1
|
Bissoyi A, Gao Y, Tomás RMF, Kinney NLH, Whale TF, Guo Q, Gibson MI. Cryopreservation and Rapid Recovery of Differentiated Intestinal Epithelial Barrier Cells at Complex Transwell Interfaces Is Enabled by Chemically Induced Ice Nucleation. ACS APPLIED MATERIALS & INTERFACES 2024; 16. [PMID: 38671549 PMCID: PMC11082836 DOI: 10.1021/acsami.4c03931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Cell-based models, such as organ-on-chips, can replace and inform in vivo (animal) studies for drug discovery, toxicology, and biomedical science, but most cannot be banked "ready to use" as they do not survive conventional cryopreservation with DMSO alone. Here, we demonstrate how macromolecular ice nucleators enable the successful cryopreservation of epithelial intestinal models supported upon the interface of transwells, allowing recovery of function in just 7 days post-thaw directly from the freezer, compared to 21 days from conventional suspension cryopreservation. Caco-2 cells and Caco-2/HT29-MTX cocultures are cryopreserved on transwell inserts, with chemically induced ice nucleation at warmer temperatures resulting in increased cell viability but crucially retaining the complex cellular adhesion on the transwell insert interfaces, which other cryoprotectants do not. Trans-epithelial electrical resistance measurements, confocal microscopy, histology, and whole-cell proteomics demonstrated the rapid recovery of differentiated cell function, including the formation of tight junctions. Lucifer yellow permeability assays confirmed that the barrier functions of the cells were intact. This work will help solve the long-standing problem of transwell tissue barrier model storage, facilitating access to advanced predictive cellular models. This is underpinned by precise control of the nucleation temperature, addressing a crucial biophysical mode of damage.
Collapse
Affiliation(s)
- Akalabya Bissoyi
- Department
of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Yanan Gao
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ruben M. F. Tomás
- Cryologyx
Ltd, Venture Centre, University of Warwick
Science Park, Coventry CV4 7EZ, U.K.
| | - Nina L. H. Kinney
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Royal
Botanic Gardens Kew, Ardingly, West Sussex RH17 6TN, U.K.
| | - Thomas F. Whale
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- School
of Earth and Environment, University of
Leeds, Leeds LS2 9JT, U.K.
| | - Qiongyu Guo
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Matthew I. Gibson
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Division
of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess Street, Manchester M1 7DN, U.K.
| |
Collapse
|
2
|
Ran R, Muñoz Briones J, Jena S, Anderson NL, Olson MR, Green LN, Brubaker DK. Detailed survey of an in vitro intestinal epithelium model by single-cell transcriptomics. iScience 2024; 27:109383. [PMID: 38523788 PMCID: PMC10959667 DOI: 10.1016/j.isci.2024.109383] [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: 07/11/2023] [Revised: 12/01/2023] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
The co-culture of two adult human colorectal cancer cell lines, Caco-2 and HT29, on Transwell is commonly used as an in vitro gut mimic, yet the translatability of insights from such a system to adult human physiological contexts is not fully characterized. Here, we used single-cell RNA sequencing on the co-culture to obtain a detailed survey of cell type heterogeneity in the system and conducted a holistic comparison with human physiology. We identified the intestinal stem cell-, transit amplifying-, enterocyte-, goblet cell-, and enteroendocrine-like cells in the system. In general, the co-culture was fetal intestine-like, with less variety of gene expression compared to the adult human gut. Transporters for major types of nutrients were found in the majority of the enterocytes-like cells in the system. TLR 4 was not expressed in the sample, indicating that the co-culture model is incapable of mimicking the innate immune aspect of the human epithelium.
Collapse
Affiliation(s)
- Ran Ran
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Javier Muñoz Briones
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Purdue Interdisciplinary Life Science Program, West Lafayette, IN, USA
| | - Smrutiti Jena
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Nicole L. Anderson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Matthew R. Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Leopold N. Green
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Douglas K. Brubaker
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH, USA
| |
Collapse
|
3
|
Watson BE, Miles JA, Moss MA. Human in vitro blood barrier models: architectures and applications. Tissue Barriers 2024; 12:2222628. [PMID: 37339009 PMCID: PMC11042067 DOI: 10.1080/21688370.2023.2222628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023] Open
Abstract
Blood barriers serve as key points of transport for essential molecules as well as lines of defense to protect against toxins. In vitro modeling of these barriers is common practice in the study of their physiology and related diseases. This review describes a common method of using an adaptable, low cost, semipermeable, suspended membrane to experimentally model three blood barriers in the human body: the blood-brain barrier (BBB), the gut-blood barrier (GBB), and the air-blood barrier (ABB). The GBB and ABB both protect from the outside environment, while the BBB protects the central nervous system from potential neurotoxic agents in the blood. These barriers share several commonalities, including the formation of tight junctions, polarized cellular monolayers, and circulatory system contact. Cell architectures used to mimic barrier anatomy as well as applications to study function, dysfunction, and response provide an overview of the versatility enabled by these cultural systems.
Collapse
Affiliation(s)
| | - Julia A. Miles
- Biomedical Engineering Program, Univ of South Carolina, Columbia, SCUSA
| | - Melissa A. Moss
- Biomedical Engineering Program, Univ of South Carolina, Columbia, SCUSA
- Department of Chemical Engineering, Univ of South Carolina, Columbia, SCUSA
| |
Collapse
|
4
|
Bhattacharya S, Sangave PC, Belemkar S, Anjum MM. pH-Sensitive Nanoparticles of Epigallocatechin-3-Gallate in Enhanced Colorectal Cancer Therapy. Nanomedicine (Lond) 2024; 19:459-481. [PMID: 38223987 DOI: 10.2217/nnm-2023-0342] [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: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/16/2024] Open
Abstract
AIM Encapsulating epigallocatechin-3-gallate (EGCG) in pH-sensitive polymeric nanoparticles for targeted delivery of drugs could revolutionize colorectal cancer treatment. MATERIALS & METHODS Nanoparticles were synthesized to release drugs at colon pH. Dynamic light scattering measured their average diameter and ζ-potential, while differential scanning calorimetry and x-ray diffraction assessed EGCG encapsulation. RESULTS The nanoparticles showed stability and bioavailability in the gastrointestinal tract, efficiently encapsulating and releasing over 93% of EGCG at pH 7.2. They enhanced cytotoxicity against HT-29 cells and demonstrated antibacterial properties, increasing apoptosis and cell cycle arrest. CONCLUSION The study underscores the potential of nanoparticles in enhancing EGCG delivery for colorectal cancer therapy, aiming to minimize side effects and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Preeti Chidambar Sangave
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Sateesh Belemkar
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Vile Parle (W), Mumbai, 400056, Maharashtra, India
| | - Md Meraj Anjum
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, UP, 226025, India
| |
Collapse
|
5
|
Jakubczyk D, Leszczyńska K, Pacyga-Prus K, Kozakiewicz D, Kazana-Płuszka W, Gełej D, Migdał P, Kruszakin R, Zabłocka A, Górska S. What happens to Bifidobacterium adolescentis and Bifidobacterium longum ssp. longum in an experimental environment with eukaryotic cells? BMC Microbiol 2024; 24:60. [PMID: 38373929 PMCID: PMC10875879 DOI: 10.1186/s12866-023-03179-z] [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: 12/02/2022] [Accepted: 12/29/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND The impact of probiotic strains on host health is widely known. The available studies on the interaction between bacteria and the host are focused on the changes induced by bacteria in the host mainly. The studies determining the changes that occurred in the bacteria cells are in the minority. Within this paper, we determined what happens to the selected Bifidobacterium adolescentis and Bifidobacterium longum ssp. longum in an experimental environment with the intestinal epithelial layer. For this purpose, we tested the bacteria cells' viability, redox activity, membrane potential and enzymatic activity in different environments, including CaCo-2/HT-29 co-culture, cell culture medium, presence of inflammatory inductor (TNF-α) and oxygen. RESULTS We indicated that the external milieu impacts the viability and vitality of bacteria. Bifidobacterium adolescentis decrease the size of the live population in the cell culture medium with and without TNF-α (p < 0.001 and p < 0.01 respectively). In contrast, Bifidobacterium longum ssp. longum significantly increased survivability in contact with the eukaryotic cells and cell culture medium (p < 0.001). Bifidobacterium adolescentis showed significant changes in membrane potential, which was decreased in the presence of eukaryotic cells (p < 0.01), eukaryotic cells in an inflammatory state (p < 0.01), cell culture medium (p < 0.01) and cell culture medium with TNF-α (p < 0.05). In contrast, Bifidobacterium longum ssp. longum did not modulate membrane potential. Instead, bacteria significantly decreased the redox activity in response to milieus such as eukaryotic cells presence, inflamed eukaryotic cells as well as the culture medium (p < 0.001). The redox activity was significantly different in the cells culture medium vs the presence of eukaryotic cells (p < 0.001). The ability to β-galactosidase production was different for selected strains: Bifidobacterium longum ssp. longum indicated 91.5% of positive cells, whereas Bifidobacterium adolescentis 4.34% only. Both strains significantly reduced the enzyme production in contact with the eukaryotic milieu but not in the cell culture media. CONCLUSION The environmental-induced changes may shape the probiotic properties of bacterial strains. It seems that the knowledge of the sensitivity of bacteria to the external environment may help to select the most promising probiotic strains, reduce research costs, and contribute to greater reproducibility of the obtained probiotic effects.
Collapse
Affiliation(s)
- Dominika Jakubczyk
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland.
| | - Katarzyna Leszczyńska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Katarzyna Pacyga-Prus
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Dominika Kozakiewicz
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Wioletta Kazana-Płuszka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Dominika Gełej
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Paweł Migdał
- Inter-Departmental Laboratory of Instrumental Analysis and Preparation, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Roksana Kruszakin
- Inter-Departmental Laboratory of Instrumental Analysis and Preparation, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Agnieszka Zabłocka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
6
|
Kapse N, Pisu V, Dhakephalkar T, Margale P, Shetty D, Wagh S, Dagar S, Dhakephalkar PK. Unveiling the Probiotic Potential of Streptococcus thermophilus MCC0200: Insights from In Vitro Studies Corroborated with Genome Analysis. Microorganisms 2024; 12:347. [PMID: 38399752 PMCID: PMC10891967 DOI: 10.3390/microorganisms12020347] [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: 12/07/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Streptococcus thermophilus is widely used as a starter culture in the dairy industry and has garnered attention as a beneficial bacterium owing to its health-promoting functionalities in humans. In this study, the probiotic potential of S. thermophilus MCC0200 isolated from a dairy product was investigated through a combinatorial approach of in vitro and in silico studies. MCC0200 demonstrated the ability to survive harsh gastrointestinal (GI) transit, adhere to intestinal mucosa and exert health-promoting traits in in vitro studies. These findings were corroborated with in silico evidence, wherein, MCC0200 genome harboured genes associated with tolerance to GI conditions, intestinal adhesion and colonization. Genome mapping also highlighted the ability of MCC0200 to produce compounds advantageous for the host (folate, bacteriocins), to release antioxidant enzymes that can quench the free radicals (superoxide dismutase, NADH peroxidase), and to metabolize food components that can be harmful to sensitive people (lactose). MCC0200 also demonstrated a positive effect on reducing cholesterol levels, proving to be a potential candidate for food and pharmaceutical applications. The absence of transmissible antibiotic resistance genes and virulence genes underscored the generally regarded as safe (GRAS) nature of MCC0200. This study explored the potential of Streptococcus thermophilus for its probable applications as a probiotic beyond the dairy industry.
Collapse
Affiliation(s)
- Neelam Kapse
- Bioenergy Group, MACS-Agharkar Research Institute, Gopal Ganesh Agarkar Road, Pune 411004, Maharashtra, India; (N.K.); (V.P.); (D.S.)
| | - Vaidehi Pisu
- Bioenergy Group, MACS-Agharkar Research Institute, Gopal Ganesh Agarkar Road, Pune 411004, Maharashtra, India; (N.K.); (V.P.); (D.S.)
