1
|
Epstein JA, Ramon GZ. In-situ measurement of the internal compaction of a soft material caused by permeation flow. J Colloid Interface Sci 2024; 673:883-892. [PMID: 38908287 DOI: 10.1016/j.jcis.2024.06.095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024]
Abstract
HYPOTHESIS The compaction of hydrogel films under permeation flow can be measured, in-situ, by tracking the internal displacements of their structure, thereby revealing the internal deformation profile. Additionally, monitoring the permeation flow rate and applied pressure over time enables determination of variations in the hydrogel's permeability due to flow-induced compaction. Hydrogels are soft porous materials capable of containing high amounts of water within their polymeric matrix. Flow-induced internal deformation can modify the hydrogel's permeability and selectivity, which are important attributes in separation processes, both industrial (e.g., membrane-based water purification) and natural (mucous filters in suspension feeders and intestinal lining) systems. Measuring the flow-induced compaction in thin hydrogels films can reveal the interplay between flow and permeability. However, the micro-scale internal compaction remains uncharted for due to experimental challenges. EXPERIMENTS A technique is demonstrated for analyzing the compaction and stratification of permeable soft materials, in-situ, created by a pressure-driven permeation flow. To this end, the internal deformations within a soft material layer are calculated, based on tracking the positions of fluorescent micro-tracers that are embedded within the soft material. We showcase the capabilities of this technique by examining a hundred-micron-thick calcium-alginate cake deposited on a nanofiltration membrane, emphasizing the achieved micro-scale resolution of the local compaction measurements. FINDINGS The results highlight the possibility to examine thin hydrogel films and their internal deformation produced by flow-induced stresses when varying the flow conditions. The method enables the simultaneous calculation of the soft material's permeance, as the pressure-driven flow conditions are continuously monitored. In summary, the proposed method provides a powerful tool for characterizing the behaviour of permeable soft materials under permeation conditions, with potential applications in engineering, biophysics and material science.
Collapse
Affiliation(s)
- José A Epstein
- Department of Civil & Environmental Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Guy Z Ramon
- Department of Civil & Environmental Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel; Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
2
|
Feng G, Wang G, Li T, Han C, Han K, Guo J, Wan Z, Yang X. Phosphatidylcholine Surface Hydration-Dependent Adsorption to Mucin Enhances Intestinal Mucus Barrier Function. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:18977-18987. [PMID: 39169607 DOI: 10.1021/acs.langmuir.4c01666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The crucial role of zwitterionic phosphatidylcholines (PC) within mucus gel is essential for maintaining intestinal homeostasis, while the underlying mechanism remains incompletely understood. Herein, we compared the dynamic interfacial adsorption behavior of saturated dipalmitoylphosphatidylcholine (DPPC) and unsaturated dioleoylphosphatidylcholine (DOPC) to intestinal mucin and their impact on the intestinal mucus barrier function. Results of quartz crystal microbalance with dissipation showed that the highly surface-hydrated DPPC vesicles exhibited significantly faster and more extensive adsorption to purified intestinal mucin than the slightly surface-hydrated DOPC vesicles. Utilizing an intestinal Caco-2/HT29-MTX coculture model, we observed that DPPC vesicles adsorbed much more to the mucus gel compared to DOPC vesicles. Additionally, DPPC vesicle adsorption displayed increased wetting, and converse for DOPC vesicles. Interestingly, both of them exhibited nearly the same protective effects against cell injury induced by peptic-tryptic digests of gliadin (PTG). The partial mechanism involved the binding of PTG to DPPC and DOPC within the mucus gel, thereby restricting PTG contact with the underlying epithelial cells. These findings shed light on the intricate interfacial dynamics of PC adsorption to mucin and their implications for maintaining the integrity of the intestinal mucus barrier.
Collapse
Affiliation(s)
- Guangxin Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Huangdao District, Qingdao 266003, Shandong Province, China
| | - Gaoshang Wang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Tanghao Li
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Chuanwu Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Kaining Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jian Guo
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Zhili Wan
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
3
|
Homolak J. A simple and affordable open-source quantitative tribometric assay and the use thereof for the analysis of a commercial water-based lubricant. Comput Methods Biomech Biomed Engin 2024; 27:1322-1331. [PMID: 37504954 DOI: 10.1080/10255842.2023.2241592] [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: 04/06/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Quantitative assessment of biotribological properties requires expensive specialized equipment. The aim was to: i) adapt an open-source load cell-based platform (PASTA) for biotribometric analysis; ii) study the effects of oxidation on the water-based lubricant using PASTA. Water-based lubricant was treated with 2,2'-azobis(2-amidinopropane) dihydrochloride and/or glutathione. The samples were analyzed with the ORP-146S redox microsensor and PASTA using a modified HX711 integrated circuit bord, NodeMCU ESP-32S, and an open-source Python script. PASTA can be adapted for affordable and reliable quantitative biotribometric assessment. Glutathione can prevent the loss of lubrication capacity of a water-based lubricant upon exposure to air.
