission of coronavirus (CoV) is considered, this understanding may also enable a better explanation and prediction of the CoV's behavior. Hence, in this Review, we summarize the variances of McLs among organs, health conditions, and experimental settings as well as recent advances in experimental measurements, data analysis, and model development for simulations.
Collapse
Affiliation(s)
- Dipesh Dinanath Pednekar
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| | - Madison A Liguori
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| | | | - Teng Zhang
- Department of Mechanical and Aerospace Engineering, Syracuse University, Syracuse, New York 13244, United States.,BioInspired Syracuse, Syracuse University, Syracuse, New York 13244, United States
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zejian Zhou
- Department of Electrical and Computer Engineering and Computer Science, University of New Haven, West Haven, Connecticut 06516, United States
| | - Kagya Amoako
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| | - Huan Gu
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, Connecticut 06516, United States
| |
Collapse
|
39
|
Singanayagam A, Footitt J, Marczynski M, Radicioni G, Cross MT, Finney LJ, Trujillo-Torralbo MB, Calderazzo M, Zhu J, Aniscenko J, Clarke TB, Molyneaux PL, Bartlett NW, Moffatt MF, Cookson WO, Wedzicha J, Evans CM, Boucher RC, Kesimer M, Lieleg O, Mallia P, Johnston SL. Airway mucins promote immunopathology in virus-exacerbated chronic obstructive pulmonary disease. J Clin Invest 2022; 132:e120901. [PMID: 35239513 PMCID: PMC9012283 DOI: 10.1172/jci120901] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
The respiratory tract surface is protected from inhaled pathogens by a secreted layer of mucus rich in mucin glycoproteins. Abnormal mucus accumulation is a cardinal feature of chronic respiratory diseases, but the relationship between mucus and pathogens during exacerbations is poorly understood. We identified elevations in airway mucin 5AC (MUC5AC) and MUC5B concentrations during spontaneous and experimentally induced chronic obstructive pulmonary disease (COPD) exacerbations. MUC5AC was more sensitive to changes in expression during exacerbation and was therefore more predictably associated with viral load, inflammation, symptom severity, decrements in lung function, and secondary bacterial infections. MUC5AC was functionally related to inflammation, as Muc5ac-deficient (Muc5ac-/-) mice had attenuated RV-induced (RV-induced) airway inflammation, and exogenous MUC5AC glycoprotein administration augmented inflammatory responses and increased the release of extracellular adenosine triphosphate (ATP) in mice and human airway epithelial cell cultures. Hydrolysis of ATP suppressed MUC5AC augmentation of RV-induced inflammation in mice. Therapeutic suppression of mucin production using an EGFR antagonist ameliorated immunopathology in a mouse COPD exacerbation model. The coordinated virus induction of MUC5AC and MUC5B expression suggests that non-Th2 mechanisms trigger mucin hypersecretion during exacerbations. Our data identified a proinflammatory role for MUC5AC during viral infection and suggest that MUC5AC inhibition may ameliorate COPD exacerbations.