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Tanisha Dhakephalkar
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 & 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Prajakta Margale
- Bioenergy Group, MACS-Agharkar Research Institute, Gopal Ganesh Agarkar Road, Pune 411004, Maharashtra, India; (N.K.); (V.P.); (D.S.)
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Deepa Shetty
- Bioenergy Group, MACS-Agharkar Research Institute, Gopal Ganesh Agarkar Road, Pune 411004, Maharashtra, India; (N.K.); (V.P.); (D.S.)
| | - Shilpa Wagh
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 & 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Sumit Dagar
- Bioenergy Group, MACS-Agharkar Research Institute, Gopal Ganesh Agarkar Road, Pune 411004, Maharashtra, India; (N.K.); (V.P.); (D.S.)
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Prashant K. Dhakephalkar
- Bioenergy Group, MACS-Agharkar Research Institute, Gopal Ganesh Agarkar Road, Pune 411004, Maharashtra, India; (N.K.); (V.P.); (D.S.)
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| |
Collapse
|
7
|
Li L, Chen J, Zheng Y, Lane J, Hu R, Zhu J, Fu X, Huang Q, Liu F, Zhang B. Gastro-Intestinal Digested Bovine Milk Osteopontin Modulates Gut Barrier Biomarkers In Vitro. Mol Nutr Food Res 2024; 68:e2200777. [PMID: 38193251 DOI: 10.1002/mnfr.202200777] [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: 11/08/2022] [Revised: 06/28/2023] [Indexed: 01/10/2024]
Abstract
SCOPE Osteopontin (OPN) is a multifunctional protein naturally present in mammals' milk, associated with immune homeostasis and intestinal maturation. This study aims to investigate the protein digestion pattern and the cellular bioactivity of bovine milk OPN digesta in vitro. METHODS AND RESULTS A modified INFOGEST static in vitro infant digestion protocol and a Caco-2/HT-29 co-culture cell model are employed to evaluate the digestion properties and the anti-inflammatory effects of OPN. OPN is resistant to gastric hydrolysis but degraded into large peptides during intestinal digestion. Its 10 kDa digesta permeate with predicted extensive bioactivities protects the co-culture cell model from the inflammation-induced dysfunction by dose-dependently recovering the expression of occludin, claudin-3, and ZO-1. Low dosage of OPN significantly decreases the production of IL-8 and IL-6, and downregulates the mRNA and protein expression of MyD88, NF-κB p65, and IκB-α, whereas a high dose evokes a mild pro-inflammatory response. Interestingly, anti-inflammatory effect of OPN digesta is stronger than lactoferrin and whey protein concentrate counterparts. CONCLUSION The findings demonstrate that the bioactive peptides released from in vitro infant gastrointestinal digestion of bovine milk OPN alleviates intestinal epithelial cell inflammation by inhibiting NF-κB pathway activation and potentiates the barrier function of the intestinal epithelium.
Collapse
Affiliation(s)
- Lu Li
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
| | - Juchun Chen
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Yuxing Zheng
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Jonathan Lane
- H&H Group, H&H Research, Global Research and Technology Centre, P61 K202 Co, Cork, Ireland
| | - Ruibiao Hu
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Jianzhong Zhu
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
| | - Xiong Fu
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
| | - Qiang Huang
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
| | - Feitong Liu
- H&H Group, H&H Research, China Research and Innovation Center, Guangzhou, 510700, China
| | - Bin Zhang
- School of Food Science and Engineering, Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health, South China University of Technology, Guangzhou, 510640, China
- Sino-Singapore International Research Institute, Guangzhou, 510555, China
| |
Collapse
|
8
|
Fan S, Zhou Y, Zhao Y, Daglia M, Zhang J, Zhu Y, Bai J, Zhu L, Xiao X. Metabolomics reveals the effects of Lactiplantibacillus plantarum dy-1 fermentation on the lipid-lowering capacity of barley β-glucans in an in vitro model of gut-liver axis. Int J Biol Macromol 2023; 253:126861. [PMID: 37714241 DOI: 10.1016/j.ijbiomac.2023.126861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/11/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
Bioactive polysaccharides known as the biological response modifiers, can directly interact with intestinal epithelium cells (IEC) and regulate key metabolic processes such as lipid metabolism. Here, the coculture of Caco-2/HT29 monolayer (>400 Ω × cm2) and HepG2 cells was developed to mimic the gut-liver interactions. This system was used to investigate the effects of raw and fermented barley β-glucans (RBG and FBG) on lipid metabolism by directly interacting with IEC. Both RBG and FBG significantly and consistently reduced the lipid droplets and triacylglycerol levels in monoculture and coculture of HepG2 overloaded with oleic acid. Notably, FBG significantly and distinctly elevated PPARα (p < 0.05) and PPARα-responsive ACOX-1 (p < 0.01) gene expressions, promoting lipid degradation in cocultured HepG2. Moreover, the metabolomics analyses revealed that FBG had a unique impact on extracellular metabolites, among them, the differential metabolite thiomorpholine 3-carboxylate was significantly and strongly correlated with PPARα (r = -0.68, p < 0.01) and ACOX-1 (r = -0.76, p < 0.01) expression levels. Taken together, our findings suggest that FBG-mediated gut-liver interactions play a key role in its lipid-lowering effects that are superior to those of RBG. These results support the application of Lactiplantibacillus fermentation for improving hypolipidemic outcomes.
Collapse
Affiliation(s)
- Songtao Fan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yurong Zhou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yansheng Zhao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
| | - Jiayan Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Ying Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Juan Bai
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Lin Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
9
|
Bendinelli P, De Noni I, Cattaneo S, Silvetti T, Brasca M, Piazzalunga F, Donetti E, Ferraretto A. Surface layer proteins from Lactobacillus helveticus ATCC® 15009™ affect the gut barrier morphology and function. Tissue Barriers 2023:2289838. [PMID: 38059583 DOI: 10.1080/21688370.2023.2289838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
Paraprobiotics and postbiotics represent a valid alternative to probiotic strains for ameliorating and preserving a healthy intestinal epithelial barrier (IEB). The present study investigated the effects of surface layer proteins (S-layer) of the dairy strain Lactobacillus helveticus ATCC® 15009™ (Lb ATCC® 15009™), as paraprobiotic, on the morpho-functional modulation of IEB in comparison to live or heat-inactivated Lb ATCC® 15009™ in an in vitro co-culture of Caco-2/HT-29 70/30 cells. Live or heat-inactivated Lb ATCC® 15009™ negatively affected transepithelial electrical resistance (TEER) and paracellular permeability, and impaired the distribution of Claudin-1, a tight junction (TJ) transmembrane protein, as detected by immunofluorescence (IF). Conversely, the addition of the S-layer improved TEER and decreased permeability in physiological conditions in co-cultures with basal TEER lower than 50 ohmcm2, indicative of a more permeable physiological IEB known as leaky gut. Transmission electron microscopy (TEM) and IF analyses suggested that the S-layer induces a structural TJ rearrangement and desmosomes' formation. S-layer also restored TEER and permeability in the presence of LPS, but not of a mixture of pro-inflammatory cytokines (TNF-α plus IFN-γ). IF analyses showed an increase in Claudin-1 staining when LPS and S-layer were co-administered with respect to LPS alone; in addition, the S-layer counteracted the reduction of alkaline phosphatase detoxification activity and the enhancement of pro-inflammatory interleukin-8 release both induced by LPS. Altogether, these data corroborate a paraprobiotic role of S-layer from Lb ATCC® 15009™ as a possible candidate for therapeutic and prophylactic uses in conditions related to gastrointestinal health and correlated with extra-intestinal disorders.
Collapse
Affiliation(s)
- Paola Bendinelli
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Ivano De Noni
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Stefano Cattaneo
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Tiziana Silvetti
- Institute of Sciences of Food Production, National Research Council (CNR-ISPA), Milan, Italy
| | - Milena Brasca
- Institute of Sciences of Food Production, National Research Council (CNR-ISPA), Milan, Italy
| | | | - Elena Donetti
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Anita Ferraretto
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Galeazzi-Sant'Ambrogio, Milan, Italy
| |
Collapse
|
10
|
Rebai Y, Wagner L, Gnaien M, Hammer ML, Kapitan M, Niemiec MJ, Mami W, Mosbah A, Messadi E, Mardassi H, Vylkova S, Jacobsen ID, Znaidi S. Escherichia coli Nissle 1917 Antagonizes Candida albicans Growth and Protects Intestinal Cells from C. albicans-Mediated Damage. Microorganisms 2023; 11:1929. [PMID: 37630490 PMCID: PMC10457924 DOI: 10.3390/microorganisms11081929] [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: 04/22/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 08/27/2023] Open
Abstract
Candida albicans is a pathobiont of the gastrointestinal tract. It can contribute to the diversity of the gut microbiome without causing harmful effects. When the immune system is compromised, C. albicans can damage intestinal cells and cause invasive disease. We hypothesize that a therapeutic approach against C. albicans infections can rely on the antimicrobial properties of probiotic bacteria. We investigated the impact of the probiotic strain Escherichia coli Nissle 1917 (EcN) on C. albicans growth and its ability to cause damage to intestinal cells. In co-culture kinetic assays, C. albicans abundance gradually decreased over time compared with C. albicans abundance in the absence of EcN. Quantification of C. albicans survival suggests that EcN exerts a fungicidal activity. Cell-free supernatants (CFS) collected from C. albicans-EcN co-culture mildly altered C. albicans growth, suggesting the involvement of an EcN-released compound. Using a model of co-culture in the presence of human intestinal epithelial cells, we further show that EcN prevents C. albicans from damaging enterocytes both distantly and through direct contact. Consistently, both C. albicans's filamentous growth and microcolony formation were altered by EcN. Taken together, our study proposes that probiotic-strain EcN can be exploited for future therapeutic approaches against C. albicans infections.
Collapse
Affiliation(s)
- Yasmine Rebai
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
| | - Lysett Wagner
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mayssa Gnaien
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
| | - Merle L. Hammer
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mario Kapitan
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
- Center for Sepsis Control and Care, 07747 Jena, Germany
| | - Maria Joanna Niemiec
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Wael Mami
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Amor Mosbah
- Laboratory of Biotechnology and Bio-Geo Resources Valorization (LR11ES31), Higher Institute of Biotechnology of Sidi Thabet (ISBST), University of Manouba, Tunis 2010, Tunisia
| | - Erij Messadi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Helmi Mardassi
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
| | - Slavena Vylkova
- Septomics Research Center, Friedrich Schiller University, 07745 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Ilse D. Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
- Center for Sepsis Control and Care, 07747 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| | - Sadri Znaidi
- Laboratoire de Microbiologie Moléculaire, Vaccinologie et Développement Biotechnologique (LR16IPT01), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1068, Tunisia; (Y.R.)
- Institut Pasteur, Institut National de la Recherche Agronomique (INRA), Département Mycologie, Unité Biologie et Pathogénicité Fongiques, 75015 Paris, France
| |
Collapse
|
11
|
Joshua Ashaolu T, Lee CC, Opeolu Ashaolu J, Pourjafar H, Jafari SM. Metal-binding peptides and their potential to enhance the absorption and bioavailability of minerals. Food Chem 2023; 428:136678. [PMID: 37418874 DOI: 10.1016/j.foodchem.2023.136678] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/07/2023] [Accepted: 06/18/2023] [Indexed: 07/09/2023]
Abstract
Minerals including calcium, iron, zinc, magnesium, and copper have several human nutritional functions due to their metabolic activities. Body tissues require sufficient levels of a variety of micronutrients to maintain their health. To achieve these micronutrient needs, dietary consumption must be adequate. Dietary proteins may regulate the biological functions of the body in addition to acting as nutrients. Some peptides encoded in the native protein sequences are primarily responsible for the absorption and bioavailability of minerals in physiological functions. Metal-binding peptides (MBPs) were discovered as potential agents for mineral supplements. Nevertheless, sufficient studies on how MBPs affect the biological functions of minerals are lacking. The hypothesis is that the absorption and bioavailability of minerals are significantly influenced by peptides, and these properties are further enhanced by the configuration and attribute of the metal-peptide complex. In this review, the production of MBPs is discussed using various key parameters such as the protein sources and amino acid residues, enzymatic hydrolysis, purification, sequencing and synthesis and in silico analysis of MBPs. The mechanisms of metal-peptide complexes as functional food ingredients are elucidated, including metal-peptide ratio, precursors and ligands, complexation reaction, absorbability and bioavailability. Finally, the characteristics and application of different metal-peptide complexes are also described.