Collapse
Affiliation(s)
- Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
4
|
Villegas-Novoa C, Wang Y, Sims CE, Allbritton NL. Creation of a spatially complex mucus bilayer on an in vitro colon model. Sci Rep 2024; 14:16849. [PMID: 39039235 PMCID: PMC11263341 DOI: 10.1038/s41598-024-67591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
The colonic epithelium is comprised of three-dimensional crypts (3D) lined with mucus secreted by a heterogeneous population of goblet cells. In this study, we report the formation of a long-lived, and self-renewing replica of human 3D crypts with a mucus layer patterned in the X-Y-Z dimensions. Primary colon cells were cultured on a shaped scaffold under an air-liquid interface to yield architecturally accurate crypts with a mucus bilayer (605 ± 180 μm thick) possessing an inner (149 ± 50 μm) and outer (435 ± 111 μm) region. Lectins with distinct carbohydrate-binding preferences demonstrated that the mucus in the intercrypt regions was chemically distinct from that above and within the crypts replicating in vivo chemical patterning. Constitutive mucus secretion ejected beads from crypt lumens in 8-10 days, while agonist-stimulated secretion increased mucus thickness by 17-fold in 8 h. The tissue was long-lived, > 50 days, the longest time assessed. In conclusion, the in vitro mucus replicated key physiology of the human mucus, including the bilayer (Z) structure and intercrypt-crypt (X-Y) zones, constitutive mucus flow, spatially complex chemical attributes, and mucus secretion response to stimulation, with the potential to reveal local and global determinants of mucus function and its breakdown in disease.
Collapse
Affiliation(s)
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Christopher E Sims
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
5
|
Wang Z, Shen J. The role of goblet cells in Crohn' s disease. Cell Biosci 2024; 14:43. [PMID: 38561835 PMCID: PMC10985922 DOI: 10.1186/s13578-024-01220-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of Crohn's disease (CD), a subtype of inflammatory bowel disease (IBD), is increasing worldwide. The pathogenesis of CD is hypothesized to be related to environmental, genetic, immunological, and bacterial factors. Current studies have indicated that intestinal epithelial cells, including columnar, Paneth, M, tuft, and goblet cells dysfunctions, are strongly associated with these pathogenic factors. In particular, goblet cells dysfunctions have been shown to be related to CD pathogenesis by direct or indirect ways, according to the emerging studies. The mucus barrier was established with the help of mucins secreted by goblet cells. Not only do the mucins mediate the mucus barrier permeability and bacterium selection, but also, they are closely linked with the endothelial reticulum stress during the synthesis process. Goblet cells also play a vital role in immune response. It was indicated that goblet cells take part in the antigen presentation and cytokines secretion process. Disrupted goblet cells related immune process were widely discovered in CD patients. Meanwhile, dysbiosis of commensal and pathogenic microbiota can induce myriad immune responses through mucus and goblet cell-associated antigen passage. Microbiome dysbiosis lead to inflammatory reaction against pathogenic bacteria and abnormal tolerogenic response. All these three pathways, including the loss of mucus barrier function, abnormal immune reaction, and microbiome dysbiosis, may have independent or cooperative effect on the CD pathogenesis. However, many of the specific mechanisms underlying these pathways remain unclear. Based on the current understandings of goblet cell's role in CD pathogenesis, substances including butyrate, PPARγagonist, Farnesoid X receptor agonist, nuclear factor-Kappa B, nitrate, cytokines mediators, dietary and nutrient therapies were all found to have potential therapeutic effects on CD by regulating the goblet cells mediated pathways. Several monoclonal antibodies already in use for the treatment of CD in the clinical settings were also found to have some goblet cells related therapeutic targets. In this review, we introduce the disease-related functions of goblet cells, their relationship with CD, their possible mechanisms, and current CD treatments targeting goblet cells.
Collapse
Affiliation(s)
- Zichen Wang
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Ministry of Health, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No.160 PuJian Road, Shanghai, 200127, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Ministry of Health, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No.160 PuJian Road, Shanghai, 200127, China.