Collapse
Affiliation(s)
- Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Joseph Footitt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthias Marczynski
- School of Engineering and Design, Department of Materials Engineering and
- Center for Protein Assemblies, Technical University of Munich, Munich, Germany
| | - Giorgia Radicioni
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael T. Cross
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lydia J. Finney
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Maria Calderazzo
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jie Zhu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Julia Aniscenko
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas B. Clarke
- Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Philip L. Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nathan W. Bartlett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- College of Health, Medicine and Wellbeing, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Miriam F. Moffatt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - William O. Cookson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jadwiga Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Christopher M. Evans
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard C. Boucher
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mehmet Kesimer
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials Engineering and
- Center for Protein Assemblies, Technical University of Munich, Munich, Germany
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
40
|
Nakahata M, Tominaga N, Saito K, Nishiyama K, Tanino Y, Saiki K, Kojima M, Sakai S. A bio‐synthetic hybrid hydrogel formed under physiological conditions consisting of mucin and a synthetic polymer carrying boronic acid. Macromol Biosci 2022; 22:e2200055. [DOI: 10.1002/mabi.202200055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/28/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Masaki Nakahata
- Department of Macromolecular Science Graduate School of Science Osaka University 1‐1 Machikaneyama‐cho Toyonaka Osaka 560‐0043 Japan
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| | - Naoki Tominaga
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| | - Keishi Saito
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| | - Keita Nishiyama
- Department of Microbiology and Immunology School of Medicine Keio University 35 Shinanomachi Shinjuku Tokyo 160–8582 Japan
| | - Yuya Tanino
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| | - Kiyoshiro Saiki
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| | - Masaru Kojima
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| | - Shinji Sakai
- Division of Chemical Engineering Department of Materials Engineering Science Graduate School of Engineering Science Osaka University 1–3 Machikaneyama‐cho Toyonaka Osaka 560–8531 Japan
| |
Collapse
|
41
|
Models using native tracheobronchial mucus in the context of pulmonary drug delivery research: Composition, structure and barrier properties. Adv Drug Deliv Rev 2022; 183:114141. [PMID: 35149123 DOI: 10.1016/j.addr.2022.114141] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/29/2021] [Accepted: 02/04/2022] [Indexed: 01/15/2023]
Abstract
Mucus covers all wet epithelia and acts as a protective barrier. In the airways of the lungs, the viscoelastic mucus meshwork entraps and clears inhaled materials and efficiently removes them by mucociliary escalation. In addition to physical and chemical interaction mechanisms, the role of macromolecular glycoproteins (mucins) and antimicrobial constituents in innate immune defense are receiving increasing attention. Collectively, mucus displays a major barrier for inhaled aerosols, also including therapeutics. This review discusses the origin and composition of tracheobronchial mucus in relation to its (barrier) function, as well as some pathophysiological changes in the context of pulmonary diseases. Mucus models that contemplate key features such as elastic-dominant rheology, composition, filtering mechanisms and microbial interactions are critically reviewed in the context of health and disease considering different collection methods of native human pulmonary mucus. Finally, the prerequisites towards a standardization of mucus models in a regulatory context and their role in drug delivery research are addressed.
Collapse
|
42
|
Kumar P, Theeyancheri L, Chakrabarti R. Chemically symmetric and asymmetric self-driven rigid dumbbells in a 2D polymer gel. SOFT MATTER 2022; 18:2663-2671. [PMID: 35311848 DOI: 10.1039/d1sm01820e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We employ computer simulations to unveil the translational and rotational dynamics of self-driven chemically symmetric and asymmetric rigid dumbbells in a two-dimensional polymer gel. Our results show that the activity or the self-propulsion always enhances the dynamics of the dumbbells. Making the self-propelled dumbbell chemically asymmetric leads to further enhancement in dynamics. Additionally, the direction of self-propulsion is a key factor for chemically asymmetric dumbbells, where self-propulsion towards the non-sticky half of the dumbbell results in faster translational and rotational dynamics compared to the case with the self-propulsion towards the sticky half of the dumbbell. Our analyses show that both the symmetric and asymmetric passive rigid dumbbells get trapped inside the mesh of the polymer gel, but the chemical asymmetry always facilitates the mesh to mesh motion of the dumbbell and it is even more pronounced when the dumbbell is self-propelled. This results in multiple peaks in the van Hove function with increasing self-propulsion. In a nutshell, we believe that our in silico study can guide researchers to design efficient artificial microswimmers possessing potential applications in site-specific delivery.
Collapse
Affiliation(s)
- Praveen Kumar
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India.
| | - Ligesh Theeyancheri
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India.
| | - Rajarshi Chakrabarti
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India.