Collapse
Affiliation(s)
- Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang 550000, Viet Nam; Faculty of Medicine, Duy Tan University, Da Nang 550000, Viet Nam
| | - Chi Ching Lee
- Istanbul Sabahattin Zaim University, Faculty of Engineering and Natural Sciences, Department of Food Engineering, Istanbul, Turkey
| | - Joseph Opeolu Ashaolu
- Department of Public Health, Faculty of Basic Medical Sciences, Redeemers University, PMB 230, Ede, Osun State, Nigeria
| | - Hadi Pourjafar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran.
| |
Collapse
|
12
|
Liu J, Fu L, Yin F, Yin L, Song X, Guo H, Liu J. Diosmetin Maintains Barrier Integrity by Reducing the Expression of ABCG2 in Colonic Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37269551 DOI: 10.1021/acs.jafc.3c00912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Crohn's disease (CD) is a relapsing and chronic inflammatory bowel disease. Recent advances have highlighted that dysfunction of the barrier function formed by a polarized monolayer of columnar epithelial cells plays a crucial role in the pathophysiology of CD. At present, we reported that diosmetin increased cell viability by reducing the levels of TNFα and IL-6 in lipopolysaccharide (LPS)-treated colonic epithelial Caco-2 cells. Meanwhile, diosmetin conferred a direct effect on maintaining barrier integrity by reducing epithelial permeability and increasing the expression of proteins associated with tight junctions, including zonula occludens-l (ZO-1), occludin, and claudin-1, in LPS-treated Caco-2 cells and in 2,4,6-trinitrobenzene sulfonic acid-induced CD mice. Additionally, diosmetin decreased the protein content of adenosine triphosphate-binding cassette efflux transporter G2 (ABCG2) in vitro and in vivo. Over-expression of ABCG2 had an important impact on the epithelial permeability and barrier-related protein levels induced by LPS in Caco-2 cells. At the same time, Ko143, a specific ABCG2 inhibitor, dramatically enhanced the role of diosmetin in ZO-1 and occludin proteins in LPS-treated Caco-2 cells. Mechanically, diosmetin significantly attenuated the role of LPS in the phosphorylation of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), phosphatidylinositol-3-kinase (PI3K)/protein kinase B (PKB/AKT), and cAMP-response element binding protein (CREB) in Caco-2 cells. The AMPK inhibitor Compound C obviously prevented the effect of diosmetin on ZO-1 and occludin expression in LPS-treated Caco-2 cells. Taken together, the results of this study suggest that AMPK/AKT/CREB-mediated ABCG2 expression plays a crucial role in diosmetin, improving the barrier dysfunction in CD.
Collapse
Affiliation(s)
- Jinfan Liu
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Lu Fu
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Fei Yin
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, People's Republic of China
| | - Li Yin
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, People's Republic of China
| | - Xiaomei Song
- Department of Gastroenterology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401147, People's Republic of China
| | - Hong Guo
- Department of Gastroenterology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401147, People's Republic of China
| | - Jianhui Liu
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, People's Republic of China
| |
Collapse
|
13
|
Romero-Arguelles R, Tamez-Guerra P, González-Ochoa G, Romo-Sáenz CI, Gomez-Flores R, Flores-Mendoza L, Aros-Uzarraga E. Bifidobacterium longum and Chlorella sorokiniana Improve the IFN Type I-Mediated Antiviral Response in Rotavirus-Infected Cells. Microorganisms 2023; 11:1237. [PMID: 37317211 DOI: 10.3390/microorganisms11051237] [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: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 06/16/2023] Open
Abstract
Probiotics are effective to treat or prevent gastrointestinal infections, and microalgae have demonstrated important health-promoting effects and in some cases function as prebiotics. In this regard, the anti-rotavirus effect of Bifidobacterium longum and Chlorella sorokiniana by reducing viral infectivity is well known. However, their effect on immune response against rotavirus has not yet been investigated. Therefore, the aim of this study was to determine the role of Bifidobacterium longum and/or Chlorella sorokiniana in influencing an IFN type I-mediated antiviral response in rotavirus-infected cells. In pre-infection experiments, HT-29 cells were treated with B. longum and C. sorokiniana alone or in combination, followed by rotavirus infection, whereas in post-infection assays, HT-29 cells were treated after infection. The cells' mRNA was then purified to determine the relative expression level of IFN-α, IFN-β, and precursors of interferons such as RIG-I, IRF-3, and IRF-5 by qPCR. We showed that combination of B. longum and C. sorokiniana significantly increased IFN-α levels in pre-infection and IFN-β in post-infection assays, as compared with individual effects. Results indicate that B. longum, C. sorokiniana, or their combination improve cellular antiviral immune response.
Collapse
Affiliation(s)
- Ricardo Romero-Arguelles
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
| | - Patricia Tamez-Guerra
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
| | - Guadalupe González-Ochoa
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Navojoa 85880, Mexico
| | - César I Romo-Sáenz
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
| | - Ricardo Gomez-Flores
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
| | - Lilian Flores-Mendoza
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Navojoa 85880, Mexico
| | - Elizama Aros-Uzarraga
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Navojoa 85880, Mexico
| |
Collapse
|
14
|
Qi Y, Yu L, Tian F, Zhao J, Zhai Q. In vitro models to study human gut-microbiota interactions: Applications, advances, and limitations. Microbiol Res 2023; 270:127336. [PMID: 36871313 DOI: 10.1016/j.micres.2023.127336] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
In vitro models of the human gut help compensate for the limitations of animal models in studying the human gut-microbiota interaction and are indispensable in the clarification the mechanism of microbial action or in the high-throughput screening and functional evaluation of probiotics. The development of these models constitutes a rapidly developing field of research. From 2D1 to 3D2 and from simple to complex, several in vitro cell and tissue models have been developed and continuously improved. In this review, we categorized and summarized these models and described their development, applications, advances, and limitations by using specific examples. We also highlighted the best ways to select an appropriate in vitro model, and we also discussed which variables to consider when imitating microbial and human gut epithelial interactions.
Collapse
Affiliation(s)
- Yuli Qi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| |
Collapse
|
15
|
Michael D, Kerry-Smith J, Webberley T, Murphy K, Plummer S, Parry L, Marchesi J. Does flow culture impact upon gut-probiotic interactions: A comparison with static culture. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
|
16
|
Osmond M, Korthals E, Zimmermann CJ, Roth EJ, Marr DW, Neeves KB. Magnetically Powered Chitosan Milliwheels for Rapid Translation, Barrier Function Rescue, and Delivery of Therapeutic Proteins to the Inflamed Gut Epithelium. ACS OMEGA 2023; 8:11614-11622. [PMID: 37008083 PMCID: PMC10061643 DOI: 10.1021/acsomega.3c00886] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
Inflammatory bowel disease (IBD) is mediated by an overexpression of tumor necrosis factor-α (TNF) by mononuclear cells in the intestinal mucosa. Intravenous delivery of neutralizing anti-TNF antibodies can cause systemic immunosuppression, and up to one-third of people are non-responsive to treatment. Oral delivery of anti-TNF could reduce adverse effects; however, it is hampered by antibody degradation in the harsh gut environment during transit and poor bioavailability. To overcome these shortcomings, we demonstrate magnetically powered hydrogel particles that roll along mucosal surfaces, provide protection from degradation, and sustain the local release of anti-TNF. Iron oxide particles are embedded into a cross-linked chitosan hydrogel and sieved to produce 100-200 μm particles called milliwheels (m-wheels). Once loaded with anti-TNF, these m-wheels release 10 to 80% of their payload over 1 week at a rate that depends on the cross-linking density and pH. A rotating magnetic field induces a torque on the m-wheels that results in rolling velocities greater than 500 μm/s on glass and mucus-secreting cells. The permeability of the TNF-challenged gut epithelial cell monolayers was rescued in the presence of anti-TNF carrying m-wheels, which both neutralized the TNF and created an impermeable patch over leaky cell junctions. With the ability to translate over mucosal surfaces at high speed, provide sustained release directly to the inflamed epithelium, and provide barrier rescue, m-wheels demonstrate a potential strategy to deliver therapeutic proteins for the treatment of IBD.
Collapse
Affiliation(s)
- Matthew
J. Osmond
- Department
of Bioengineering, University of Colorado
Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department
of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Elizabeth Korthals
- Department
of Bioengineering, University of Colorado
Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Coy J. Zimmermann
- Department
of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Eric J. Roth
- Department
of Bioengineering, University of Colorado
Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - David W.M. Marr
- Department
of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Keith B. Neeves
- Department
of Bioengineering, University of Colorado
Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department
of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
17
|
Schimpel C, Passegger C, Egger S, Tam-Amersdorfer C, Strobl H. A novel 3D cell culture model to study the human small intestinal immune landscape. Eur J Immunol 2023; 53:e2250131. [PMID: 36527196 DOI: 10.1002/eji.202250131] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/21/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Several subsets of mononuclear phagocytes and DCs (MDC) populate the small intestine (SI), and these cells reportedly exert specialized functions in anti-microbial immunity and tolerance. Given the specialized phenotype of these cells, differing from other MDC family members, including their putative circulating blood precursors, local intestinal factors play key instructive roles in their differentiation. We designed an SI cell culture model composed of three intestinal epithelial cell (IEC) types, including absorptive enterocytes (E cells), antigen delivering microfold (M) cells, and mucus-producing goblet (G) cells plus T lymphocytes and soluble B cell-derived factors. This model was used to study the differentiation fate of CD34+ hematopoietic progenitor cell-derived monocyte/DC precursors. Progeny cells can be analyzed after a 3-week co-culture period, mimicking the physiologic turn-over time of intestinal MDC. A dominant monocyte differentiation pathway was suppressed, in favor of partial differentiation along DC and macrophage pathways, with low percentages of cells acquired DC or macrophage markers. Moreover, E and G cells play opposing roles in CX3CR1+ vs CD103dim cell differentiation, indicating that both together might counter-balance M/DC differentiation. Thus, SI epithelial cells suppress M/DC differentiation, supporting a key role for exogenous factors in M/DC differentiation.
Collapse
Affiliation(s)
- Christa Schimpel
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Christina Passegger
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Simone Egger
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Herbert Strobl
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| |
Collapse
|
18
|
Yoshinaga N, Zhou JK, Xu C, Quek CH, Zhu Y, Tang D, Hung LY, Najjar SA, Shiu CYA, Margolis KG, Lao YH, Leong KW. Phenylboronic Acid-Functionalized Polyplexes Tailored to Oral CRISPR Delivery. NANO LETTERS 2023; 23:757-764. [PMID: 36648291 PMCID: PMC10375565 DOI: 10.1021/acs.nanolett.2c02306] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Effective delivery of the CRISPR-Cas9 components is crucial to realizing the therapeutic potential. Although many delivery approaches have been developed for this application, oral delivery has not been explored due to the degradative nature of the gastrointestinal tract. For this issue, we developed a series of novel phenylboronic acid (PBA)-functionalized chitosan-polyethylenimine (CS-PEI) polymers for oral CRISPR delivery. PBA functionalization equipped the polyplex with higher stability, smooth transport across the mucus, and efficient endosomal escape and cytosolic unpackaging in the cells. From a library of 12 PBA-functionalized CS-PEI polyplexes, we identified a formulation that showed the most effective penetration in the intestinal mucosa after oral gavage to mice. The optimized formulation performed feasible CRISPR-mediated downregulation of the target protein and reduction in the downstream cholesterol. As the first oral CRISPR carrier, this study suggests the potential of addressing the needs of both local and systemic editing in a patient-compliant manner.