| |
Collapse
|
6
|
Moran ET, Bedford MR. Basis for the diversity and extent in loss of digestible nutrients created by dietary phytin: Emphasis on fowl and swine. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:422-428. [PMID: 38379939 PMCID: PMC10876675 DOI: 10.1016/j.aninu.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 02/22/2024]
Abstract
Phytin is the Ca2+-Mg2+-K+ salt of phytic acid that is created and deposited in the aleurone layer and/or germ of grains and legumes. Its high presence in feedstuffs for fowl and swine diets results in it being a universal and significant impediment to optimum performance. Phytin impairs gastrointestinal recovery of a wide array of nutrients, the effect varying with the nutrient concerned. On exposure to low pH during gastric digestion, phytin dissociates into phytic acid and solubilized Ca2+. Even at low gastric pH, phytic acid is negatively charged which forms the basis of its anti-nutritive behavior. Pepsinogen has extensive basic amino acids on its activation peptide that are presented as cations at low pH which are targeted by pepsin for activation. Partially crystalized Ca2+ near the enzyme's active site further stabilizes its newly formed structure. Thus, phytic acid appears to interfere with gastric digestion by several mechanisms; interfering with pepsinogen activation by binding to the polypeptide's basic amino acids; coordinating free Ca2+, destabilizing pepsin; binding some dietary proteins directly, further compromising gastric proteolysis. Upon digesta attaining neutrality in the duodenum, Ca2+ and other cations re-bind with accessible anions, phytic acid being a significant contender. Phytate not only binds free cations but can also strip them from enzymes (e.g. Ca2+, Zn2+) which reduces their structural resistance to autolysis and ability as co-factors (e.g. Zn2+) to increase enzyme activity. Goblet cells initially employ Ca2+ as an electronic shield between mucin layers enabling granule formation and cell storage. After mucin granule release, Ca2+ is progressively displaced by Na+ to free the viscous mucins enabling its translocation. Mucin entangles with the glycocalyx of adjacent enterocytes thereby constructing the unstirred water layer (USWL). Excessive removal of Ca2+ from mucin by phytic acid increases its fluidity facilitating its loss from the USWL with its associated Na+. This partly explains increased mucin and Na+ losses noted with high phytate diets. This review suggests that phytic acid binding of Ca2+ and less so Zn2+ is the basis for the diversity in nutrient losses encountered and that such losses are in proportion to dietary phytate content.
Collapse
Affiliation(s)
- Edwin T. Moran
- Poultry Science Department, Auburn University, AL, 36830-5416, USA
| | - Michael R. Bedford
- AB Vista, Woodstock Court, Blenheim Road, Marlborough, Wiltshire, SN8 4AN, UK
| |
Collapse
|
7
|
Park J, Ghanim R, Rahematpura A, Gerage C, Abramson A. Electromechanical convective drug delivery devices for overcoming diffusion barriers. J Control Release 2024; 366:650-667. [PMID: 38190971 PMCID: PMC10922834 DOI: 10.1016/j.jconrel.2024.01.008] [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: 09/08/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
Drug delivery systems which rely on diffusion for mass transport, such as hydrogels and nanoparticles, have enhanced drug targeting and extended delivery profiles to improve health outcomes for patients suffering from diseases including cancer and diabetes. However, diffusion-dependent systems often fail to provide >0.01-1% drug bioavailability when transporting macromolecules across poorly permeable physiological tissues such as the skin, solid tumors, the blood-brain barrier, and the gastrointestinal walls. Convection-enabling robotic ingestibles, wearables, and implantables physically interact with tissue walls to improve bioavailability in these settings by multiple orders of magnitude through convective mass transfer, the process of moving drug molecules via bulk fluid flow. In this Review, we compare diffusive and convective drug delivery systems, highlight engineering techniques that enhance the efficacy of convective devices, and provide examples of synergies between the two methods of drug transport.
Collapse
Affiliation(s)
- Jihoon Park
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ramy Ghanim
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Adwik Rahematpura
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Caroline Gerage
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alex Abramson
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
8
|
Li T, Han K, Feng G, Guo J, Wan Z, Yang X. Condensation of Soy Protein Peptides Contributes to Sequester Bile Acids and Mitigate LPS-Induced Inflammation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1266-1275. [PMID: 38109330 DOI: 10.1021/acs.jafc.3c06480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Soy protein is widely known to have serum triglyceride (TG) and cholesterol-lowering effects associated with a reduced risk of cardiovascular disease. Recent studies highlighted that the extension region (ER) domain of soy 7S globulin (β-conglycinin) is a key component responsible for the serum TG-lowering effect via modulation of bile acid (BA) homeostasis. Here, we studied the sequestration of BAs by ER peptides during intestinal digestion in vitro and assessed the anti-inflammatory effects of ER peptides using Caco-2/HT29-MTX/RAW264.7 triple-cell cocultures as an intestine cell model. Results show that ER peptides, which share characteristics of intrinsically disordered regions (IDRs), are capable of forming peptide condensates and exhibit the capability to sequester BA-containing colloidal structures during intestinal digestion in vitro. Moreover, BAs enhance the penetration of peptide condensates within the mucus layer, enabling ER peptides to mitigate lipopolysaccharide (LPS)-induced gut inflammation. These results provide a possible explanation for the molecular mechanisms underlying the modulation of BA homeostasis by soybean proteins.