| |
Collapse
|
43
|
Wardzala C, Wood AM, Belnap DM, Kramer JR. Mucins Inhibit Coronavirus Infection in a Glycan-Dependent Manner. ACS CENTRAL SCIENCE 2022; 8:351-360. [PMID: 35345395 PMCID: PMC8864775 DOI: 10.1021/acscentsci.1c01369] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Indexed: 05/11/2023]
Abstract
Mucins are a diverse and heterogeneous family of glycoproteins that comprise the bulk of mucus and the epithelial glycocalyx. Mucins are intimately involved in viral transmission. Mucin and virus laden particles can be expelled from the mouth and nose to later infect others. Viruses must also penetrate the mucus layer before cell entry and replication. The role of mucins and their molecular structure have not been well-characterized in coronavirus transmission studies. Laboratory studies predicting high rates of fomite transmission have not translated to real-world infections, and mucins may be one culprit. Here, we probed both surface and direct contact transmission scenarios for their dependence on mucins and their structure. We utilized disease-causing, bovine-derived, human coronavirus OC43. We found that bovine mucins could inhibit the infection of live cells in a concentration- and glycan-dependent manner. The effects were observed in both mock fomite and direct contact transmission experiments and were not dependent upon surface material or time-on-surface. However, the effects were abrogated by removal of the glycans or in a cross-species infection scenario where bovine mucin could not inhibit the infection of a murine coronavirus. Together, our data indicate that the mucin molecular structure plays a complex and important role in host defense.
Collapse
Affiliation(s)
- Casia
L. Wardzala
- Department
of Biomedical Engineering, University of
Utah, 36 South Wasatch Drive, Salt Lake City, Utah 84112, United States
| | - Amanda M. Wood
- Department
of Biomedical Engineering, University of
Utah, 36 South Wasatch Drive, Salt Lake City, Utah 84112, United States
| | - David M. Belnap
- Department
of Biochemistry, University of Utah, 36 South Wasatch Drive, Salt Lake City, Utah 84112, United States
- School
of Biological Sciences, University of Utah, 36 South Wasatch Drive, Salt Lake City, Utah 84112, United States
| | - Jessica R. Kramer
- Department
of Biomedical Engineering, University of
Utah, 36 South Wasatch Drive, Salt Lake City, Utah 84112, United States
- E-mail:
| |
Collapse
|
44
|
Müller WEG, Schröder HC, Neufurth M, Wang X. An unexpected biomaterial against SARS-CoV-2: Bio-polyphosphate blocks binding of the viral spike to the cell receptor. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2021; 51:504-524. [PMID: 34366696 PMCID: PMC8326012 DOI: 10.1016/j.mattod.2021.07.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/22/2021] [Accepted: 07/26/2021] [Indexed: 05/15/2023]
Abstract
No other virus after the outbreak of the influenza pandemic of 1918 affected the world's population as hard as the coronavirus SARS-CoV-2. The identification of effective agents/materials to prevent or treat COVID-19 caused by SARS-CoV-2 is an urgent global need. This review aims to survey novel strategies based on inorganic polyphosphate (polyP), a biologically formed but also synthetically available polyanionic polymeric material, which has the potential of being a potent inhibitor of the SARS-CoV-2 virus-cell-docking machinery. This virus attaches to the host cell surface receptor ACE2 with its receptor binding domain (RBD), which is present at the tips of the viral envelope spike proteins. On the surface of the RBD an unusually conserved cationic groove is exposed, which is composed of basic amino acids (Arg, Lys, and His). This pattern of cationic amino acids, the cationic groove, matches spatially with the anionic polymeric material, with polyP, allowing an electrostatic interaction. In consequence, the interaction between the RBD and ACE2 is potently blocked. PolyP is a physiological inorganic polymer, synthesized by cells and especially enriched in the blood platelets, which releases metabolically useful energy through enzymatic degradation and coupled ADP/ATP formation. In addition, this material upregulates the steady-state-expression of the mucin genes in the epithelial cells. We propose that polyP, with its two antiviral properties (blocking the binding of the virus to the cells and reinforcing the defense barrier against infiltration of the virus) has the potential to be a novel protective/therapeutic anti-COVID-19 agent.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany
| |
Collapse
|
45
|
Wright L, Joyce P, Barnes TJ, Prestidge CA. Mimicking the Gastrointestinal Mucus Barrier: Laboratory-Based Approaches to Facilitate an Enhanced Understanding of Mucus Permeation. ACS Biomater Sci Eng 2021. [PMID: 34784462 DOI: 10.1021/acsbiomaterials.1c00814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The gastrointestinal mucus layer plays a significant role in maintaining gut homeostasis and health, offering protective capacities against the absorption of harmful pathogens as well as commensal gut bacteria and buffering stomach acid to protect the underlying epithelium. Despite this, the mucus barrier is often overlooked during preclinical pharmaceutical development and may pose a significant absorption barrier to high molecular weight or lipophilic drug species. The complex chemical and physical nature of the dynamic mucus layer has proven problematic to reliably replicate in a laboratory setting, leading to the development of multiple mucus models with varying complexity and predictive capacity. This, coupled with the wide range of analysis methods available, has led to a plethora of possible approaches to quantifying mucus permeation; however, the field remains significantly under-represented in biomedical research. For this reason, the development of a concise collation of the available approaches to mucus permeation is essential. In this review, we explore widely utilized mucus mimics ranging in complexity from simple mucin solutions to native mucus preparations for their predictive capacity in mucus permeation analysis. Furthermore, we highlight the diverse range of laboratory-based models available for the analysis of mucus interaction and permeability with a specific focus on in vitro, ex vivo, and in situ models. Finally, we highlight the predictive capacity of these models in correlation with in vivo pharmacokinetic data. This review provides a comprehensive and critical overview of the available technologies to analyze mucus permeation, facilitating the efficient selection of appropriate tools for further advancement in oral drug delivery.
