1
|
Kwon SY, Jo SH, Park J, Park JH, Kim YR, Baek JH, Kim MG, Choi BG, Hong NY, Jung HK, Ryu HW, Jeon JS, Kim YG. Development of the Gut Microbial Immune and Epithelial Cellular System (GutMICS) to Investigate the Immunological Role of Gut Anaerobes. Biotechnol Bioeng 2025. [PMID: 40411261 DOI: 10.1002/bit.29031] [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: 02/02/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/26/2025]
Abstract
The gut microbiota plays an essential role in host health by regulating gut barrier function and immune system homeostasis. However, research into the physiological and immunological functions of the gut microbiota using In Vitro models that mimic the immune environment of the gut remains limited. Herein, we developed the Gut Microbial Immune & Epithelial Cellular System (GutMICS), a device for coculturing anaerobic gut microbes with host cells, including intestinal epithelial and immune cells. Coculturing Akkermansia muciniphila with GutMICS sustained host cell viability and microbial activity for 72 h. In a lipopolysaccharide- and tumor necrosis factor-α (TNF-α)-induced inflammation model, A. muciniphila enhanced the intestinal barrier function, prevented barrier disruption, reduced pro-inflammatory cytokines (interleukin (IL)-6, TNF-α), and increased anti-inflammatory cytokines (IL-10). Additionally, A. muciniphila protected against Salmonella Typhimurium infection by reducing adhesion and invasion, thereby preventing pathogen-induced cell death. This study used GutMICS to characterize the anti-inflammatory properties of A. muciniphila and its ability to inhibit pathogen infection, demonstrating that GutMICS is a valuable tool for assessing the effects of anaerobic gut microbes on host cells. The ability of the system to simulate various inflammatory environments is expected to have broad applications in the study of host-microbe interactions.
Collapse
Affiliation(s)
- Seo-Young Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Joonha Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ji-Hyeon Park
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ye-Rim Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ji-Hyun Baek
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Min-Gyu Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Bo-Gyeong Choi
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | | | | | - Hee-Wook Ryu
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Kamlárová A, Kvaková M, Ambro Ľ, Link R, Bertková I, Hertelyová Z, Janíčko M, Hijová E, Štofilová J. Improvement of the inflammation-damaged intestinal barrier and modulation of the gut microbiota in ulcerative colitis after FMT in the SHIME® model. BMC Complement Med Ther 2025; 25:145. [PMID: 40259351 PMCID: PMC12013018 DOI: 10.1186/s12906-025-04889-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/09/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) seems to be a promising approach in ulcerative colitis (UC) management with the aim of repopulating a patient's dysbiotic microbiota with beneficial bacteria and restore its metabolic activity to its healthy characteristics. Metabolites present after FMT may improve the function and integrity of the intestinal barrier, reduce inflammation, and thus induce remission in an UC patient. In this study we evaluated whether the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) model may be a suitable non-invasive alternative for studying and modifying the dysbiotic microbiota in UC by FMT application. METHODS SHIME® model was used to investigate microbial and metabolic changes in the gut microbiota of UC patient induced by FMT application. FMT-modified metabolites from SHIME® were applied to an in vitro model of the intestinal barrier (differentiated Caco-2 and HT-29-MTX-E12 cell lines) compromised by pro-inflammatory cytokines to study the effect of FMT on the intestinal barrier. RESULTS Qualitative and quantitative microbial analyses showed that FMT increased the diversity and variability of the microbiota in UC patient associated with a significant increase in total bacteria, Bacteroidota and Lactobacillus, as well as an increase in butyrate levels. In addition, an increase in the relative abundance of some important species such as Faecalibacterium prausnitzii and Bifidobacterium longum was observed, and there was also an enrichment of the microbiota with new species such as Blautia obeum, Roseburia faecis, Bifidobacterium adolescentis, Fusicatenibacter saccharivorans and Eubacterium rectale. Furthermore, microbial metabolites modulated by FMT from the SHIME® model prevented intestinal barrier damage and inhibited interleukin 8 (IL-8) and monocyte chemoattractant protein 1 (MCP-1) secretion when cell barriers were pretreated with FMT medium for 24 h. In summary, this study confirmed that a single dose of FMT beneficially modulated the composition and metabolic activity of the UC microbiota in the SHIME® model. CONCLUSIONS FMT favorably modulates the gut microbiota of UC patient cultured in the SHIME® model. FMT-modulated SHIME-derived microbial metabolites improve intact and inflamed intestinal barrier properties in vitro. Repeated applications are necessary to maintain the beneficial effect of FMT in SHIME® model.
Collapse
Affiliation(s)
- Anna Kamlárová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Monika Kvaková
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Ľuboš Ambro
- Center for Interdisciplinary Biosciences, Technology and Innovation Park, P.J. Šafárik University, Jesenna 5, Košice, 040 01, Slovakia
| | - René Link
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Izabela Bertková
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Zdenka Hertelyová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Martin Janíčko
- 2nd Department of Internal Medicine, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Emília Hijová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Jana Štofilová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia.