Collapse
Affiliation(s)
- Tanghao Li
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Kaining Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Guangxin Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jian Guo
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Zhili Wan
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
9
|
Milian D, Robert de Saint Vincent M, Patarin J, Bodiguel H. Gastropod Slime-Based Gel as an Adjustable Synthetic Model for Human Airway Mucus. Biomacromolecules 2024; 25:400-412. [PMID: 38124283 DOI: 10.1021/acs.biomac.3c01043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Airway mucus works as a protective barrier in the human body, as it entraps pathogens that will be later cleared from the airways by ciliary transport or by coughing, thus featuring the rheological properties of a highly stretchable gel. Nonetheless, the study of these physical barrier as well as transport properties remains limited due to the restricted and invasive access to lungs and bronchi to retrieve mucus and to the poor repeatability inherent to native mucus samples. To overcome these limits, we report on a biobased synthetic mucus prepared from snail slime and multibranched thiol cross-linker, which are able to establish disulfide bonds, in analogy with the disulfide bonding of mucins, and therefore build viscoelastoplastic hydrogels. The gel macroscopic properties are tuned by modifying the cross-linker and slime concentrations and can quantitatively match those of native sputum from donors with cystic fibrosis (CF) or non-cystic fibrosis bronchiectasis (NCFB) both in the small- and large-deformation regimes. Heterogeneous regimes were locally found in the mucus model by passive microrheology, in which both diffusive and non-diffusive motion are present, similar to what is observed in sputa. The biobased synthetic approach proposed in the present study thus allows to produce, with commercially available components, a promising model to native respiratory mucus regarding both mechanical and, to a lesser extent, physicochemical aspects.
Collapse
Affiliation(s)
- Diego Milian
- Univ. Grenoble Alpes, CNRS, Grenoble INP, LRP, 38000 Grenoble, France
- Rheonova, 1 Allée de Certèze, 38610 Gières, France
| | | | | | - Hugues Bodiguel
- Univ. Grenoble Alpes, CNRS, Grenoble INP, LRP, 38000 Grenoble, France
| |
Collapse
|
10
|
Donahue R, Sahoo JK, Rudolph S, Chen Y, Kaplan DL. Mucosa-Mimetic Materials for the Study of Intestinal Homeostasis and Disease. Adv Healthc Mater 2023; 12:e2300301. [PMID: 37329337 DOI: 10.1002/adhm.202300301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Mucus is a viscoelastic hydrogel that lines and protects the epithelial surfaces of the body that houses commensal microbiota and functions in host defense against pathogen invasion. As a first-line physical and biochemical barrier, intestinal mucus is involved in immune surveillance and spatial organization of the microbiome, while dysfunction of the gut mucus barrier is implicated in several diseases. Mucus can be collected from a variety of mammalian sources for study, however, established methods are challenging in terms of scale and efficiency, as well as with regard to rheological similarity to native human mucus. Therefore, there is a need for mucus-mimetic hydrogels that more accurately reflect the physical and chemical profile of the in vivo human epithelial environment to enable the investigation of the role of mucus in human disease and interactions with the intestinal microbiome. This review will evaluate the material properties of synthetic mucus mimics to date designed to address the above need, with a focus toward an improved understanding of the biochemical and immunological functions of these biopolymers related to utility for research and therapeutic applications.
Collapse
Affiliation(s)
- Rebecca Donahue
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|
11
|
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
|
12
|
Wang CM, Fernez MT, Woolston BM, Carrier RL. Native gastrointestinal mucus: Critical features and techniques for studying interactions with drugs, drug carriers, and bacteria. Adv Drug Deliv Rev 2023; 200:114966. [PMID: 37329985 PMCID: PMC11184232 DOI: 10.1016/j.addr.2023.114966] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Gastrointestinal mucus plays essential roles in modulating interactions between intestinal lumen contents, including orally delivered drug carriers and the gut microbiome, and underlying epithelial and immune tissues and cells. This review is focused on the properties of and methods for studying native gastrointestinal mucus and its interactions with intestinal lumen contents, including drug delivery systems, drugs, and bacteria. The properties of gastrointestinal mucus important to consider in its analysis are first presented, followed by a discussion of different experimental setups used to study gastrointestinal mucus. Applications of native intestinal mucus are then described, including experimental methods used to study mucus as a barrier to drug delivery and interactions with intestinal lumen contents that impact barrier properties. Given the significance of the microbiota in health and disease, its impact on drug delivery and drug metabolism, and the use of probiotics and microbe-based delivery systems, analysis of interactions of bacteria with native intestinal mucus is then reviewed. Specifically, bacteria adhesion to, motility within, and degradation of mucus is discussed. Literature noted is focused largely on applications of native intestinal mucus models as opposed to isolated mucins or reconstituted mucin gels.