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Joyce K Zhou
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Chai Hoon Quek
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Yuefei Zhu
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Ding Tang
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Lin Yung Hung
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York 10032, United States
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York 10010, United States
| | - Sarah A Najjar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York 10032, United States
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York 10010, United States
| | - Chin Ying Angela Shiu
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Kara Gross Margolis
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York 10032, United States
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York 10010, United States
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
- Department of Systems Biology, Columbia University Medical Center, New York, New York 10032, United States
| |
Collapse
|
19
|
Differential Effects of Oligosaccharides, Antioxidants, Amino Acids and PUFAs on Heat/Hypoxia-Induced Epithelial Injury in a Caco-2/HT-29 Co-Culture Model. Int J Mol Sci 2023; 24:ijms24021111. [PMID: 36674626 PMCID: PMC9861987 DOI: 10.3390/ijms24021111] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
(1) Exposure of intestinal epithelial cells to heat and hypoxia causes a (heat) stress response, resulting in the breakdown of epithelial integrity. There are indications that several categories of nutritional components have beneficial effects on maintaining the intestinal epithelial integrity under stress conditions. This study evaluated the effect of nine nutritional components, including non-digestible oligosaccharides (galacto-oligosaccharides (GOS), fructo-oligosaccharides (FOS), chitosan oligosaccharides (COS)), antioxidants (α-lipoic acid (ALA), resveratrol (RES)), amino acids (l-glutamine (Glu), l-arginine (Arg)) and polyunsaturated fatty acids (PUFAs) (docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)), on heat/hypoxia-induced epithelial injury. (2) Two human colonic cell lines, Caco-2 and HT-29, were co-cultured and pre-treated with the nutritional components for 48 h. After pre-treatment, the cells were exposed to heat/hypoxia (42 °C, 5% O2) for 2 h. Epithelial integrity was evaluated by measuring trans-epithelial electrical resistance (TEER), paracellular Lucifer Yellow (LY) permeability, and tight junction (TJ) protein expression. Heat stress and oxidative stress levels were evaluated by determining heat-shock protein-70 (HSP-70) expression and the concentration of the lipid peroxidation product malondialdehyde (MDA). (3) GOS, FOS, COS, ALA, RES, Arg, and EPA presented protective effects on epithelial damage in heat/hypoxia-exposed Caco-2/HT-29 cells by preventing the decrease in TEER, the increase in LY permeability, and/or decrease in TJ proteins zonula occludens-1 (ZO-1) and claudin-3 expression. COS, RES, and EPA demonstrated anti-oxidative stress effects by suppressing the heat/hypoxia-induced MDA production, while Arg further elevated the heat/hypoxia-induced increase in HSP-70 expression. (4) This study indicates that various nutritional components have the potential to counteract heat/hypoxia-induced intestinal injury and might be interesting candidates for future in vivo studies and clinical trials in gastrointestinal disorders related to heat stress and hypoxia.
Collapse
|
20
|
El-Harakeh M, Saliba J, Sharaf Aldeen K, Haidar M, El Hajjar L, Awad MK, Hashash JG, Shirinian M, El-Sabban M. Expression of the methylcytosine dioxygenase ten-eleven translocation-2 and connexin 43 in inflammatory bowel disease and colorectal cancer. World J Gastroenterol 2022; 28:5845-5864. [PMID: 36353202 PMCID: PMC9639657 DOI: 10.3748/wjg.v28.i40.5845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) constitutes a substantial risk factor for colorectal cancer. Connexin 43 (Cx43) is a protein that forms gap junction (GJ) complexes involved in intercellular communication, and its expression is altered under pathological conditions, such as IBD and cancer. Recent studies have implicated epigenetic processes modulating DNA methylation in the pathogenesis of diverse inflammatory and malignant diseases. The ten-eleven translocation-2 (TET-2) enzyme catalyzes the demethylation, hence, regulating the activity of various cancer-promoting and tumor-suppressor genes.
AIM To investigate Cx43 and TET-2 expression levels and presence of 5-hydroxymethylcytosine (5-hmC) marks under inflammatory conditions both in vitro and in vivo.
METHODS TET-2 expression was evaluated in parental HT-29 cells and in HT-29 cells expressing low or high levels of Cx43, a putative tumor-suppressor gene whose expression varies in IBD and colorectal cancer, and which has been implicated in the inflammatory process and in tumor onset. The dextran sulfate sodium-induced colitis model was reproduced in BALB/c mice to evaluate the expression of TET-2 and Cx43 under inflammatory conditions in vivo. In addition, archived colon tissue sections from normal, IBD (ulcerative colitis), and sporadic colon adenocarcinoma patients were obtained and evaluated for the expression of TET-2 and Cx43. Expression levels were reported at the transcriptional level by quantitative real-time polymerase chain reaction, and at the translational level by Western blotting and immunofluorescence.
RESULTS Under inflammatory conditions, Cx43 and TET-2 expression levels increased compared to non-inflammatory conditions. TET-2 upregulation was more pronounced in Cx43-deficient cells. Moreover, colon tissue sections from normal, ulcerative colitis, and sporadic colon adenocarcinoma patients corroborated that Cx43 expression increased in IBD and decreased in adenocarcinoma, compared to tissues from non-IBD subjects. However, TET-2 expression and 5-hmC mark levels decreased in samples from patients with ulcerative colitis or cancer. Cx43 and TET-2 expression levels were also investigated in an experimental colitis mouse model. Interestingly, mice exposed to carbenoxolone (CBX), a GJ inhibitor, had upregulated TET-2 levels. Collectively, these results show that TET-2 levels and activity increased under inflammatory conditions, in cells downregulating gap junctional protein Cx43, and in colon tissues from mice exposed to CBX.
CONCLUSION These results suggest that TET-2 expression levels, as well as Cx43 expression levels, are modulated in models of intestinal inflammation. We hypothesize that TET-2 may demethylate genes involved in inflammation and tumorigenesis, such as Cx43, potentially contributing to intestinal inflammation and associated carcinogenesis.
Collapse
Affiliation(s)
- Mohammad El-Harakeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
- UR GPF Laboratory of Biodiversity and Functional Genomics, Faculty of Science, Université Saint-Joseph de Beyrouth, Beirut 1107, Lebanon
| | - Jessica Saliba
- Department of Biology, Faculty of Sciences, Lebanese University, Beirut 1533, Lebanon
- Department of Public Health, Faculty of Health Sciences, University of Balamand, Dekwaneh, Sin el Fil 1552, Lebanon
| | - Kawthar Sharaf Aldeen
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - May Haidar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Layal El Hajjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Mireille Kallassy Awad
- UR GPF Laboratory of Biodiversity and Functional Genomics, Faculty of Science, Université Saint-Joseph de Beyrouth, Beirut 1107, Lebanon
| | - Jana G Hashash
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Margret Shirinian
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
21
|
Rudolph SE, Longo BN, Tse MW, Houchin MR, Shokoufandeh MM, Chen Y, Kaplan DL. Crypt-Villus Scaffold Architecture for Bioengineering Functional Human Intestinal Epithelium. ACS Biomater Sci Eng 2022; 8:4942-4955. [PMID: 36191009 PMCID: PMC10379436 DOI: 10.1021/acsbiomaterials.2c00851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Crypt-villus architecture in the small intestine is crucial for the structural integrity of the intestinal epithelium and maintenance of gut homeostasis. We utilized three-dimensional (3D) printing and inverse molding techniques to form three-dimensional (3D) spongy scaffold systems that resemble the intestinal crypt-villus microarchitecture. The scaffolds consist of silk fibroin protein with curved lumens with rows of protruding villi with invaginating crypts to generate the architecture. Intestinal cell (Caco-2, HT29-MTX) attachment and growth, as well as long-term culture support were demonstrated with cell polarization and tissue barrier properties compared to two-dimensional (2D) Transwell culture controls. Further, physiologically relevant oxygen gradients were generated in the 3D system. The various advantages of this system may be ascribed to the more physiologically relevant 3D environment, offering a system for the exploration of disease pathogenesis, host-microbiome interactions, and therapeutic discovery.
Collapse
Affiliation(s)
- Sara E Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Brooke N Longo
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Megan W Tse
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Megan R Houchin
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Mina M Shokoufandeh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
22
|
McCright J, Sinha A, Maisel K. Generating an In Vitro Gut Model with Physiologically Relevant Biophysical Mucus Properties. Cell Mol Bioeng 2022; 15:479-491. [PMID: 36444342 PMCID: PMC9700528 DOI: 10.1007/s12195-022-00740-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/15/2022] [Indexed: 11/03/2022] Open
Abstract
Introduction Gastrointestinal (GI) in vitro models have received lasting attention as an effective tool to model drug and nutrient absorption, study GI diseases, and design new drug delivery vehicles. A complete model of the GI epithelium should at a minimum include the two key functional components of the GI tract: mucus and the underlying epithelium. Mucus plays a key role in protecting and lubricating the GI tract, poses a barrier to orally administered therapies and pathogens, and serves as the microenvironment for the GI microbiome. These functions are reliant on the biophysical material properties of the mucus produced, including viscosity and pore size. Methods In this study, we generated in vitro models containing Caco-2 enterocyte-like cells and HT29-MTX goblet-like cells and determined the effects of coculture and mucus layer on epithelial permeability and biophysical properties of mucus using multiple particle tracking (MPT). Results We found that mucus height increased as the amount of HT29-MTX goblet-like cells increased. Additionally, we found that increasing the amount of HT29-MTX goblet-like cells within culture corresponded to an increase in mucus pore size and mucus microviscosity, measured using MPT. When compared to ex vivo mucus samples from mice and pigs, we found that a 90:10 ratio of Caco-2:HT29-MTX coculture displayed similar mucus pore size to porcine jejunum and that the mucus produced from 90:10 and 80:20 ratios of cells shared mechanical properties to porcine jejunum and ileum mucus. Conclusions GI coculture models are valuable tools in simulating the mucus barrier and can be utilized for a variety of applications including the study of GI diseases, food absorption, or therapeutic development.
Collapse
Affiliation(s)
- Jacob McCright
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Arnav Sinha
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
23
|
Pitsalidis C, van Niekerk D, Moysidou CM, Boys AJ, Withers A, Vallet R, Owens RM. Organic electronic transmembrane device for hosting and monitoring 3D cell cultures. SCIENCE ADVANCES 2022; 8:eabo4761. [PMID: 36112689 PMCID: PMC9481123 DOI: 10.1126/sciadv.abo4761] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
3D cell models have made strides in the past decades in response to failures of 2D cultures to translate targets during the drug discovery process. Here, we report on a novel multiwell plate bioelectronic platform, namely, the e-transmembrane, capable of supporting and monitoring complex 3D cell architectures. Scaffolds made of PEDOT:PSS [poly(3,4-ethylenedioxythiophene):polystyrene sulfonate] are microengineered to function as separating membranes for compartmentalized cell cultures, as well as electronic components for real-time in situ recordings of cell growth and function. Owing to the high surface area-to-volume ratio, the e-transmembrane allows generation of deep, stratified tissues within the porous bulk and cell polarization at the apico-basal domains. Impedance spectroscopy measurements carried out throughout the tissue growth identified signatures from different cellular systems and allowed extraction of critical functional parameters. This platform has the potential to become a universal tool for biologists for the next generation of high-throughput drug screening assays.