| |
Collapse
|
3
|
Hosseinzadeh N, Asqardokht-Aliabadi A, Sarabi-Aghdam V, Hashemi N, Dogahi PR, Sarraf-Ov N, Homayouni-Rad A. Antioxidant Properties of Postbiotics: An Overview on the Analysis and Evaluation Methods. Probiotics Antimicrob Proteins 2025; 17:606-624. [PMID: 39395091 DOI: 10.1007/s12602-024-10372-7] [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] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Antioxidants found naturally in foods have a significant effect on preventing several human diseases. However, the use of synthetic antioxidants in studies has raised concerns about their potential link to liver disease and cancer. The findings show that postbiotics have the potential to act as a suitable alternative to chemical antioxidants in the food and pharmaceutical sectors. Postbiotics are bioactive compounds generated by probiotic bacteria as they ferment prebiotic fibers in the gut. These compounds can also be produced from a variety of substrates, including non-prebiotic carbohydrates such as starches and sugars, as well as proteins and organic acids, all of which probiotics utilize during the fermentation process. These are known for their antioxidant, antibacterial, anti-inflammatory, and anti-cancer properties that help improve human health. Various methodologies have been suggested to assess the antioxidant characteristics of postbiotics. While there are several techniques to evaluate the antioxidant properties of foods and their bioactive compounds, the absence of a convenient and uncomplicated method is remarkable. However, cell-based assays have become increasingly important as an intermediate method that bridges the gap between chemical experiments and in vivo research due to the limitations of in vitro and in vivo assays. This review highlights the necessity of transitioning towards more biologically relevant cell-based assays to effectively evaluate the antioxidant activity of postbiotics. These experiments are crucial for assessing the biological efficacy of dietary antioxidants. This review focuses on the latest applications of the Caco-2 cell line in the assessment of cellular antioxidant activity (CAA) and bioavailability. Understanding the impact of processing processes on the biological properties of postbiotic antioxidants can facilitate the development of new food and pharmaceutical products.
Collapse
Affiliation(s)
- Negin Hosseinzadeh
- Student Research Committee, Department of Food Science and Technology, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Abolfazl Asqardokht-Aliabadi
- Department of Food Science and Technology, Faculty of Agricultural Engineering, Sari Agricultural Sciences and Natural Resources University, Sari, Iran
| | - Vahideh Sarabi-Aghdam
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Hashemi
- University of Applied Science & Technology, Center of Pardisan Hospitality & Tourism Management, Mashhad, Iran
| | - Parisa Rahimi Dogahi
- Department of Food Science and Technology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Narges Sarraf-Ov
- Student Research Committee, Department of Food Science and Technology, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aziz Homayouni-Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Ngew E, Kollipara R, Bessissow T, Karboune S, George S. Nanoencapsulation enhanced the performance of β-carotene for ameliorating inflammation in patient-derived organoids. Nanomedicine (Lond) 2025; 20:663-675. [PMID: 39943855 PMCID: PMC11970773 DOI: 10.1080/17435889.2025.2465247] [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: 10/22/2024] [Accepted: 02/07/2025] [Indexed: 04/02/2025] Open
Abstract
AIM This study aims to develop a nanocarrier system for the oral delivery of β-Carotene (BC) (as a model therapeutic agent) and to test its efficacy in ameliorating inflammation in an ulcerative colitis (UC) patient-derived organoid. MATERIALS & METHODS BC was encapsulated in a zein protein nano-cage surface-functionalized with pectin and polyethyleneglycol (PEG). The nanoencapsulated BC (nBC) was characterized for physicochemical properties (size, charge, surface chemistry) and functional properties (radical scavenging, mucoadhesion and penetration, release in simulated digestive fluids). Further, we evaluated the performance of nBC in ameliorating inflammation in Caco-2 and UC patient-derived organoid models. RESULTS nBC achieved 75% encapsulation efficiency with improved stability and functional properties when compared to free BC. The nanocarrier was non-cytotoxic and improved mucoadhesion, mucopenetration, and the anti-inflammatory potential of BC. In UC organoids, nBC suppressed dextran sulfate sodium (DSS)-induced TNF-α and IL-8 production by approximately 70% and 31%, respectively, which was significantly higher than free BC at comparable concentrations. CONCLUSIONS The protein-polymer nanoencapsulation strategy showed promise in protecting BC and overcoming intestinal mucus barriers for an improved anti-inflammatory effect in the organoid model. Further studies using animal models are warranted for establishing pharmacokinetics, tissue distribution, and therapeutic index of orally delivered nBC.
Collapse
Affiliation(s)
- Estee Ngew
- Department of Food Science and Agricultural Chemistry, McGill University, Québec, Canada
| | - Revathi Kollipara
- Department of Food Science and Agricultural Chemistry, McGill University, Québec, Canada
| | - Talat Bessissow
- Division of Gastroenterology, McGill University Health Centre, Quebec, Canada
| | - Salwa Karboune
- Department of Food Science and Agricultural Chemistry, McGill University, Québec, Canada
| | - Saji George
- Department of Food Science and Agricultural Chemistry, McGill University, Québec, Canada
| |
Collapse
|
5
|
Dey C, Sommerfeld IK, Bojarová P, Kodra N, Vrbata D, Zimolová Vlachová M, Křen V, Pich A, Elling L. Color-coded galectin fusion proteins as novel tools in biomaterial science. Biomater Sci 2025; 13:1482-1500. [PMID: 39907577 DOI: 10.1039/d4bm01148a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The inherent carbohydrate-binding specificities of human galectins can serve as recognition elements in both biotechnological and biomedical applications. The combination of the carbohydrate-recognition domain (CRD) of galectins fused to peptides or proteins for purification, immobilization, and imaging enables multifunctional utilization within a single protein. We present here a library of color-coded galectin fusion proteins that incorporate a His6-tag, a fluorescent protein, and a SpyCatcher or SpyTag unit to enable immobilization procedures. These galectin fusion proteins exhibit similar binding properties to the non-fused galectins with micromolar apparent binding affinities. N- and C-terminal fusion partners do not interfere with the SpyCatcher/SpyTag immobilization. By applying SpyCatcher/SpyTag-mediated SC-ST-Gal-3 conjugates, we show the stepwise formation of a three-layer ECM-like structure in vitro. Additionally, we demonstrate the SpyCatcher/SpyTag-mediated immobilization of galectins in microgels, which can serve as a transport platform for localized targeting applications. The proof of concept is provided by the galectin-mediated binding of microgels to colorectal cancer cells.