Collapse
Affiliation(s)
- Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Matthew T Fernez
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Benjamin M Woolston
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, USA; Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
13
|
Homolak J, De Busscher J, Zambrano-Lucio M, Joja M, Virag D, Babic Perhoc A, Knezovic A, Osmanovic Barilar J, Salkovic-Petrisic M. Altered Secretion, Constitution, and Functional Properties of the Gastrointestinal Mucus in a Rat Model of Sporadic Alzheimer's Disease. ACS Chem Neurosci 2023; 14:2667-2682. [PMID: 37477640 PMCID: PMC10401635 DOI: 10.1021/acschemneuro.3c00223] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023] Open
Abstract
The gastrointestinal (GI) system is affected in Alzheimer's disease (AD); however, it is currently unknown whether GI alterations arise as a consequence of central nervous system (CNS) pathology or play a causal role in the pathogenesis. GI mucus is a possible mediator of GI dyshomeostasis in neurological disorders as the CNS controls mucus production and secretion via the efferent arm of the brain-gut axis. The aim was to use a brain-first model of sporadic AD induced by intracerebroventricular streptozotocin (STZ-icv; 3 mg/kg) to dissect the efferent (i.e., brain-to-gut) effects of isolated central neuropathology on the GI mucus. Morphometric analysis of goblet cell mucigen granules revealed altered GI mucus secretion in the AD model, possibly mediated by the insensitivity of AD goblet cells to neurally evoked mucosal secretion confirmed by ex vivo cholinergic stimulation of isolated duodenal rings. The dysfunctional efferent control of the GI mucus secretion results in altered biochemical composition of the mucus associated with reduced mucin glycoprotein content, aggregation, and binding capacity in vitro. Finally, functional consequences of the reduced barrier-forming capacity of the mucin-deficient AD mucus are demonstrated using the in vitro two-compartment caffeine diffusion interference model. Isolated central AD-like neuropathology results in the loss of efferent control of GI homeostasis via the brain-gut axis and is characterized by the insensitivity to neurally evoked mucosal secretion, altered mucus constitution with reduced mucin content, and reduced barrier-forming capacity, potentially increasing the susceptibility of the STZ-icv rat model of AD to GI and systemic inflammation induced by intraluminal toxins, microorganisms, and drugs.
Collapse
Affiliation(s)
- Jan Homolak
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Croatian
Institute for Brain Research, University
of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | | | - Miguel Zambrano-Lucio
- School
of Medicine, Autonomous University of Nuevo
Leon, Monterrey, Nuevo Leon 66455, Mexico
| | - Mihovil Joja
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Department
of Infection and Immunity, Luxembourg Institute
of Health, L-4354 Esch-sur-Alzette, Luxembourg
- Faculty
of
Science, Technology and Medicine, University
of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Davor Virag
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Croatian
Institute for Brain Research, University
of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Ana Babic Perhoc
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Croatian
Institute for Brain Research, University
of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Ana Knezovic
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Croatian
Institute for Brain Research, University
of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Croatian
Institute for Brain Research, University
of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Melita Salkovic-Petrisic
- Department
of Pharmacology, University of Zagreb School
of Medicine, 10 000 Zagreb, Croatia
- Croatian
Institute for Brain Research, University
of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| |
Collapse
|
14
|
Yang S, Duncan G. Synthetic Mucus Biomaterials for Antimicrobial Peptide Delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531025. [PMID: 36945438 PMCID: PMC10028879 DOI: 10.1101/2023.03.07.531025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
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 AMPs and enhancing their therapeutic effect. LL37 loaded SM hydrogels demonstrated controlled release of LL37 over 8 hours as a result of charge-mediated interactions between mucins and LL37 AMPs. Compared to treatment with LL37 alone where antimicrobial activity was reduced after 3 hours, LL37-SM hydrogels inhibited Pseudomonas aeruginosa PAO1 growth over 12 hours. LL37-SM hydrogel treatment reduced PAO1 viability over 6 hours 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
- University of Maryland, Fischell Department of Bioengineering, College Park, MD
| | - Gregg Duncan
- University of Maryland, Fischell Department of Bioengineering, College Park, MD
| |
Collapse
|
15
|
Wright L, Barnes TJ, Joyce P, Prestidge CA. Optimisation of a High-Throughput Model for Mucus Permeation and Nanoparticle Discrimination Using Biosimilar Mucus. Pharmaceutics 2022; 14:2659. [PMID: 36559151 PMCID: PMC9782027 DOI: 10.3390/pharmaceutics14122659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
High-throughput permeation models are essential in drug development for timely screening of new drug and formulation candidates. Nevertheless, many current permeability assays fail to account for the presence of the gastrointestinal mucus layer. In this study, an optimised high-throughput mucus permeation model was developed employing a highly biorelevant mucus mimic. While mucus permeation is primarily conducted in a simple mucin solution, the complex chemistry, nanostructure and rheology of mucus is more accurately modelled by a synthetic biosimilar mucus (BSM) employing additional protein, lipid and rheology-modifying polymer components. Utilising BSM, equivalent permeation of various molecular weight fluorescein isothiocyanate-dextrans were observed, compared with native porcine jejunal mucus, confirming replication of the natural mucus permeation barrier. Furthermore, utilising synthetic BSM facilitated the analysis of free protein permeation which could not be quantified in native mucus due to concurrent proteolytic degradation. Additionally, BSM could differentiate between the permeation of poly (lactic-co-glycolic) acid nanoparticles (PLGA-NP) with varying surface chemistries (cationic, anionic and PEGylated), PEG coating density and size, which could not be achieved by a 5% mucin solution. This work confirms the importance of utilising highly biorelevant mucus mimics in permeation studies, and further development will provide an optimal method for high-throughput mucus permeation analysis.