Collapse
Affiliation(s)
- Charalampos Pitsalidis
- Department of Physics and Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, P. O. Box 127788, Abu Dhabi, UAE
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Douglas van Niekerk
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Alexander J. Boys
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Aimee Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | | | - Róisín M. Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| |
Collapse
|
24
|
Moysidou CM, Withers AM, Nisbet AJ, Price DRG, Bryant CE, Cantacessi C, Owens RM. Investigation of Host-Microbe-Parasite Interactions in an In Vitro 3D Model of the Vertebrate Gut. Adv Biol (Weinh) 2022; 6:e2200015. [PMID: 35652159 DOI: 10.1002/adbi.202200015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/23/2022] [Indexed: 01/28/2023]
Abstract
In vitro models of the gut-microbiome axis are in high demand. Conventionally, intestinal monolayers grown on Transwell setups are used to test the effects of commensals/pathogens on the barrier integrity, both under homeostatic and pathophysiological conditions. While such models remain valuable for deepening the understanding of host-microbe interactions, often, they lack key biological components that mediate this intricate crosstalk. Here, a 3D in vitro model of the vertebrate intestinal epithelium, interfaced with immune cells surviving in culture for over 3 weeks, is developed and applied to proof-of-concept studies of host-microbe interactions. More specifically, the establishment of stable host-microbe cocultures is described and functional and morphological changes in the intestinal barrier induced by the presence of commensal bacteria are shown. Finally, evidence is provided that the 3D vertebrate gut models can be used as platforms to test host-microbe-parasite interactions. Exposure of gut-immune-bacteria cocultures to helminth "excretory/secretory products" induces in vivo-like up-/down-regulation of certain cytokines. These findings support the robustness of the modular in vitro cell systems for investigating the dynamics of host-microbe crosstalk and pave the way toward new approaches for systems biology studies of pathogens that cannot be maintained in vitro, including parasitic helminths.
Collapse
Affiliation(s)
- Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Aimee M Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Alasdair J Nisbet
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Daniel R G Price
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| |
Collapse
|
25
|
Johnson B, Panek P, Yu A, Fischer E, Koba M, Mendoza Hermosillo D, Capaldo CT. Interferon gamma upregulates the cytokine receptors IFNGR1 and TNFRSF1A in HT-29-MTX E12 cells. Cytokine 2022; 156:155892. [PMID: 35653895 PMCID: PMC9636846 DOI: 10.1016/j.cyto.2022.155892] [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: 12/09/2021] [Revised: 03/24/2022] [Accepted: 04/15/2022] [Indexed: 11/22/2022]
Abstract
The intestinal mucosa protects the body from physical damage, pathogens, and antigens. However, inflammatory bowel diseases (IBDs) patients suffer from poor mucosal tissue function, including the lack of an effective cellular and/or mucus barrier. We investigated the mucus producing human colonic epithelial cell line HT29-MTX E12 to study its suitability as an in vitro model of cell/mucus barrier adaption during IBD. It was found that the proinflammatory cytokine interferon-gamma (IFN-γ), but not tumor necrosis factor-alpha (TNF-α), reduced cell viability. IFN-γ and TNF-α were found to synergize to decrease barrier function, as measured by trans-epithelial electric resistance (TER) and molecular flux assays. Cells cultured under an air-liquid interface produced an adherent mucus layer, and under these conditions reduced barrier function was found after cytokine exposure. Furthermore, IFN-γ, but not TNF-α treatment, upregulated the IFN-γ receptor 1 (IFNGR1) and TNF-α receptor super family 1A (TNFRSF1A) subunit mRNA in vitro. Co-stimulation resulted in increased mRNA expression of CLDN 2 and 5, two gene known to play a role in epithelial barrier integrity. Analysis of IBD patient samples revealed IFNGR1 and TNFRSF mRNA increased coincidently with guanylate binding protein 1 (GBP1) expression, an indicator of NFkB activity. Lastly, CLDN2 was found at higher levels in IBD patients while HNF4a was suppressed with disease. In conclusion, IFN-γ and TNF-α degrade epithelial/mucus barriers coincident with changes in CLDN gene and cytokine receptor subunit mRNA expression in HT29-MTX E12 cells. These changes largely reflect those observed in IBD patient samples.
Collapse
Affiliation(s)
- Brandon Johnson
- Center for Biomedical Research at The Queen's Medical Center, Honolulu, HI 96813, USA
| | - Paulina Panek
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | - Andy Yu
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | - Elizabeth Fischer
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | - Marli Koba
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | | | | |
Collapse
|
26
|
Milani N, Parrott N, Ortiz Franyuti D, Godoy P, Galetin A, Gertz M, Fowler S. Application of a gut-liver-on-a-chip device and mechanistic modelling to the quantitative in vitro pharmacokinetic study of mycophenolate mofetil. LAB ON A CHIP 2022; 22:2853-2868. [PMID: 35833849 DOI: 10.1039/d2lc00276k] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microphysiological systems (MPS) consisting of multiple linked organ-on-a-chip (OoC) components are highly promising tools with potential to provide more relevant in vitro to in vivo translation of drug disposition, efficacy and toxicity. A gut-liver OoC system was employed with Caco2 cells in co-culture with HT29 cells in the intestinal compartment and single donor primary hepatocytes in the hepatic compartment for the investigation of intestinal permeability, metabolism (intestinal and hepatic) and potential interplay of those processes. The prodrug mycophenolate mofetil was tested for quantitative evaluation of the gut-liver OoC due to the contribution of both gut and liver in its metabolism. Conversion of mycophenolate mofetil to active drug mycophenolic acid and further metabolism to a glucuronide metabolite was assessed over time in the gut apical, gut basolateral and liver compartments. Mechanistic modelling of experimental data was performed to estimate clearance and permeability parameters for the prodrug, active drug and glucuronide metabolite. Integration of gut-liver OoC data with in silico modelling allowed investigation of the complex combination of intestinal and hepatic processes, which is not possible with standard single tissue in vitro systems. A comprehensive evaluation of the mechanistic model, including structural model and parameter identifiability and global sensitivity analysis, enabled a robust experimental design and estimation of in vitro pharmacokinetic parameters. We propose that similar methodologies may be applied to other multi-organ microphysiological systems used for drug metabolism studies or wherever quantitative knowledge of changing drug concentration with time enables better understanding of biological effect.
Collapse
Affiliation(s)
- Nicoló Milani
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, UK
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Daniela Ortiz Franyuti
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Patricio Godoy
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, UK
| | - Michael Gertz
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Stephen Fowler
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
27
|
Hu W, Feng P, Zhang M, Tian T, Wang S, Zhao B, Li Y, Wang S, Wu C. Endotoxins Induced ECM-Receptor Interaction Pathway Signal Effect on the Function of MUC2 in Caco2/HT29 Co-Culture Cells. Front Immunol 2022; 13:916933. [PMID: 35757703 PMCID: PMC9226665 DOI: 10.3389/fimmu.2022.916933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Endotoxins are toxic substances that widely exist in the environment and can enter the intestine with food and other substances. Intestinal epithelial cells are protected by a mucus layer that contains MUC2 as its main structural component. However, a detailed understanding of the mechanisms involved in the function of the mucus barrier in endotoxin penetration is lacking. Here, we established the most suitable proportion of Caco-2/HT-29 co-culture cells as a powerful tool to evaluate the intestinal mucus layer. Our findings significantly advance current knowledge as focal adhesion and ECM-receptor interaction were identified as the two most significantly implicated pathways in MUC2 small interfering RNA (siRNA)-transfected Caco-2/HT-29 co-culture cells after 24 h of LPS stimulation. When the mucus layer was not intact, LPS was found to damage the tight junctions of Caco-2/HT29 co-cultured cells. Furthermore, LPS was demonstrated to inhibit the integrin-mediated focal adhesion structure and damage the matrix network structure of the extracellular and actin microfilament skeletons. Ultimately, LPS inhibited the interactive communication between the extracellular matrix and the cytoskeleton for 24 h in the siMUC2 group compared with the LPS(+) and LPS(-) groups. Overall, we recognized the potential of MUC2 as a tool for barrier function in several intestinal bacterial diseases.
Collapse
Affiliation(s)
- Wenxiang Hu
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Ping Feng
- College of Life Sciences, Yulin University, Yulin, China
| | - Mingming Zhang
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Tian Tian
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Shengxiang Wang
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Baoyu Zhao
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Yajie Li
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Shuo Wang
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| | - Chenchen Wu
- College of Animal Veterinary Medicine, Northwest A & F University, Yanling City, China
| |
Collapse
|
28
|
Tunca Koyun M, Sirin S, Aslim B, Taner G, Nigdelioglu Dolanbay S. Characterization of prodigiosin pigment by Serratia marcescens and the evaluation of its bioactivities. Toxicol In Vitro 2022; 82:105368. [PMID: 35476923 DOI: 10.1016/j.tiv.2022.105368] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 11/19/2022]
Abstract
The aim of the present study is to discover a bacterial pigment providing protection and prevention of neurological damage and cancer development, which can have a role as a non-synthetic food additive in the food industry as well as an active drug ingredient of anticancer drugs and pharmaceuticals for neural injury. Within this scope, Serratia marcescens MB703 strain was used to produce prodigiosin. Characterization of the prodigiosin was carried out using UV-VIS, and FT-IR. In addition, its inhibitory action on AChE and antioxidant activities were determined. The cytotoxic, genotoxic and antigenotoxic activities of the prodigiosin as well as its antiproliferative activities were detected. It was determined that the maximum production of the prodigiosin (72 mg/L). The prodigiosin was found to cause no significant difference in its inhibitory effect on AChE. The prodigiosin was found effective on all antioxidant parameters tested. The IC50 values of the prodigiosin on SK-MEL-30 and HT-29 cells were calculated as 70 and 47 μM, respectively. This IC50 values of the prodigiosin showed no cytotoxic effect on L929 cells. Prodigiosin did not have genotoxic effect alone and also seem to decrease DNA damage induced by H2O2 in L929 cells. The findings of in vitro experimental studies suggest that using the prodigiosin pigment as a drug candidate for cancer and neurodegenerative disease therapy is both effective and safe.
Collapse
Affiliation(s)
- Merve Tunca Koyun
- Department of Biology, Faculty of Science, Gazi University, 06500 Teknikokullar, Ankara, Turkey; Department of Bioengineering, Faculty of Engineering and Natural Sciences, Bursa Technical University, Bursa, Turkey.
| | - Seda Sirin
- Department of Biology, Faculty of Science, Gazi University, 06500 Teknikokullar, Ankara, Turkey
| | - Belma Aslim
- Department of Biology, Faculty of Science, Gazi University, 06500 Teknikokullar, Ankara, Turkey
| | - Gokce Taner
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Bursa Technical University, Bursa, Turkey
| | | |
Collapse
|
29
|
Impact of Caveolin-Mediated Endocytosis on the Trafficking of HIV within the Colonic Barrier. J Virol 2022; 96:e0020222. [PMID: 35297667 PMCID: PMC9006927 DOI: 10.1128/jvi.00202-22] [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] [Indexed: 11/20/2022] Open
Abstract
In the United States, most new cases of human immunodeficiency virus (HIV) belong to the at-risk group of gay and bisexual men. Developing therapies to reverse viral latency and prevent spread is paramount for the HIV cure agenda. In gay and bisexual men, a major, yet poorly characterized, route of HIV entry is via transport across the colonic epithelial barrier. While colonic tears and paracellular transport contribute to infection, we hypothesize that HIV entry through the colonic mucosa proceeds via a process known as transcytosis, involving (i) virion binding to the apical surface of the colonic epithelium, (ii) viral endocytosis, (iii) transport of virions across the cell, and (iv) HIV release from the basolateral membrane. Using Caco-2 colonic epithelial cells plated as a polarized monolayer in transwells, we characterized the mechanism of HIV transport. After exposing the monolayer to HIV apically, reverse transcription quantitative PCR (RT-qPCR) of the viral genome present in the basolateral chamber revealed that transport is dose dependent, cooperative, and inefficient, with released virus first detectable at 12 h. Inefficiency may be associated with >50% decline in detectable intracellular virus that correlates temporally with increased association of the virion with lysosomal-associated membrane protein 1 (LAMP-1+) endosomes. Microscopy revealed green fluorescent protein (GFP)-labeled HIV within the confines of the epithelial monolayer, with no virus detectable between cells, suggesting that viral transport is transcellular. Treatment of the monolayer with endocytosis inhibitors, cholesterol reducing agents, and small interfering RNA (siRNA) to caveolin showed that viral endocytosis is mediated by caveolin-coated endosomes contained in lipid rafts. These results indicate that HIV transport across the intestinal epithelial barrier via transcytosis is a viable mechanism for viral spread and a potential therapeutic target. IMPORTANCE Despite the success of combination antiretroviral therapy in suppressing HIV replication and the emergence and effectiveness of PrEP-based prevention strategies, in 2018, 37,968 people in the United States received a new HIV diagnosis, accompanied by 15,820 deaths. While the annual number of new diagnoses decreased 7% from 2014 to 2018, 14% of people with HIV did not know they were infected. Gay and bisexual men accounted for 69% of all HIV diagnoses and 83% of diagnoses among males. Due to the scope of the HIV epidemic, determining and understanding precise routes of infection and the mechanisms of viral spread are paramount to ending the epidemic. Since transcellular transport of HIV across an intact colonic epithelial barrier is poorly understood, our overall goal is to characterize the molecular events involved in HIV transcytosis across the intestinal epithelial cell.