Collapse
Affiliation(s)
- Carina Dey
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| | - Isabel K Sommerfeld
- DWI - Leibniz-Institute for Interactive Materials, e.V. Forckenbeckstr. 50, 52074 Aachen, Germany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
| | - Pavla Bojarová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
- Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, nám. Sítná 3105, 27201 Kladno, Czech Republic
| | - Nikol Kodra
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| | - David Vrbata
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Miluše Zimolová Vlachová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Andrij Pich
- DWI - Leibniz-Institute for Interactive Materials, e.V. Forckenbeckstr. 50, 52074 Aachen, Germany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| |
Collapse
|
6
|
Hwang S, Sung SI, Kim YE, Yang M, Koh A, Ahn SY, Chang YS. Thrombin-preconditioned mesenchymal stromal cell-derived extracellular vesicles attenuate experimental necrotizing enterocolitis. Stem Cell Res Ther 2025; 16:101. [PMID: 40022236 PMCID: PMC11871789 DOI: 10.1186/s13287-025-04243-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a critical gastrointestinal disease in preterm infants, for which no specific treatment is established. We previously demonstrated that thrombin-preconditioned mesenchymal stromal cell-derived extracellular vesicles (thMSC-EVs) enhance protection against other neonatal tissue injuries. Therefore, this study aimed to evaluate the therapeutic potential of thMSC-EVs in modified in vitro, in vivo, and organoid models of NEC. METHODS In vitro, the effects of thMSC-EVs and naïveMSC-EVs were compared in hyperosmotic, ischemic, and hypothermic (HIT)-stressed IEC-6 cells and LPS-treated peritoneal macrophages. In vivo, NEC was induced in P4 mouse pups by three cycles of formula feeding, oral LPS administration, hypoxia, and hypothermia, followed by overnight dam care. 2 × 109 thMSC-EVs were intraperitoneally administered daily for three days, and the therapeutic effects were assessed macroscopically, histologically, and biochemically. NEC mouse-derived organoids were established to evaluate the thMSC-EVs' effect in mature enterocytes. LC-MS/MS was performed to analyze the EV proteomics. RESULTS In vitro, compared with naïveMSC-EVs, thMSC-EVs significantly improved cellular viability in HIT-induced IEC-6 cells and reduced pro-inflammatory (IL-1α, IL-1β, TNF-α) but increased anti-inflammatory (TGF-b) cytokine levels in LPS-treated peritoneal macrophages. In vivo, thMSC-EVs significantly attenuated clinical symptoms, reduced intestinal damage, and retained intestinal stem cell markers, showing more significant localization in NEC-induced intestines than in healthy intestines. In NEC mouse-derived organoids, thMSC-EVs significantly increased OLFM4 and claudin-4 expression and reduced stress-related markers such as sucrase-isomaltase, defensin, and chromogranin A. Proteomic analysis revealed that thMSC-EVs were greater enriched in anti-apoptotic, anti-inflammatory, cell adhesion, and Wnt signaling pathways than naïveMSC-EVs. CONCLUSION thMSC-EVs improved cellular viability, reduced apoptosis, attenuated inflammation, and upregulated key intestinal stem cell markers, collectively suggesting their tissue-protective effects and highlighting their potential as a treatment for NEC.
Collapse
Affiliation(s)
- Sein Hwang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Se In Sung
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Young Eun Kim
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Misun Yang
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, South Korea
| | - So Yoon Ahn
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Yun Sil Chang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
- Cell and Gene Therapy Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea.
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea.
| |
Collapse
|
7
|
Zhou C, Zou Y, Huang H, Zhao F, Fan X, Bai L, Zhang X, Ye K. Virulence expression difference to intestinal cells of different pathogenic Listeria monocytogenes contaminating sausages after simulated digestive tract. Int J Food Microbiol 2025; 430:111067. [PMID: 39813952 DOI: 10.1016/j.ijfoodmicro.2025.111067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
This study investigated the difference in survival among Listeria monocytogenes (LM) 10403S (highly pathogenic strain) and M7 (low pathogenic strain) in sausage under a simulated digestive environment, and established intestinal organoids and macrophages co-culture model to further explore the virulence expression difference to intestinal cells between LM 10403S and M7 after in vitro gastrointestinal digestion. Results showed that, compared with LM M7, LM 10403S exhibited a high survival rate during in vitro digestion, which may be due to the increased expression of stress response-related genes. In addition, the expression of virulence genes in LM 10403S was significantly higher than in LM M7 under the gastrointestinal environment. Furthermore, in the intestinal organoids and macrophages co-culture model infected by LM 10403S and M7 after in vitro gastrointestinal digestion, results showed that, compared with the LM M7 group, the LM 10403S group had significantly lower budding rate and significantly higher mortality of organoids. Also, the significantly increased LDH release and inflammatory factors (TNF-α and IL-1β) in the LM 10403S group were observed, and the main virulence genes (iap, inlA, inlB, actA, hly, plcA, and plcB) of 10403S were significantly highly expressed than LM M7 during the cell infection. These results reflected that the reason for the different pathogenicity between LM 10403S and M7 may be due to the high tolerance and the expression of virulence genes than LM M7 during gastrointestinal digestion and cell infection, which would be expected to provide a better understanding of the infection mechanisms among different pathogenic strains of L. monocytogenes in food.