Collapse
Affiliation(s)
- Leah Wright
- UniSA: Clinical and Health Sciences, Bradley Building, North Terrace, University of South Australia, Adelaide, 5001, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, North Terrace, University of South Australia, Adelaide, 5001, Australia
| | - Paul Joyce
- UniSA: Clinical and Health Sciences, Bradley Building, North Terrace, University of South Australia, Adelaide, 5001, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, North Terrace, University of South Australia, Adelaide, 5001, Australia
| | - Timothy J Barnes
- UniSA: Clinical and Health Sciences, Bradley Building, North Terrace, University of South Australia, Adelaide, 5001, Australia
| | - Clive A Prestidge
- UniSA: Clinical and Health Sciences, Bradley Building, North Terrace, University of South Australia, Adelaide, 5001, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, North Terrace, University of South Australia, Adelaide, 5001, Australia
| |
Collapse
|
46
|
Marczynski M, Kimna C, Lieleg O. Purified mucins in drug delivery research. Adv Drug Deliv Rev 2021; 178:113845. [PMID: 34166760 DOI: 10.1016/j.addr.2021.113845] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022]
Abstract
One of the main challenges in the field of drug delivery remains the development of strategies to efficiently transport pharmaceuticals across mucus barriers, which regulate the passage and retention of molecules and particles in all luminal spaces of the body. A thorough understanding of the molecular mechanisms, which govern such selective permeability, is key for achieving efficient translocation of drugs and drug carriers. For this purpose, model systems based on purified mucins can contribute valuable information. In this review, we summarize advances that were made in the field of drug delivery research with such mucin-based model systems: First, we give an overview of mucin purification procedures and discuss the suitability of model systems reconstituted from purified mucins to mimic native mucus. Then, we summarize techniques to study mucin binding. Finally, we highlight approaches that made use of mucins as building blocks for drug delivery platforms or employ mucins as active compounds.
Collapse
|
47
|
Marczynski M, Lieleg O. Forgotten but not gone: Particulate matter as contaminations of mucosal systems. BIOPHYSICS REVIEWS 2021; 2:031302. [PMID: 38505633 PMCID: PMC10903497 DOI: 10.1063/5.0054075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/14/2021] [Indexed: 03/21/2024]
Abstract
A decade ago, environmental issues, such as air pollution and the contamination of the oceans with microplastic, were prominently communicated in the media. However, these days, political topics, as well as the ongoing COVID-19 pandemic, have clearly taken over. In spite of this shift in focus regarding media representation, researchers have made progress in evaluating the possible health risks associated with particulate contaminations present in water and air. In this review article, we summarize recent efforts that establish a clear link between the increasing occurrence of certain pathological conditions and the exposure of humans (or animals) to airborne or waterborne particulate matter. First, we give an overview of the physiological functions mucus has to fulfill in humans and animals, and we discuss different sources of particulate matter. We then highlight parameters that govern particle toxicity and summarize our current knowledge of how an exposure to particulate matter can be related to dysfunctions of mucosal systems. Last, we outline how biophysical tools and methods can help researchers to obtain a better understanding of how particulate matter may affect human health. As we discuss here, recent research has made it quite clear that the structure and functions of those mucosal systems are sensitive toward particulate contaminations. Yet, our mechanistic understanding of how (and which) nano- and microparticles can compromise human health via interacting with mucosal barriers is far from complete.