Collapse
|
30
|
Cai Y, Liu L, Xia M, Tian C, Wu W, Dong B, Chu X. SEDDS facilitate cinnamaldehyde crossing the mucus barrier: The perspective of mucus and Caco-2/HT29 co-culture models. Int J Pharm 2022; 614:121461. [PMID: 35026310 DOI: 10.1016/j.ijpharm.2022.121461] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/19/2021] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
Self-emulsifying drug delivery systems (SEDDS) have potential applications in the delivery of hydrophobic components. Oral drugs are readily captured and cleared by intestinal mucus, a natural barrier that covers the mucosal epithelium and prevents the entry of foreign substances. In this study, we investigated for the first time the ability of SEDDS to deliver the lipophilic aldehyde cinnamaldehyde (CA-SEDDS) in rat mucus, mucin solution, Caco-2 and Caco-2/HT29 co-culture monolayer systems. CA-SEDDS was characterized by particle size, Zeta potential and the logDSEDDS/release medium. The capacity of CA-SEDDS to enhance mucus permeability was investigated in rat intestinal mucus gel and mucin solution with the period of in 12 h by Transwell® diffusion. We evaluated the potential of CA-SEDDS delivery of CA in a co-culture system of absorptive Caco-2 and mucus-secreting HT29 cells. CA-SEDDS exhibited excellent mucus permeability in mucus and mucin solutions, 5.1- and 2.8-fold higher than the free CA group, respectively. CA-SEDDS penetration increased by 2.5-fold compared with free CA when using the mucus-secreting co-culture cell model as a barrier. The relative oral bioavailability of CA-SEDDS was 242% compared to CA without formulation. These findings suggest that SEDDS exhibited good release and superior mucus permeability, displaying great potential for the future of hydrophobic oral applications.
Collapse
Affiliation(s)
- Ye Cai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China
| | - Liu Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China
| | - Mengqiu Xia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China
| | - Chunling Tian
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China
| | - Wenqing Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China
| | - Baoqi Dong
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, China; Engineering Technology Research Center of Modern Pharmaceutical Preparation, Anhui Province, China.
| |
Collapse
|
31
|
Delon L, Gibson R, Prestidge C, Thierry B. Mechanisms of uptake and transport of particulate formulations in the small intestine. J Control Release 2022; 343:584-599. [PMID: 35149142 DOI: 10.1016/j.jconrel.2022.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 10/19/2022]
Abstract
Micro- and nano-scale particulate formulations are widely investigated towards improving the oral bioavailability of both biologics and drugs with low solubility and/or low intestinal permeability. Particulate formulations harnessing physiological intestinal transport pathways have recently yielded remarkably high oral bioavailabilities, illustrating the need for better understanding the specific pathways underpinning particle small intestinal absorption and the relative role of intestinal cells. Mechanistic knowledge has been hampered by the well acknowledged limitations of current in vitro, in vivo and ex vivo models relevant to the human intestinal physiology and the lack of standardization in studies reporting absorption data. Here we review the relevant literature and critically discusses absorption pathways with a focus on the role of specific intestinal epithelial and immune cells. We conclude that while Microfold (M) cells are a valid target for oral vaccines, enterocytes play a greater role in the systemic bioavailability of orally administrated particulate formulations, particularly within the sub-micron size range. We also comment on less-reported mechanisms such as paracellular permeability of particles, persorption due to cell damage and uptake by migratory immune cells.
Collapse
Affiliation(s)
- Ludivine Delon
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia; Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Rachel Gibson
- Australia School of Allied Health Science and Practice, University of Adelaide, South Australia 5005, Australia
| | - Clive Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| |
Collapse
|
32
|
Samprasit W, Opanasopit P, Chamsai B. Alpha-mangostin and resveratrol, dual-drugs-loaded mucoadhesive thiolated chitosan-based nanoparticles for synergistic activity against colon cancer cells. J Biomed Mater Res B Appl Biomater 2021; 110:1221-1233. [PMID: 34919783 DOI: 10.1002/jbm.b.34992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 10/25/2021] [Accepted: 12/05/2021] [Indexed: 02/04/2023]
Abstract
Alpha-mangostin (M) and resveratrol (R), dual-drugs-loaded mucoadhesive thiolated chitosan-based nanoparticles (NPs) coated by Eudragit® S100 (S) were developed for colon-specific delivery and synergistic activity against colon cancer cells. The NPs were prepared by the ionotropic gelation method and coated with S. The particle size and zeta potential of NPs before and after the coating process were observed. The M and R loading efficiency, mucoadhesive properties, as well as release patterns were examined. Moreover, the activity against colon cancer cells of M, R, and NPs were studied for their synergistic activity. M and R-loaded NPs (MR-TNPs) were spherical in shape with sizes of around 540 nm and zeta potential of +39 mV. The S coating of MR-TNPs provided larger particle sizes which offered lower zeta potential. However, it created an increase in M and R loading, prevented M and R release at the upper gastrointestinal tract, and enhanced M and R reaching the colon. S dissolved at pH > 7.0 while thiolated chitosan formed the mucoadhesion, resulting in M and R remaining in the colon and allowing them to enter the colon cancer cells. The half-maximal inhibitory concentration values of NPs was dramatically decreased when M and R were dually loaded into the NPs, which indicated significantly higher activity against colon cancer cells. Moreover, M and R loading at this ratio applied synergistic efficiency. The results illustrated that NPs successfully loaded drugs and achieved synergistic efficiency. This system could be promising in facilitating targeted nanomedicines for the treatment of colon cancer.
Collapse
Affiliation(s)
- Wipada Samprasit
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Praneet Opanasopit
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Benchawan Chamsai
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| |
Collapse
|
33
|
Rahman S, Ghiboub M, Donkers JM, van de Steeg E, van Tol EAF, Hakvoort TBM, de Jonge WJ. The Progress of Intestinal Epithelial Models from Cell Lines to Gut-On-Chip. Int J Mol Sci 2021; 22:ijms222413472. [PMID: 34948271 PMCID: PMC8709104 DOI: 10.3390/ijms222413472] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past years, several preclinical in vitro and ex vivo models have been developed that helped to understand some of the critical aspects of intestinal functions in health and disease such as inflammatory bowel disease (IBD). However, the translation to the human in vivo situation remains problematic. The main reason for this is that these approaches fail to fully reflect the multifactorial and complex in vivo environment (e.g., including microbiota, nutrition, and immune response) in the gut system. Although conventional models such as cell lines, Ussing chamber, and the everted sac are still used, increasingly more sophisticated intestinal models have been developed over the past years including organoids, InTESTine™ and microfluidic gut-on-chip. In this review, we gathered the most recent insights on the setup, advantages, limitations, and future perspectives of most frequently used in vitro and ex vivo models to study intestinal physiology and functions in health and disease.
Collapse
Affiliation(s)
- Shafaque Rahman
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
| | - Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
- Department of Pediatric Gastroenterology and Nutrition, Amsterdam University Medical Centers, Emma Children’s Hospital, 1105 AZ Amsterdam, The Netherlands
| | - Joanne M. Donkers
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (J.M.D.); (E.v.d.S.); (E.A.F.v.T.)
| | - Evita van de Steeg
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (J.M.D.); (E.v.d.S.); (E.A.F.v.T.)
| | - Eric A. F. van Tol
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (J.M.D.); (E.v.d.S.); (E.A.F.v.T.)
| | - Theodorus B. M. Hakvoort
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
- Department of Surgery, University of Bonn, 53113 Bonn, Germany
- Correspondence:
| |
Collapse
|
34
|
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: 2.7] [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
|
35
|
Measuring the oral bioavailability of protein hydrolysates derived from food sources: A critical review of current bioassays. Biomed Pharmacother 2021; 144:112275. [PMID: 34628165 DOI: 10.1016/j.biopha.2021.112275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Food proteins are a source of hydrolysates with potentially useful biological attributes. Bioactive peptides from food-derived proteins are released from hydrolysates using exogenous industrial processes or endogenous intestinal enzymes. Current in vitro permeability assays have limitations in predicting the oral bioavailability (BA) of bioactive peptides in humans. There are also difficulties in relating the low blood levels of food-derived bioactive peptides detected in preclinical in vivo models to pharmacodynamic read-outs relevant for humans. SCOPE AND APPROACH In this review, we describe in vitro assays of digestion, permeation, and metabolism as indirect predictors of the potential oral BA of hydrolysates and their constituent bioactive peptides. We discuss the relationship between industrial hydrolysis processes and the oral BA of hydrolysates and their peptide by-products. KEY FINDINGS Hydrolysates are challenging for analytical detection methods due to capacity for enzymatic generation of peptides with novel sequences and also new modifications of these peptides during digestion. Mass spectrometry and peptidomics can improve the capacity to detect individual peptides released from complex hydrolysates in biological milieu.
Collapse
|
36
|
Hoffmann P, Burmester M, Langeheine M, Brehm R, Empl MT, Seeger B, Breves G. Caco-2/HT29-MTX co-cultured cells as a model for studying physiological properties and toxin-induced effects on intestinal cells. PLoS One 2021; 16:e0257824. [PMID: 34618824 PMCID: PMC8496855 DOI: 10.1371/journal.pone.0257824] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/12/2021] [Indexed: 12/28/2022] Open
Abstract
Infectious gastrointestinal diseases are frequently caused by toxins secreted by pathogens which may impair physiological functions of the intestines, for instance by cholera toxin or by heat-labile enterotoxin. To obtain a functional model of the human intestinal epithelium for studying toxin-induced disease mechanisms, differentiated enterocyte-like Caco-2 cells were co-cultured with goblet cell-like HT29-MTX cells. These co-cultures formed a functional epithelial barrier, as characterized by a high electrical resistance and the presence of physiological intestinal properties such as glucose transport and chloride secretion which could be demonstrated electrophysiologically and by measuring protein expression. When the tissues were exposed to cholera toxin or heat-labile enterotoxin in the Ussing chamber, cholera toxin incubation resulted in an increase in short-circuit currents, indicating an increase in apical chloride secretion. This is in line with typical cholera toxin-induced secretory diarrhea in humans, while heat-labile enterotoxin only showed an increase in short-circuit-current in Caco-2 cells. This study characterizes for the first time the simultaneous measurement of physiological properties on a functional and structural level combined with the epithelial responses to bacterial toxins. In conclusion, using this model, physiological responses of the intestine to bacterial toxins can be investigated and characterized. Therefore, this model can serve as an alternative to the use of laboratory animals for characterizing pathophysiological mechanisms of enterotoxins at the intestinal level.