Collapse
Affiliation(s)
- Cong Zhou
- China National Center for Food Safety Risk Assessment Key Laboratory of Food Safety Risk Assessment, Beijing, PR China; State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Yafang Zou
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Haorui Huang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Fanwen Zhao
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Xia Fan
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Li Bai
- China National Center for Food Safety Risk Assessment Key Laboratory of Food Safety Risk Assessment, Beijing, PR China
| | - Xinhao Zhang
- China National Center for Food Safety Risk Assessment Key Laboratory of Food Safety Risk Assessment, Beijing, PR China
| | - Keping Ye
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
8
|
Steyn JD, Haasbroek-Pheiffer A, Pheiffer W, Weyers M, van Niekerk SE, Hamman JH, van Staden D. Evaluation of Drug Permeation Enhancement by Using In Vitro and Ex Vivo Models. Pharmaceuticals (Basel) 2025; 18:195. [PMID: 40006008 PMCID: PMC11859300 DOI: 10.3390/ph18020195] [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: 12/18/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Drugs administered by means of extravascular routes of drug administration must be absorbed into the systemic circulation, which involves the movement of the drug molecules across biological barriers such as epithelial cells that cover mucosal surfaces or the stratum corneum that covers the skin. Some drugs exhibit poor permeation across biological membranes or may experience excessive degradation during first-pass metabolism, which tends to limit their bioavailability. Various strategies have been used to improve drug bioavailability. Absorption enhancement strategies include the co-administration of chemical permeation enhancers, enzymes, and/or efflux transporter inhibitors, chemical changes, and specialized dosage form designs. Models with physiological relevance are needed to evaluate the efficacy of drug absorption enhancement techniques. Various in vitro cell culture models and ex vivo tissue models have been explored to evaluate and quantify the effectiveness of drug permeation enhancement strategies. This review deliberates on the use of in vitro and ex vivo models for the evaluation of drug permeation enhancement strategies for selected extravascular drug administration routes including the nasal, oromucosal, pulmonary, oral, rectal, and transdermal routes of drug administration.
Collapse
Affiliation(s)
- Johan D. Steyn
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.D.S.); (A.H.-P.); (M.W.); (S.E.v.N.); (J.H.H.)
| | - Anja Haasbroek-Pheiffer
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.D.S.); (A.H.-P.); (M.W.); (S.E.v.N.); (J.H.H.)
| | - Wihan Pheiffer
- Preclinical Drug Development Platform, Faculty of Health Sciences, North-West University, Potchefstroom 2531, South Africa;
| | - Morné Weyers
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.D.S.); (A.H.-P.); (M.W.); (S.E.v.N.); (J.H.H.)
| | - Suzanne E. van Niekerk
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.D.S.); (A.H.-P.); (M.W.); (S.E.v.N.); (J.H.H.)
| | - Josias H. Hamman
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.D.S.); (A.H.-P.); (M.W.); (S.E.v.N.); (J.H.H.)
| | - Daniélle van Staden
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.D.S.); (A.H.-P.); (M.W.); (S.E.v.N.); (J.H.H.)
| |
Collapse
|
9
|
Özkan A, Merry GE, Chou DB, Posey RR, Stejskalova A, Calderon K, Sperry M, Horvath V, Ferri LE, Carlotti E, McDonald SAC, Winton DJ, Riccardi R, Bordeianou L, Hall S, Goyal G, Ingber DE. Human Organ Chips Reveal New Inflammatory Bowel Disease Drivers. RESEARCH SQUARE 2025:rs.3.rs-5627712. [PMID: 39975910 PMCID: PMC11838723 DOI: 10.21203/rs.3.rs-5627712/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Inflammatory bowel disease (IBD) patients exhibit compromised intestinal barrier function and decreased mucus accumulation, as well as increased inflammation, fibrosis, and cancer risk, with symptoms often being exacerbated in women during pregnancy. Here, we show that these IBD hallmarks can be replicated using human Organ Chips lined by IBD patient-derived colon epithelial cells interfaced with matched fibroblasts cultured under flow. Use of heterotypic tissue recombinants revealed that IBD fibroblasts are the primary drivers of multiple IBD symptoms. Inflammation and fibrosis are accentuated by peristalsis-like motions in IBD Chips and when exposed to pregnancy-associated hormones in female IBD Chips. Carcinogen exposure also increases inflammation, gene mutations, and chromosome duplication in IBD Chips, but not in Healthy Chips. These data enabled by human Organ Chip technology suggest that the intestinal stroma, sex hormones, and peristalsis-associated mechanical deformations play a key role in driving inflammation, fibrosis, and disease progression in male and female IBD patients.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Gwenn E. Merry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - David B. Chou
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Ryan R. Posey
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Karina Calderon
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Megan Sperry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Viktor Horvath
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Current address: Entact Bio, Watertown, MA
| | - Lorenzo E. Ferri
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Department of Experimental Surgery and Department of Surgery, McGill University, Montreal, QC, Canada
| | - Emanuela Carlotti
- Clonal Dynamics in Epithelia Laboratory, Queen Mary University of London, London, United Kingdom
| | - Stuart A. C. McDonald
- Clonal Dynamics in Epithelia Laboratory, Queen Mary University of London, London, United Kingdom
| | - Douglas J. Winton
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Rocco Riccardi
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | | | - Sean Hall
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Current address: Iovance Therapeutics, Tampa, FL
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Vascular Biology Program and Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA
| |
Collapse
|
10
|