Collapse
Affiliation(s)
| | | | | | - Clive A. Prestidge
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| |
Collapse
|
16
|
Subramanian DA, Langer R, Traverso G. Mucus interaction to improve gastrointestinal retention and pharmacokinetics of orally administered nano-drug delivery systems. J Nanobiotechnology 2022; 20:362. [PMID: 35933341 PMCID: PMC9356434 DOI: 10.1186/s12951-022-01539-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
Oral delivery of therapeutics is the preferred route of administration due to ease of administration which is associated with greater patient medication adherence. One major barrier to oral delivery and intestinal absorption is rapid clearance of the drug and the drug delivery system from the gastrointestinal (GI) tract. To address this issue, researchers have investigated using GI mucus to help maximize the pharmacokinetics of the therapeutic; while mucus can act as a barrier to effective oral delivery, it can also be used as an anchoring mechanism to improve intestinal residence. Nano-drug delivery systems that use materials which can interact with the mucus layers in the GI tract can enable longer residence time, improving the efficacy of oral drug delivery. This review examines the properties and function of mucus in the GI tract, as well as diseases that alter mucus. Three broad classes of mucus-interacting systems are discussed: mucoadhesive, mucus-penetrating, and mucolytic drug delivery systems. For each class of system, the basis for mucus interaction is presented, and examples of materials that inform the development of these systems are discussed and reviewed. Finally, a list of FDA-approved mucoadhesive, mucus-penetrating, and mucolytic drug delivery systems is reviewed. In summary, this review highlights the progress made in developing mucus-interacting systems, both at a research-scale and commercial-scale level, and describes the theoretical basis for each type of system.
Collapse
Affiliation(s)
- Deepak A Subramanian
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni Traverso
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Feng G, Han K, Yang Q, Feng W, Guo J, Wang J, Yang X. Interaction of Pyrogallol-Containing Polyphenols with Mucin Reinforces Intestinal Mucus Barrier Properties. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9536-9546. [PMID: 35852590 DOI: 10.1021/acs.jafc.2c03564] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
High consumption of polyphenol-rich green tea, coffee, fruits, and vegetables is associated with a low risk of human chronic diseases. Recent studies highlight the relevance of polyphenol-mediated gut microbiota modulation and its impact on mucus barrier. Herein, we study the direct interaction of epicatechin (EC), epigallocatechin gallate (EGCG), and tannic acid (TA) with intestinal mucin by isothermal titration calorimetry and multiple particle tracking and the impact on mucus barrier using ex vivo mucus and Caco-2/HT29-MTX cocultures. Results show that pyrogallol-containing polyphenols EGCG and TA exhibit strong binding to intestinal mucin and reinforce mucus barrier, whereas EC does not. ECGG and TA also mitigate gliadin-mediated cytotoxicity and inflammation. The chemical binding of EGCG and TA to the nucleophilic thiol groups of mucins shows their roles as cross-linkers of mucin networks. These results bring a novel understanding of the health benefits of polyphenols and provide support for the consumption of pyrogallol-containing beverages like green tea as a potential dietary therapy for gluten-related disorders.
Collapse
Affiliation(s)
- Guangxin Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Kaining Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qian Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Weiting Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jian Guo
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jinmei Wang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
18
|
Sardelli L, Vangosa FB, Merli M, Ziccarelli A, Visentin S, Visai L, Petrini P. Bioinspired in vitro intestinal mucus model for 3D-dynamic culture of bacteria. BIOMATERIALS ADVANCES 2022; 139:213022. [PMID: 35891596 DOI: 10.1016/j.bioadv.2022.213022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/27/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
The intestinal mucus is a biological barrier that supports the intestinal microbiota growth and filters molecules. To perform these functions, mucus possesses optimized microstructure and viscoelastic properties and it is steadily replenished thus flowing along the gut. The available in vitro intestinal mucus models are useful tools in investigating the microbiota-human cells interaction, and are used as matrices for bacterial culture or as static component of microfluidic devices like gut-on-chips. The aim of this work is to engineer an in vitro mucus models (I-Bac3Gel) addressing in a single system physiological viscoelastic properties (i.e., 2-200 Pa), 3D structure and suitability for dynamic bacterial culture. Homogeneously crosslinked alginate hydrogels are optimized in composition to obtain target viscoelastic and microstructural properties. Then, rheological tests are exploited to assess a priori the hydrogels capability to withstand the flow dynamic condition. We experimentally assess the suitability of I-Bac3Gels in the evolving field of microfluidics by applying a dynamic flow to a bacterial-loaded mucus model and by monitoring E. coli growth and survival. The engineered models represent a step forward in the modelling of the mucus, since they can answer to different urgent needs such as a 3D structure, bioinspired properties and compatibility with dynamic system.