Collapse
Affiliation(s)
- Pascal Hoffmann
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Burmester
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Langeheine
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ralph Brehm
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Michael T. Empl
- Institute for Food Toxicology, University of Veterinary Medicine, Hannover, Germany
| | - Bettina Seeger
- Institute for Food Toxicology, University of Veterinary Medicine, Hannover, Germany
| | - Gerhard Breves
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
37
|
Pinho SC, Faria MA, Melo A, Pinto E, Almeida A, Alves R, Cabrita AR, Fonseca AJ, Ferreira IM. Effect of skimmed milk on intestinal tract: Prevention of increased reactive oxygen species and nitric oxide formation. Int Dairy J 2021. [DOI: 10.1016/j.idairyj.2021.105046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
38
|
Hypoxia and heat stress affect epithelial integrity in a Caco-2/HT-29 co-culture. Sci Rep 2021; 11:13186. [PMID: 34162953 PMCID: PMC8222227 DOI: 10.1038/s41598-021-92574-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia and hyperthermia, which can be induced by high environmental temperature or strenuous exercise, are two common stressors that affect intestinal epithelial integrity and lead to multiple clinical symptoms. In this study, we developed an in-vitro intestinal monolayer model using two human colonic epithelial cell lines, Caco-2 and HT-29, co-cultured in Transwell inserts, and investigated the effects of heat treatment and/or hypoxia on the epithelial barrier function. The monolayer with a ratio of 9:1 (Caco-2:HT-29) showed high trans-epithelial electrical resistance (TEER), low Lucifer Yellow permeability and high mucin production. Hyperthermia and/or hypoxia exposure (2 h) triggered heat shock and oxidative stress responses. HSP-70 and HSF-1 protein levels were up-regulated by hyperthermia, which were further enhanced when hyperthermia was combined with hypoxia. Increased HIF-1α protein expression and Nrf2 nuclear translocation was only caused by hypoxia. Hyperthermia and/or hypoxia exposure disrupted the established monolayer by increasing paracellular permeability, decreasing ZO-1, claudin-3 and occludin protein/mRNA expression, while enhancing E-cadherin protein expression. Tight junction protein distribution in the monolayer was also modulated by the hyperthermia and/or hypoxia exposure. In addition, transcription levels of mucin genes, MUC-2 and MUC-5AC, were increased after 2 h of hyperthermia and/or hypoxia exposure. In conclusion, this Caco-2/HT-29 cell model is valid and effective for studying detrimental effects of hyperthermia and/or hypoxia on intestinal barrier function and related heat shock and oxidative stress pathways and can be used to investigate possible interventions to reverse hyperthermia and/or hypoxia-induced intestinal epithelial injury.
Collapse
|
39
|
Corrochano AR, Cal R, Kennedy K, Wall A, Murphy N, Trajkovic S, O’Callaghan S, Adelfio A, Khaldi N. Characterising the efficacy and bioavailability of bioactive peptides identified for attenuating muscle atrophy within a Vicia faba-derived functional ingredient. Curr Res Food Sci 2021; 4:224-232. [PMID: 33937870 PMCID: PMC8079236 DOI: 10.1016/j.crfs.2021.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 12/20/2022] Open
Abstract
Characterising key components within functional ingredients as well as assessing efficacy and bioavailability is an important step in validating nutritional interventions. Machine learning can assess large and complex data sets, such as proteomic data from plants sources, and so offers a prime opportunity to predict key bioactive components within a larger matrix. Using machine learning, we identified two potentially bioactive peptides within a Vicia faba derived hydrolysate, NPN_1, an ingredient which was previously identified for preventing muscle loss in a murine disuse model. We investigated the predicted efficacy of these peptides in vitro and observed that HLPSYSPSPQ and TIKIPAGT were capable of increasing protein synthesis and reducing TNF-α secretion, respectively. Following confirmation of efficacy, we assessed bioavailability and stability of these predicted peptides and found that as part of NPN_1, both HLPSYSPSPQ and TIKIPAGT survived upper gut digestion, were transported across the intestinal barrier and exhibited notable stability in human plasma. This work is a first step in utilising machine learning to untangle the complex nature of functional ingredients to predict active components, followed by subsequent assessment of their efficacy, bioavailability and human plasma stability in an effort to assist in the characterisation of nutritional interventions.
Collapse
Affiliation(s)
| | - Roi Cal
- Nuritas Ltd., D02 RY95, Dublin, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Screening for Effects of Inhaled Nanoparticles in Cell Culture Models for Prolonged Exposure. NANOMATERIALS 2021; 11:nano11030606. [PMID: 33671010 PMCID: PMC7997552 DOI: 10.3390/nano11030606] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/24/2022]
Abstract
Respiratory exposure of humans to environmental and therapeutic nanoparticles repeatedly occurs at relatively low concentrations. To identify adverse effects of particle accumulation under realistic conditions, monocultures of Calu-3 and A549 cells and co-cultures of A549 and THP-1 macrophages in the air–liquid interphase culture were exposed repeatedly to 2 µg/cm2 20 nm and 200 nm polystyrene particles with different functionalization. Particle accumulation, transepithelial electrical resistance, dextran (3–70 kDa) uptake and proinflammatory cytokine secretion were determined over 28 days. Calu-3 cells showed constant particle uptake without any change in barrier function and cytokine release. A549 cells preferentially ingested amino- and not-functionalized particles combined with decreased endocytosis. Cytokine release was transiently increased upon exposure to all particles. Carboxyl-functionalized demonstrated higher uptake and higher cytokine release than the other particles in the A549/THP-1 co-cultures. The evaluated respiratory cells and co-cultures ingested different amounts and types of particles and caused small (partly transient) effects. The data suggest that the healthy cells can adapt to low doses of non-cytotoxic particles.
Collapse
|
41
|
Alfarafisa NM, Kitaguchi K, Yabe T. Diospyros kaki extract protects myoblasts from oxidative stress-induced cytotoxicity via secretions derived from intestinal epithelium. Biosci Biotechnol Biochem 2021; 85:430-439. [PMID: 33604620 DOI: 10.1093/bbb/zbaa048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022]
Abstract
Under oxidative stress, reactive oxygen species (ROS) alter signal transduction and induce macromolecular damage in cells. Such oxidative damage can lead to sarcopenia, an age-related syndrome characterized by a progressive loss of mass and strength of skeletal muscles. Because food components do not directly come in contact with muscle cells, we focused on the effects of secretions produced by stimulated intestinal epithelial cells on oxidative stress in myoblast cells. An extract of Diospyros kaki was fractionated using different concentrations of ethanol. Each fraction showed different levels of antioxidant and phenolic compounds. The biological activity was evaluated using a Caco-2 cell coculture system. Secretions from Caco-2 cells exposed to 0.5 mg/mL D. kaki extract attenuated the oxidative stress-induced reduction of C2C12 cell viability, suggesting that the D. kaki extract could stimulate intestinal epithelial cells to produce secretions that reduce oxidative stress in myoblasts in vitro.
Collapse
Affiliation(s)
| | - Kohji Kitaguchi
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Tomio Yabe
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan.,Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| |
Collapse
|
42
|
Pinto D, Silva AM, Freitas V, Vallverdú-Queralt A, Delerue-Matos C, Rodrigues F. Microwave-Assisted Extraction as a Green Technology Approach to Recover Polyphenols from Castanea sativa Shells. ACTA ACUST UNITED AC 2021. [DOI: 10.1021/acsfoodscitech.0c00055] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Diana Pinto
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal
| | - Ana M. Silva
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal
| | - Vitor Freitas
- REQUIMTE/LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre 1021/1055, 4169-007 Porto, Portugal
| | - Anna Vallverdú-Queralt
- Department of Nutrition, Food Science and Gastronomy, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- Consorcio CIBER, M. P. Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal
| | - Francisca Rodrigues
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal
| |
Collapse
|
43
|
Frühbeck G, Mentxaka A, Ahechu P, Gómez-Ambrosi J, Ramírez B, Becerril S, Rodríguez A, Unamuno X, Cienfuegos JA, Casado M, Burrell MA, Martín M, Baixauli J, Valentí V, Moncada R, Reina G, Silva C, Catalán V. The Differential Expression of the Inflammasomes in Adipose Tissue and Colon Influences the Development of Colon Cancer in a Context of Obesity by Regulating Intestinal Inflammation. J Inflamm Res 2021; 14:6431-6446. [PMID: 34880645 PMCID: PMC8646233 DOI: 10.2147/jir.s335882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Inflammasomes maintain tissue homeostasis and their altered regulation in the colon, and the adipose tissue (AT) leads to chronic activation of inflammatory pathways promoting colon cancer (CC) development. We aimed to analyze the potential involvement of inflammasomes in obesity-associated CC. METHODS Ninety-nine volunteers [61 with obesity (OB) and 38 normoponderal (NP)] further subclassified according to the approved protocol for the diagnosis of CC (58 without CC and 41 with CC) were included in the case-control study. RESULTS CC (P<0.01) and obesity (P<0.01) were accompanied by increased mRNA levels of NLRP3, NLRP6, ASC, IL1B and NOD2 in VAT. Contrarily, patients with CC exhibited a downregulation of NLRP6 and IL18 in their colon. Additionally, we revealed that the decreased Nlrp1 (P<0.05), Nlrp3 (P<0.01) and Nlrp6 (P<0.01) mRNA levels in the colon from obese rats significantly increase (P<0.05) after caloric restriction. Adipocyte-conditioned media obtained from subjects with obesity reduced (P<0.01) the mRNA of NLRP3 as well as molecules involved in maintaining the intestinal integrity (MUC2, CLDN1 and TJP1) and the anti-inflammatory factors FGF21, KLF4, and IL33 and in HT-29 cells. We also found that the knockdown of NLRP6 in HT-29 cells significantly upregulated (P<0.05) the mRNA of NLRP1 and NLRP3 and inhibited (P<0.05) the expression levels of MUC2. Finally, we showed that the incubation of HT-29 with Akkermansia muciniphila influence (P<0.05) the inflammasome expression profile as well as intestinal integrity-related genes and aberrant inflammation. CONCLUSIONS These findings provide evidence that the downregulated levels of NLRP6 and IL18 in the colon from patients with CC may be responsible for a reduced intestinal-barrier integrity, triggering local inflammation, which in turn acts on the dysfunctional AT in obesity, increasing the expression of different inflammasome components and flaring up a vicious cycle of uncontrollable inflammatory cascades that favours a pro-tumorigenic microenvironment.
Collapse
Affiliation(s)
- Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Mentxaka
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
| | - Patricia Ahechu
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Medical Engineering Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Marcos Casado
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - María A Burrell
- Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Marina Martín
- Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Jorge Baixauli
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victor Valentí
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Reina
- Microbiology Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
44
|
Contino M, Guglielmo S, Riganti C, Antonello G, Perrone MG, Giampietro R, Rolando B, Fruttero R, Colabufo NA. One molecule two goals: A selective P-glycoprotein modulator increases drug transport across gastro-intestinal barrier and recovers doxorubicin toxicity in multidrug resistant cancer cells. Eur J Med Chem 2020; 208:112843. [DOI: 10.1016/j.ejmech.2020.112843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022]
|
45
|
Bottani M, Cattaneo S, Pica V, Stuknytė M, Gomarasca M, Lombardi G, Banfi G, Noni ID, Ferraretto A. Gastrointestinal In Vitro Digests of Infant Biscuits Formulated with Bovine Milk Proteins Positively Affect In Vitro Differentiation of Human Osteoblast-Like Cells. Foods 2020; 9:foods9101510. [PMID: 33096628 PMCID: PMC7589107 DOI: 10.3390/foods9101510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 11/17/2022] Open
Abstract
Infant biscuits (IBs) are part of complementary feeding from weaning up to the age of five years. They normally contain bovine milk proteins, which can influence bone development. This potential effect was investigated using experimental baked IBs, which were prepared from doughs containing different type of dairy proteins: milk protein concentrate (IB1), whey protein isolate (IB2), and skimmed milk powder (IB3). Dairy protein-free (IB0) and gluten-free (IB4) biscuits were also formulated. The in vitro gastrointestinal digests of IBs (IBDs) were tested on a co-culture of Caco-2/HT-29 70/30 cells as an in vitro model of human small intestine. None of the IBDs influenced cell viability and monolayer integrity, while IBD0 and IBD4 increased Peptide-YY production. The basolateral contents of Transwell plates seeded with Caco-2/HT-29 70/30 co-culture, mimicking metabolized IBDs (MIBDs), were tested on Saos-2 cells, an in vitro model of human osteoblast-like cells. After incubation, MIBD0, lacking dairy proteins, decreased the cell viability, while MIBD2, containing whey protein isolate, increased both the viability and the number of cells. MIBD2 and MIBD4, the latter containing both casein and whey proteins, increased alkaline phosphatase activity, a bone differentiation marker. These results highlight that IBs containing dairy proteins positively affect bone development.