Özkan A, Merry G, Chou DB, Posey RR, Stejskalova A, Calderon K, Sperry M, Horvath V, Ferri LE, Carlotti E, McDonald SAC, Winton DJ, Riccardi R, Bordeianou L, Hall S, Goyal G, Ingber DE. Inflammatory Bowel Disease Drivers Revealed in Human Organ Chips. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.05.24318563. [PMID: 39677416 PMCID: PMC11643285 DOI: 10.1101/2024.12.05.24318563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Inflammatory bowel disease (IBD) patients exhibit compromised intestinal barrier function and decreased mucus accumulation, as well as increased inflammation, fibrosis, and cancer risk, with symptoms often being exacerbated in women during pregnancy. Here, we show that these IBD hallmarks can be replicated using human Organ Chips lined by IBD patient-derived colon epithelial cells interfaced with matched fibroblasts cultured under flow. Use of heterotypic tissue recombinants revealed that IBD fibroblasts are the primary drivers of multiple IBD symptoms. Inflammation and fibrosis are accentuated by peristalsis-like motions in IBD Chips and when exposed to pregnancy-associated hormones in female IBD Chips. Carcinogen exposure also increases inflammation, gene mutations, and chromosome duplication in IBD Chips, but not in Healthy Chips. These data enabled by human Organ Chip technology suggest that the intestinal stroma and peristalsis-associated mechanical deformations play a key role in driving inflammation and disease progression in male and female IBD patients.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Gwenn Merry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - David B. Chou
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Ryan R. Posey
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Karina Calderon
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Megan Sperry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Viktor Horvath
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Current address: Entact Bio, Watertown, MA
| | - Lorenzo E. Ferri
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Department of Experimental Surgery and Department of Surgery, McGill University, Montreal, QC, Canada
| | - Emanuela Carlotti
- Clonal Dynamics in Epithelia Laboratory, Queen Mary University of London, London, United Kingdom
| | - Stuart A. C. McDonald
- Clonal Dynamics in Epithelia Laboratory, Queen Mary University of London, London, United Kingdom
| | - Douglas J. Winton
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Rocco Riccardi
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | | | - Sean Hall
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Current address: Iovance Therapeutics, Tampa, FL
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
- Vascular Biology Program and Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA
| |
Collapse
|
11
|
Uemura I, Takahashi-Suzuki N, Kita F, Satoh T. Establishment of an in-vitro inflammatory bowel disease model using immunological differentiation of Caco-2 cells. MethodsX 2024; 13:102952. [PMID: 39329151 PMCID: PMC11426153 DOI: 10.1016/j.mex.2024.102952] [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: 05/27/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
Studies on intestinal cell differentiation, particularly in dextran sodium sulfate (DSS)-induced inflammatory bowel disease (IBD), have predominantly focused on the disruption of intestinal crypts and suppressive effects on the intestinal microbiota; however, repeated administration of DSS is required to induce inflammatory pathology, and there is a lack of observation of early responses and consideration of differentiation stages. Although colonic adenocarcinoma (Caco-2) cells can be used as intestinal cell models, research on these cells in an immature state is limited. We, therefore, investigated the relationship between Caco-2 cell culture duration and immunological differentiation using α-defensin5 (DEFA5) as an indicator of intestinal immunity and differentiation. Changes in protein and gene expression levels in response to DSS were examined at each differentiation stage. Expression of immune- and differentiation-related proteins, including DEFA5 and lysozyme, was evident from Day 8 of culture. Immune responses to DSS varied with the differentiation stage, affecting cell viability and cytokine expression.•Caco-2 cell culture duration correlates with the differentiation stage of Paneth cells.•DSS exposure elicits different effects depending on the differentiation stage.•Our in-vitro model of IBD facilitates the characterization of the cell differentiation process and provides a methodology to help elucidate the causal mechanisms of IBD.
Collapse
Affiliation(s)
- Ippei Uemura
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan
| | - Natsuko Takahashi-Suzuki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan
| | - Fumiya Kita
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan
| | - Takashi Satoh
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan
| |
Collapse
|
12
|
Walraven T, Busch M, Wang J, Donkers JM, Duijvestein M, van de Steeg E, Kramer NI, Bouwmeester H. Elevated risk of adverse effects from foodborne contaminants and drugs in inflammatory bowel disease: a review. Arch Toxicol 2024; 98:3519-3541. [PMID: 39249550 PMCID: PMC11489187 DOI: 10.1007/s00204-024-03844-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
The global burden of Inflammatory bowel disease (IBD) has been rising over the last decades. IBD is an intestinal disorder with a complex and largely unknown etiology. The disease is characterized by a chronically inflamed gastrointestinal tract, with intermittent phases of exacerbation and remission. This compromised intestinal barrier can contribute to, enhance, or even enable the toxicity of drugs, food-borne chemicals and particulate matter. This review discusses whether the rising prevalence of IBD in our society warrants the consideration of IBD patients as a specific population group in toxicological safety assessment. Various in vivo, ex vivo and in vitro models are discussed that can simulate hallmarks of IBD and may be used to study the effects of prevalent intestinal inflammation on the hazards of these various toxicants. In conclusion, risk assessments based on healthy individuals may not sufficiently cover IBD patient safety and it is suggested to consider this susceptible subgroup of the population in future toxicological assessments.