Collapse
|
48
|
Impact of artificial sputum media formulation on Pseudomonas aeruginosa secondary metabolite production. J Bacteriol 2021; 203:e0025021. [PMID: 34398662 PMCID: PMC8508215 DOI: 10.1128/jb.00250-21] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
In vitro culture media are being developed to understand how host site-specific nutrient profiles influence microbial pathogenicity and ecology. To mimic the cystic fibrosis (CF) lung environment, a variety of artificial sputum media (ASM) have been created. However, the composition of these ASM vary in the concentration of key nutrients, including amino acids, lipids, DNA, and mucin. In this work, we used feature-based molecular networking (FBMN) to perform comparative metabolomics of Pseudomonas aeruginosa, the predominant opportunistic pathogen infecting the lungs of people with CF, cultured in nine different ASM. We found that the concentration of aromatic amino acids and iron from mucin added to the media contributes to differences in the production of P. aeruginosa virulence-associated secondary metabolites. IMPORTANCE Different media formulations aiming to replicate in vivo infection environments contain different nutrients, which affects interpretation of experimental results. Inclusion of undefined components, such as commercial porcine gastric mucin (PGM), in an otherwise chemically defined medium can alter the nutrient content of the medium in unexpected ways and influence experimental outcomes.
Collapse
|
49
|
Ott IM, Strine MS, Watkins AE, Boot M, Kalinich CC, Harden CA, Vogels CBF, Casanovas-Massana A, Moore AJ, Muenker MC, Nakahata M, Tokuyama M, Nelson A, Fournier J, Bermejo S, Campbell M, Datta R, Dela Cruz CS, Farhadian SF, Ko AI, Iwasaki A, Grubaugh ND, Wilen CB, Wyllie AL. Stability of SARS-CoV-2 RNA in Nonsupplemented Saliva. Emerg Infect Dis 2021; 27:1146-1150. [PMID: 33754989 PMCID: PMC8007305 DOI: 10.3201/eid2704.204199] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The expense of saliva collection devices designed to stabilize severe acute respiratory syndrome coronavirus 2 RNA is prohibitive to mass testing. However, virus RNA in nonsupplemented saliva is stable for extended periods and at elevated temperatures. Simple plastic tubes for saliva collection will make large-scale testing and continued surveillance easier.
Collapse
|
50
|
Size shifting of solid lipid nanoparticle system triggered by alkaline phosphatase for site specific mucosal drug delivery. Eur J Pharm Biopharm 2021; 163:109-119. [PMID: 33775852 DOI: 10.1016/j.ejpb.2021.03.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022]
Abstract
We aim to prepare a size-shifting nanocarrier for site-targeting mucosal drug delivery that can penetrate through mucus gel layer and remain close to the absorption membrane. As nanocarriers can be engineered to penetrate mucus but they can also back diffuse into outer mucus regions, a size shifting to micron range once they have reached the absorption membrane would prevent back-diffusion effect and extend drug release over a long period of time. For this purpose, we loaded solid lipid nanoparticles (SLN) with a phosphate ester surfactant and octadecylamine. Alkaline phosphatase (AP), a membrane bound enzyme was for the first time utilized as an in situ partner for triggering the size conversion at epithelial cell surface. Having the size of ~120 nm, SLN with hydrophilic and phosphate-decorated shells were shown to penetrate through mucus gel and form aggregates above cell layer surface. Aggregates of 5-8 µm were formed due to interparticle interactions induced by enzymatic phosphate removal after ~30 min in contact with isolated AP. The developed SLN system could be a potential tool for mucosal drug delivery to AP-expressing tissues like colon, lung, cervix, vagina and some mucus-secreting tumors.
Collapse
|