Collapse
Affiliation(s)
- Michela Bottani
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Via Galeazzi 4, 20161 Milan, Italy; (M.B.); (M.G.); (G.L.); (G.B.)
| | - Stefano Cattaneo
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milan, Italy; (S.C.); (V.P.)
| | - Valentina Pica
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milan, Italy; (S.C.); (V.P.)
| | - Milda Stuknytė
- Unitech COSPECT—University Technological Platforms Office, University of Milan, Via Golgi 19, 20133 Milan, Italy;
| | - Marta Gomarasca
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Via Galeazzi 4, 20161 Milan, Italy; (M.B.); (M.G.); (G.L.); (G.B.)
| | - Giovanni Lombardi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Via Galeazzi 4, 20161 Milan, Italy; (M.B.); (M.G.); (G.L.); (G.B.)
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Królowej Jadwigi 27/39, 61-871 Poznań, Poland
| | - Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Via Galeazzi 4, 20161 Milan, Italy; (M.B.); (M.G.); (G.L.); (G.B.)
- Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Ivano De Noni
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milan, Italy; (S.C.); (V.P.)
- Correspondence: ; Tel.: +39-02-503-16680
| | - Anita Ferraretto
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy;
| |
Collapse
|
46
|
Aguilar-Rojas A, Olivo-Marin JC, Guillen N. Human intestinal models to study interactions between intestine and microbes. Open Biol 2020; 10:200199. [PMID: 33081633 PMCID: PMC7653360 DOI: 10.1098/rsob.200199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Implementations of suitable in vitro cell culture systems of the human intestine have been essential tools in the study of the interaction among organs, commensal microbiota, pathogens and parasites. Due to the great complexity exhibited by the intestinal tissue, researchers have been developing in vitro/ex vivo systems to diminish the gap between conventional cell culture models and the human intestine. These models are able to reproduce different structures and functional aspects of the tissue. In the present review, information is recapitulated on the most used models, such as cell culture, intestinal organoids, scaffold-based three-dimensional models, and organ-on-a-chip and their use in studying the interaction between human intestine and microbes, and their advantages and limitations are also discussed.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Medicina Reproductiva, Unidad Médica de Alta Especialidad en Ginecología y Obstetricia No. 4 ‘Dr. Luis Castelazo Ayala’, Av. Río Magdalena No. 289, Col. Tizapán San Ángel, C.P. 01090 Ciudad de México, México
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, UMR3691, 25 Rue du Dr Roux, 75015 Paris, France
| | - Nancy Guillen
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, ERL9195, 25 Rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
47
|
Reale O, Huguet A, Fessard V. Co-culture model of Caco-2/HT29-MTX cells: A promising tool for investigation of phycotoxins toxicity on the intestinal barrier. CHEMOSPHERE 2020; 273:128497. [PMID: 34756374 DOI: 10.1016/j.chemosphere.2020.128497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 06/13/2023]
Abstract
Most lipophilic phycotoxins have been involved in human intoxications but some of these toxins have never been proven to induce human gastro-intestinal symptoms, although intestinal damage in rodents has been documented. For investigating the in vitro toxicological profile of lipophilic phycotoxins on intestine, the epithelial Caco-2 cell line has been the most commonly used model. Nevertheless, considering the complexity of the intestinal epithelium, in vitro co-cultures integrating enterocyte-like and mucus-secreting cell types are expected to provide more relevant data. In this study, the toxic effects (viability, inflammation, cellular monolayer integrity, modulation of cell type proportion and production of mucus) of four lipophilic phycotoxins (PTX2, YTX, AZA1 and OA) were evaluated in Caco-2/HT29-MTX co-cultured cells. The four toxins induced a reduction of viability from 20% to 50% and affected the monolayer integrity. Our results showed that the HT29-MTX cells population were more sensitive to OA and PTX2 than Caco-2 cells. Among the four phycotoxins, OA induced inflammation (28-fold increase of IL-8 release) and also a slight increase of both mucus production (up-regulation of mucins mRNA expression) and mucus secretion (mucus area and density). For PTX2 we observed an increase of IL-8 release but weaker than OA. Intestinal cell models integrating several cell types can contribute to improve hazard characterization and to describe more accurately the modes of action of phycotoxins.
Collapse
Affiliation(s)
- Océane Reale
- ANSES, Fougères Laboratory, Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères, 35306, France.
| | - Antoine Huguet
- ANSES, Fougères Laboratory, Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères, 35306, France.
| | - Valérie Fessard
- ANSES, Fougères Laboratory, Toxicology of Contaminants Unit, French Agency for Food, Environmental and Occupational Health & Safety, Fougères, 35306, France.
| |
Collapse
|
48
|
Allaw M, Manca ML, Caddeo C, Recio MC, Pérez-Brocal V, Moya A, Fernàndez-Busquets X, Manconi M. Advanced strategy to exploit wine-making waste by manufacturing antioxidant and prebiotic fibre-enriched vesicles for intestinal health. Colloids Surf B Biointerfaces 2020; 193:111146. [PMID: 32485579 DOI: 10.1016/j.colsurfb.2020.111146] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Grape extract-loaded fibre-enriched vesicles, nutriosomes, were prepared by combining antioxidant extracts obtained from grape pomaces and a prebiotic, soluble fibre (Nutriose®FM06). The nutriosomes were small in size (from ∼140 to 260 nm), homogeneous (polydispersity index < 0.2) and highly negative (∼ -79 mV). The vesicles were highly stable during 12 months of storage at 25 °C. When diluted with warmed (37 °C) acidic medium (pH 1.2) of high ionic strength, the vesicles only displayed an increase of the mean diameter and a low release of the extract, which were dependent on Nutriose concentration. The formulations were highly biocompatible and able to protect intestinal cells (Caco-2) from oxidative stress damage. In vivo results underlined that the composition of mouse microbiota was not affected by the vesicular formulations. Overall results support the potential application of grape nutriosomes as an alternative strategy for the protection of the intestinal tract.
Collapse
Affiliation(s)
- Mohamad Allaw
- Dept. of Scienze della Vita e dell'Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Maria Letizia Manca
- Dept. of Scienze della Vita e dell'Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy.
| | - Carla Caddeo
- Dept. of Scienze della Vita e dell'Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Maria Carmen Recio
- Dept. of Pharmacology, Faculty of Pharmacy, University of Valencia, Avda Vicent Andrés Estellés sn, 46100 Burjassot-Valencia, Spain
| | - Vicente Pérez-Brocal
- Dept. of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain; CIBER in Epidemiology and Public Health (CIBEResp), Madrid, Spain
| | - Andres Moya
- Dept. of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain; CIBER in Epidemiology and Public Health (CIBEResp), Madrid, Spain; Institute for Integrative Systems Biology (I2SysBio), The University of Valencia and The Spanish National Research Council (CSIC)-UVEG), Valencia, Spain
| | - Xavier Fernàndez-Busquets
- Institute for Bioengineering of Catalonia, (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Maria Manconi
- Dept. of Scienze della Vita e dell'Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| |
Collapse
|
49
|
Elkateb H, Tatham LM, Cauldbeck H, Niezabitowska E, Owen A, Rannard S, McDonald T. Optimization of the synthetic parameters of lipid polymer hybrid nanoparticles dual loaded with darunavir and ritonavir for the treatment of HIV. Int J Pharm 2020; 588:119794. [PMID: 32828978 DOI: 10.1016/j.ijpharm.2020.119794] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022]
Abstract
Human Immunodeficiency Virus (HIV) is a global health concern to which nanomedicine approaches provide opportunities to improve the bioavailability of existing drugs used to treat HIV.In this article, lipid polymer hybrid nanoparticles (LPHNs) were developed as a system to provide a combination drug delivery of two leading antiretroviral drugs; darunavir (DRV) and its pharmacokinetic enhancer ritonavir (RTV).The LPHNs were designed with a poly(D, l-lactide-co-glycolide) (PLGA) core, and soybean lecithin (SBL) and Brij 78 as the stabilizers. The LPHNs were prepared by modified nanoprecipitation and the effect of synthetic conditions on the particle properties was studied, which included the Z-average diameter and polydispersity index of LPHNs in water and phosphate buffered saline, and the morphology of the particles. This investigation aimed to prepare a formulation that could be stored in its dry and redispersible form, therefore avoiding the challenges associated with storage of dispersions. The optimum ratio of stabilizer to polymer core was established at 20% w/w, and Brij 78 was found to be crucial in providing colloidal stability in physiological solutions; the minimum amount of Brij 78 required to provide stability in phosphate buffered saline was 70% w/w of the total stabilizer mass. Viable formulations of LPHNs containing DRV and RTV in the clinically used 8:1 ratio were prepared containing 20% w/w DRV with respect to the PLGA mass. The use of cryoprotectant, polyethylene glycol, combined with freeze-drying yielded LPHNs with a Z-average diameter of 150 nm when the particles were re-dispersed in water. The oral absorption behavior was assessed using an in vitro triple culture model. Whilst the use of cryoprotectant and freeze-drying led to no improvement of the transcellular permeability compared to the unformulated drugs, the non-freeze-dried samples with the highest soybean lecithin led to increased transcellular permeability, revealing the potential of LPHNs for enhancing HIV treatment.
Collapse
Affiliation(s)
- Heba Elkateb
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, El Gomhouria Street, 35516, Egypt
| | - Lee M Tatham
- Department of Molecular and Clinical Pharmacology, Materials Innovation Factory, University of Liverpool, Liverpool L7 3NY, UK; Tandem Nano Ltd., Liverpool, UK
| | - Helen Cauldbeck
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, UK
| | - Edyta Niezabitowska
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, UK
| | - Andrew Owen
- Department of Molecular and Clinical Pharmacology, Materials Innovation Factory, University of Liverpool, Liverpool L7 3NY, UK; Tandem Nano Ltd., Liverpool, UK
| | - Steve Rannard
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, UK; Tandem Nano Ltd., Liverpool, UK
| | - Tom McDonald
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, UK.
| |
Collapse
|
50
|
Schoultz I, Keita ÅV. The Intestinal Barrier and Current Techniques for the Assessment of Gut Permeability. Cells 2020; 9:E1909. [PMID: 32824536 PMCID: PMC7463717 DOI: 10.3390/cells9081909] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/08/2023] Open
Abstract
The intestinal barrier is essential in human health and constitutes the interface between the outside and the internal milieu of the body. A functional intestinal barrier allows absorption of nutrients and fluids but simultaneously prevents harmful substances like toxins and bacteria from crossing the intestinal epithelium and reaching the body. An altered intestinal permeability, a sign of a perturbed barrier function, has during the last decade been associated with several chronic conditions, including diseases originating in the gastrointestinal tract but also diseases such as Alzheimer and Parkinson disease. This has led to an intensified interest from researchers with diverse backgrounds to perform functional studies of the intestinal barrier in different conditions. Intestinal permeability is defined as the passage of a solute through a simple membrane and can be measured by recording the passage of permeability markers over the epithelium via the paracellular or the transcellular route. The methodological tools to investigate the gut barrier function are rapidly expanding and new methodological approaches are being developed. Here we outline and discuss, in vivo, in vitro and ex vivo techniques and how these methods can be utilized for thorough investigation of the intestinal barrier.
Collapse
Affiliation(s)
- Ida Schoultz
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, 703 62 Örebro, Sweden;
| | - Åsa V. Keita
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|