Collapse
Affiliation(s)
- Tom Walraven
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands.
| | - Mathias Busch
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Jingxuan Wang
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Joanne M Donkers
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Marjolijn Duijvestein
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Nynke I Kramer
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
13
|
Arcuri S, Pennarossa G, Prasadani M, Gandolfi F, Brevini TAL. Use of Decellularized Bio-Scaffolds for the Generation of a Porcine Artificial Intestine. Methods Protoc 2024; 7:76. [PMID: 39452790 PMCID: PMC11510128 DOI: 10.3390/mps7050076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
In recent years, great interest has been focused on the development of highly reproducible 3D in vitro models that are able to mimic the physiological architecture and functionality of native tissues. To date, a wide range of techniques have been proposed to recreate an intestinal barrier in vitro, including synthetic scaffolds and hydrogels, as well as complex on-a-chip systems and organoids. Here, we describe a novel protocol for the generation of an artificial intestine based on the creation of decellularized bio-scaffolds and their repopulation with intestinal stromal and epithelial cells. Organs collected at the local slaughterhouse are subjected to a decellularization protocol that includes a freezing/thawing step, followed by sequential incubation in 1% SDS for 12 h, 1% Triton X-100 for 12 h, and 2% deoxycholate for 12 h. At the end of the procedure, the generated bio-scaffolds are repopulated with intestinal fibroblasts and then with epithelial cells. The protocol described here represents a promising and novel strategy to generate an in vitro bioengineered intestine platform able to mimic some of the complex functions of the intestinal barrier, thus constituting a promising 3D strategy for nutritional, pharmaceutical, and toxicological studies.
Collapse
Affiliation(s)
- Sharon Arcuri
- Department of Veterinary Medicine, Università degli Studi di Sassari, Via Vienna, 07100 Sassari, Italy;
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy;
| | - Georgia Pennarossa
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy;
| | - Madhusha Prasadani
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 50411 Tartu, Estonia;
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy;
| |
Collapse
|
14
|
Nakazawa M, Nagao I, Ambrosini YM. Proinflammatory cytokines suppress stemness-related properties and expression of tight junction in canine intestinal organoids. In Vitro Cell Dev Biol Anim 2024; 60:916-925. [PMID: 38914841 PMCID: PMC11419940 DOI: 10.1007/s11626-024-00936-w] [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: 01/09/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024]
Abstract
Recent advancements in canine intestinal organoid research have paved the way for the development of enhanced in vitro models, crucial for exploring intestinal physiology and diseases. Despite these strides, there is a notable gap in creating specific in vitro models that focus on intestinal inflammation. Our study aims to bridge this gap by investigating the impact of proinflammatory cytokines on canine intestinal epithelial cells (IECs) within the context of organoid models. Canine intestinal organoids were treated with proinflammatory cytokines TNF-α, IFN-γ, and IL-1β. The expression of stem cell markers Lgr5, Sox9, Hopx, and Olfm4 was evaluated through RT-qPCR, while membrane integrity was assessed using immunofluorescence staining for tight junction proteins and transport assays for permeability. IFN-γ significantly decreased Lgr5 expression, a key intestinal stem cell marker, at both 24 and 48 h post-treatment (p=0.030 and p=0.002, respectively). Conversely, TNF-α increased Olfm4 expression during the same intervals (p=0.018 and p=0.011, respectively). A reduction in EdU-positive cells, indicative of decreased cell proliferation, was observed following IFN-γ treatment. Additionally, a decrease in tight junction proteins E-cadherin and ZO-1 (p<0.001 and p=0.003, respectively) and increased permeability in IECs (p=0.012) were noted, particularly following treatment with IFN-γ. The study highlights the profound impact of proinflammatory cytokines on canine IECs, influencing both stem cell dynamics and membrane integrity. These insights shed light on the intricate cellular processes underlying inflammation in the gut and open avenues for more in-depth research into the long-term effects of inflammation on intestinal health.
Collapse
Affiliation(s)
- Meg Nakazawa
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Itsuma Nagao
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoko M Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
15
|
Rehman NU, Shin SA, Lee CS, Song M, Kim HJ, Chung HJ. Short Caco-2 model for evaluation of drug permeability: A sodium valerate-assisted approach. Int J Pharm 2024; 661:124415. [PMID: 38960340 DOI: 10.1016/j.ijpharm.2024.124415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/10/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
The human colorectal adenocarcinoma cell line Caco-2, widely used for studying intestinal drug permeability, is typically grown on permeable filter supports and matures in 21 days with frequent media changes. The process is labor-intensive, prone to contamination, and has low throughput, contributing to the overall high utilization cost. Efforts to establish a low-cost, high-throughput, and short-duration model have encountered obstacles, such as weaker tight junctions causing monolayer leaks, incomplete differentiation resulting in low transporter expression, intricate and challenging protocols, and cytotoxicity, limiting the usability. Hence, this study aimed to develop a low-cost, efficient, and short-duration model by addressing the aforementioned concerns by customizing the media and finding a safe differentiation inducer. We generated a new rapid model using sodium valerate, which demonstrated sufficient transporter activity, improved monolayer integrity, and higher levels of differentiation markers than the 21-day model. Furthermore, this model exhibited consistent and reliable results when used to evaluate drug permeability over multiple days of repeated use. This study demonstrates the potential of a sodium valerate-assisted abbreviated model for drug permeability assessment with economic and practical advantages.