Collapse
Affiliation(s)
- Lorenzo Sardelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy.
| | - Francesco Briatico Vangosa
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Marta Merli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Anna Ziccarelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Sonja Visentin
- Molecular Biotechnology and Health Sciences Department, University of Torino, Torino, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy; Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici (ICS) Maugeri, IRCCS, Pavia, Italy
| | - Paola Petrini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| |
Collapse
|
19
|
Han K, Feng G, Li T, Deng Z, Zhang Z, Wang J, Yang X. Digestion Resistance of Soybean 7S Protein and Its Implications for Reinforcing the Gastric Mucus Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8776-8787. [PMID: 35802804 DOI: 10.1021/acs.jafc.2c02603] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Previous studies have found that soybean protein, especially soybean 7S protein (β-conglycinin), exhibits digestion resistance, but the mechanism of digestion resistance and its implications for human health are still unclear. Here, we show that the extracted soybean 7S protein contains both oligomer globulins and amyloid aggregates, while the gastric digested soybean 7S protein only contains amyloid aggregates and thus exhibits digestion resistance. An animal experiment shows that un-digestible soybean 7S protein effectively prevents aspirin-induced acute gastric mucosa damage. The impacts of un-digestible soybean 7S protein on gastric mucus barrier properties are investigated using quartz crystal microbalance with dissipation (QCM-D), Langmuir monolayer, and multiple particle tracking (MPT). Results show that these un-digestible protein aggregates can penetrate into gastric mucus, increase the viscosity and compactness of the mucin layer, and reinforce the gastric mucus barrier properties. The findings are helpful to understand that high consumption of non-fermented soybean foods is associated with a decreased risk of gastric cancer.
Collapse
Affiliation(s)
- Kaining Han
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510641, China
| | - Guangxin Feng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510641, China
| | - Tanghao Li
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510641, China
| | - Zhuoyao Deng
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510641, China
| | - Zhao Zhang
- Guangdong Longsee Biomedical Co.,Ltd., Guangzhou 510700, China
| | - Jinmei Wang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510641, China
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids, School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
20
|
Sharma A, Raman V, Lee J, Forbes NS. Microbial Imbalance Induces Inflammation by Promoting Salmonella Penetration through the Mucosal Barrier. ACS Infect Dis 2022; 8:969-981. [PMID: 35404574 DOI: 10.1021/acsinfecdis.1c00530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The balance of microbial species in the intestine must be maintained to prevent inflammation and disease. Healthy bacteria suppress infection by pathogens and prevent disorders such as inflammatory bowel diseases (IBDs). The role of mucus in the relation between pathogens and the intestinal microbiota is poorly understood. Here, we hypothesized that healthy bacteria inhibit infection by preventing pathogens from penetrating the mucus layer and that microbial imbalance leads to inflammation by promoting the penetration of the mucosal barrier. We tested this hypothesis with an in vitro model that contains mucus, an epithelial cell layer, and resident immune cells. We found that, unlike probiotic VSL#3 bacteria, Salmonella penetrated the mucosal layers and induced the production of interleukin-8 (IL-8) and tumor necrosis factor (TNF)-α. At ratios greater than 104:1, probiotic bacteria suppressed the growth and penetration of Salmonella and reduced the production of inflammatory cytokines. Counterintuitively, low densities of healthy bacteria increased both pathogen penetration and cytokine production. In all cases, mucus increased Salmonella penetration and the production of cytokines. These results suggest that mucus lessens the protective effect of probiotic bacteria by promoting barrier penetration. In this model, a more imbalanced microbial population caused infection and inflammation by selecting pathogens that are more invasive and immunogenic. Combined, the results suggest that the depletion of commensal bacteria or an insufficient dosage of probiotics could worsen an infection and cause increased inflammation. A better understanding of the interactions between pathogens, healthy microbes, and the mucosal barrier will improve the treatment of infections and inflammatory diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
21
|
Wang Y, Cui H, Esworthy T, Mei D, Wang Y, Zhang LG. Emerging 4D Printing Strategies for Next-Generation Tissue Regeneration and Medical Devices. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109198. [PMID: 34951494 DOI: 10.1002/adma.202109198] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/17/2021] [Indexed: 06/14/2023]
Abstract
The rapid development of 3D printing has led to considerable progress in the field of biomedical engineering. Notably, 4D printing provides a potential strategy to achieve a time-dependent physical change within tissue scaffolds or replicate the dynamic biological behaviors of native tissues for smart tissue regeneration and the fabrication of medical devices. The fabricated stimulus-responsive structures can offer dynamic, reprogrammable deformation or actuation to mimic complex physical, biochemical, and mechanical processes of native tissues. Although there is notable progress made in the development of the 4D printing approach for various biomedical applications, its more broad-scale adoption for clinical use and tissue engineering purposes is complicated by a notable limitation of printable smart materials and the simplistic nature of achievable responses possible with current sources of stimulation. In this review, the recent progress made in the field of 4D printing by discussing the various printing mechanisms that are achieved with great emphasis on smart ink mechanisms of 4D actuation, construct structural design, and printing technologies, is highlighted. Recent 4D printing studies which focus on the applications of tissue/organ regeneration and medical devices are then summarized. Finally, the current challenges and future perspectives of 4D printing are also discussed.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Fluid Power and Mechatronics Systems, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Deqing Mei
- State Key Laboratory of Fluid Power and Mechatronics Systems, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yancheng Wang
- State Key Laboratory of Fluid Power and Mechatronics Systems, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Electrical and Computer Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Medicine, The George Washington University, Washington, DC, 20052, USA
| |
Collapse
|
22
|
Marczynski M, Jiang K, Blakeley M, Srivastava V, Vilaplana F, Crouzier T, Lieleg O. Structural Alterations of Mucins Are Associated with Losses in Functionality. Biomacromolecules 2021; 22:1600-1613. [PMID: 33749252 DOI: 10.1021/acs.biomac.1c00073] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Commercial mucin glycoproteins are routinely used as a model to investigate the broad range of important functions mucins fulfill in our bodies, including lubrication, protection against hostile germs, and the accommodation of a healthy microbiome. Moreover, purified mucins are increasingly selected as building blocks for multifunctional materials, i.e., as components of hydrogels or coatings. By performing a detailed side-by-side comparison of commercially available and lab-purified variants of porcine gastric mucins, we decipher key molecular motifs that are crucial for mucin functionality. As two main structural features, we identify the hydrophobic termini and the hydrophilic glycosylation pattern of the mucin glycoprotein; moreover, we describe how alterations in those structural motifs affect the different properties of mucins-on both microscopic and macroscopic levels. This study provides a detailed understanding of how distinct functionalities of gastric mucins are established, and it highlights the need for high-quality mucins-for both basic research and the development of mucin-based medical products.