Collapse
Affiliation(s)
- Naveed Ur Rehman
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Miyoung Song
- Department of Anatomy and Convergence Medical Sciences, College of Medicine, Institute of Medical Sciences, Tyrosine Peptide Multiuse Research Group, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Hyun Joon Kim
- Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Anatomy and Convergence Medical Sciences, College of Medicine, Institute of Medical Sciences, Tyrosine Peptide Multiuse Research Group, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Hye Jin Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| |
Collapse
|
16
|
Banerjee P, Senapati S. Translational Utility of Organoid Models for Biomedical Research on Gastrointestinal Diseases. Stem Cell Rev Rep 2024; 20:1441-1458. [PMID: 38758462 DOI: 10.1007/s12015-024-10733-3] [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] [Accepted: 05/01/2024] [Indexed: 05/18/2024]
Abstract
Organoid models have recently been utilized to study 3D human-derived tissue systems to uncover tissue architecture and adult stem cell biology. Patient-derived organoids unambiguously provide the most suitable in vitro system to study disease biology with the actual genetic background. With the advent of much improved and innovative approaches, patient-derived organoids can potentially be used in regenerative medicine. Various human tissues were explored to develop organoids due to their multifold advantage over the conventional in vitro cell line culture approach and in vivo models. Gastrointestinal (GI) tissues have been widely studied to establish organoids and organ-on-chip for screening drugs, nutraceuticals, and other small molecules having therapeutic potential. The function of channel proteins, transporters, and transmembrane proteins was also explained. The successful application of genome editing in organoids using the CRISPR-Cas approach has been reported recently. GI diseases such as Celiac disease (CeD), Inflammatory bowel disease (IBD), and common GI cancers have been investigated using several patient-derived organoid models. Recent advancements on organoid bio-banking and 3D bio-printing contributed significantly in personalized disease management and therapeutics. This article reviews the available literature on investigations and translational applications of patient-derived GI organoid models, notably on elucidating gut-microbial interaction and epigenetic modifications.
Collapse
Affiliation(s)
- Pratibha Banerjee
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
17
|
Fontes NFDA, Fernandes M, González-Ballesteros N, Rodríguez-Argüelles MC, Gomes AC, Duarte ASG. Exploring the Therapeutic Potential of Green-Synthesized Gold Nanoparticles and Ericaria selaginoides Extract for Inflammatory Bowel Disease. Antioxidants (Basel) 2024; 13:884. [PMID: 39199130 PMCID: PMC11351725 DOI: 10.3390/antiox13080884] [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: 05/22/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024] Open
Abstract
Addressing disease remission and treatment adherence in inflammatory bowel diseases (IBDs), such as Crohn's disease, poses significant challenges due to underlying oxidative and inflammatory processes. Nanotechnology emerges as a promising avenue for enhancing therapeutic outcomes in IBD by optimizing drug bioactivity, reducing toxicity, and extending circulation time. Gold nanoparticles, known for their resistance to gastrointestinal pH and possessing antioxidant and anti-inflammatory properties, offer particular promise. They can be produced by green synthesis with seaweed Ericaria selaginoides (ES), itself associated with gastroprotective and anti-inflammatory activities. In a murine model of Crohn's disease induced with 8% acetic acid, pretreatment with dexamethasone (0.2 mL/30 g) or Au@ES (25 and 50 mg/kg) effectively mitigated inflammatory features. Notably, ES (50 mg/kg) and Au@ES (50 mg/kg) administration resulted in significant reductions in both macroscopic and microscopic inflammation scores compared to the disease control group. Furthermore, these treatments normalized inflammatory cytokine expression while safeguarding myenteric plexus glial cells. They also impeded neutrophil activation, leading to reduced myeloperoxidase activity and lipid peroxidation, coupled with increased glutathione levels. In conclusion, ES and Au@ES exhibit potent efficacy in counteracting inflammation and oxidation processes in an experimental Crohn's disease model, suggesting their potential as alternative therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Nayana Freire de Almeida Fontes
- Departamento de Morfologia, Faculdade de Medicina, Centro de Ciências da Saúde, Universidade Federal do Ceará, Fortaleza 60440-900, Brazil (A.S.G.D.)
| | - Mário Fernandes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | | | | | - Andreia Castro Gomes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Antoniella Souza Gomes Duarte
- Departamento de Morfologia, Faculdade de Medicina, Centro de Ciências da Saúde, Universidade Federal do Ceará, Fortaleza 60440-900, Brazil (A.S.G.D.)