Collapse
Affiliation(s)
- Matthias Marczynski
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany.,Center for Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer Str. 8, 85748 Garching, Germany
| | - Kun Jiang
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden.,AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, 114 28 Stockholm, Sweden.,Department of Neuroscience, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Matthew Blakeley
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Vaibhav Srivastava
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Francisco Vilaplana
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Thomas Crouzier
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden.,AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, 114 28 Stockholm, Sweden.,Department of Neuroscience, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Oliver Lieleg
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany.,Center for Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer Str. 8, 85748 Garching, Germany
| |
Collapse
|
23
|
Li YL, Han KN, Feng GX, Wan ZL, Wang GS, Yang XQ. Salt reduction in bread via enrichment of dietary fiber containing sodium and calcium. Food Funct 2021; 12:2660-2671. [PMID: 33650606 DOI: 10.1039/d0fo03126g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The high intake of sodium and low intake of dietary fiber are two major dietary risk factors for preventable deaths worldwide, highlighting the need and implementations for developing health foods with low-salt/high-dietary fibers. Bread as a staple food contributes about 25% to the daily intake of sodium in many countries, and salt reduction in bread still remains a great technical challenge. In this study, we developed a simple method to reformulate the white bread in terms of reducing salt contents via dietary fiber fortification, while maintaining the taste and texture qualities. Low molecular weight water-extractable arabinoxylans (LMW-WEAX) as a soluble dietary fiber was first hydrated in salt water before dough mixing, leading to an inhomogeneous spatial distribution of sodium in bread and accelerating the release of sodium ions from crumbs, allowing 20% salt reduction in bread without impacting the salt perception. Data from the moisture content, crumb structure, water distribution, dough rheology and bread texture properties suggest that the pre-hydrated incorporation of LMW-WEAX mitigates the detrimental effect of dietary fiber on the dough and bread quality. The modulation of Ca2+ on the permeability of Na+ through the mucus layer and implication in salt enhancement of the bread were investigated. Results show that the pre-hydrated incorporation of WEAX containing Na+ and Ca2+ (1.0%) makes it possible to reduce 30% salt content in breads, which have implications in the large-scale production of low-salt/high-dietary fiber bread.
Collapse
Affiliation(s)
- Yan-Lei Li
- Laboratory of Food Proteins and Colloids, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | | | | | | | | | | |
Collapse
|
24
|
Sharma A, Raman V, Lee J, Forbes NS. Mucus blocks probiotics but increases penetration of motile pathogens and induces TNF-α and IL-8 secretion. Biotechnol Bioeng 2020; 117:2540-2555. [PMID: 32396232 PMCID: PMC7806204 DOI: 10.1002/bit.27383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/29/2022]
Abstract
The mucosal barrier in combination with innate immune system are the first line of defense against luminal bacteria at the intestinal mucosa. Dysfunction of the mucus layer and bacterial infiltration are linked to tissue inflammation and disease. To study host-bacterial interactions at the mucosal interface, we created an experimental model that contains luminal space, a mucus layer, an epithelial layer, and suspended immune cells. Reconstituted porcine small intestinal mucus formed an 880 ± 230 µm thick gel layer and had a porous structure. In the presence of mucus, sevenfold less probiotic and nonmotile VSL#3 bacteria transmigrated across the epithelial barrier compared to no mucus. The higher bacterial transmigration caused immune cell differentiation and increased the concentration of interleukin-8 (IL-8) and tumor necrosis factor-alpha (TNF-α; p < .01). Surprisingly, the mucus layer increased transmigration of pathogenic Salmonella and increased secretion of TNF-α and IL-8 (p < .05). Nonmotile, flagella knockout Salmonella had lower transmigration and caused lower IL-8 and TNF-α secretion (p < .05). These results demonstrate that motility enables pathogenic bacteria to cross the mucus and epithelial layers, which could lead to infection. Using an in vitro coculture platform to understand the interactions of bacteria with the intestinal mucosa has the potential to improve the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|