| |
Collapse
|
18
|
Ingegneri M, Braghini MR, Piccione M, De Stefanis C, Mandrone M, Chiocchio I, Poli F, Imbesi M, Alisi A, Smeriglio A, Trombetta D. Citrus Pomace as a Source of Plant Complexes to Be Used in the Nutraceutical Field of Intestinal Inflammation. Antioxidants (Basel) 2024; 13:869. [PMID: 39061937 PMCID: PMC11274116 DOI: 10.3390/antiox13070869] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
This study aims to recover the main by-product of Citrus fruits processing, the raw pomace, known also as pastazzo, to produce plant complexes to be used in the treatment of inflammatory bowel disease (IBD). Food-grade extracts from orange (OE) and lemon (LE) pomace were obtained by ultrasound-assisted maceration. After a preliminary phytochemical and biological screening by in vitro assays, primary and secondary metabolites were characterized by proton nuclear magnetic resonance (1H-NMR) and liquid chromatography coupled to diode array detection and electrospray ionization mass spectrometry (LC-DAD-ESI-MS) analyses. The intestinal bioaccessibility and antioxidant and anti-inflammatory properties were investigated by in vitro simulated gastro-intestinal digestion followed by treatments on a lipopolysaccharide (LPS)-stimulated human colorectal adenocarcinoma cell line (Caco-2). The tight junctions-associated structural proteins (ZO-1, Claudin-1, and Occludin), transepithelial electrical resistance (TEER), reactive oxygen species (ROS)-levels, expression of some key antioxidant (CAT, NRF2 and SOD2) and inflammatory (IL-1β, IL-6, TNF-α, IL-8) genes, and pNFkB p65 nuclear translocation, were evaluated. The OE and LE digesta, which did not show any significant difference in terms of phytochemical profile, showed significant effects in protecting against the LPS-induced intestinal barrier damage, oxidative stress and inflammatory response. In conclusion, both OE and LE emerged as potential candidates for further preclinical studies on in vivo IBD models.
Collapse
Affiliation(s)
- Mariarosaria Ingegneri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (M.I.); (M.I.); (D.T.)
| | - Maria Rita Braghini
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy;
| | - Michela Piccione
- Core Facilities, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy; (M.P.); (C.D.S.)
| | - Cristiano De Stefanis
- Core Facilities, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy; (M.P.); (C.D.S.)
| | - Manuela Mandrone
- Department of Pharmacy and Biotechnology (FaBit), Alma Mater Studiorum, University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (M.M.); (I.C.); (F.P.)
| | - Ilaria Chiocchio
- Department of Pharmacy and Biotechnology (FaBit), Alma Mater Studiorum, University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (M.M.); (I.C.); (F.P.)
| | - Ferruccio Poli
- Department of Pharmacy and Biotechnology (FaBit), Alma Mater Studiorum, University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (M.M.); (I.C.); (F.P.)
| | - Martina Imbesi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (M.I.); (M.I.); (D.T.)
| | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00165 Rome, Italy;
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (M.I.); (M.I.); (D.T.)
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (M.I.); (M.I.); (D.T.)
| |
Collapse
|
19
|
Gulnaz A, Lee KR, Kang MJ, Chang JE, Chae YJ. Roles of breast cancer resistance protein and organic anion transporting polypeptide 2B1 in gastrointestinal toxicity induced by SN-38 under inflammatory conditions. Toxicol Lett 2024; 394:57-65. [PMID: 38423481 DOI: 10.1016/j.toxlet.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Drug transporters are among the factors that determine the pharmacokinetic profiles after drug administration. In this study, we investigated the roles of drug transporters involved in transport of SN-38, which is an active metabolite of irinotecan, in the intestine under inflammatory conditions in vitro and determined their functional consequences. The expression alterations of breast cancer resistance protein (BCRP) and organic anion transporting polypeptide (OATP) 2B1 were determined at the mRNA and protein levels, and the subsequent functional alterations were evaluated via an accumulation study with the representative transporter substrates [prazosin and dibromofluorescein (DBF)] and SN-38. We also determined the cytotoxicity of SN-38 under inflammatory conditions. Decreased BCRP expression and increased OATP2B1 expression were observed under inflammatory conditions in vitro, which led to altered accumulation profiles of prazosin, DBF, and SN-38, and the subsequent cytotoxic profiles of SN-38. Treatment with rifampin or novobiocin supported the significant roles of BCRP and OATP2B1 in the transport and cytotoxic profile of SN-38. Collectively, these results suggest that BCRP and OATP2B1 are involved in the increased cytotoxicity of SN-38 under inflammatory conditions in vitro. Further comprehensive research is warranted to completely understand SN-38-induced gastrointestinal cytotoxicity and aid in the successful treatment of cancer with irinotecan.
Collapse
Affiliation(s)
- Aneela Gulnaz
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Bioscience, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Min-Ji Kang
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Ji-Eun Chang
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea.
| |
Collapse
|
20
|
Jamtsho T, Yeshi K, Perry MJ, Loukas A, Wangchuk P. Approaches, Strategies and Procedures for Identifying Anti-Inflammatory Drug Lead Molecules from Natural Products. Pharmaceuticals (Basel) 2024; 17:283. [PMID: 38543070 PMCID: PMC10974486 DOI: 10.3390/ph17030283] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 04/28/2025] Open
Abstract
Natural products (NPs) have played a vital role in human survival for millennia, particularly for their medicinal properties. Many traditional medicine practices continue to utilise crude plants and animal products for treating various diseases, including inflammation. In contrast, contemporary medicine focuses more on isolating drug-lead compounds from NPs to develop new and better treatment drugs for treating inflammatory disorders such as inflammatory bowel diseases. There is an ongoing search for new drug leads as there is still no cure for many inflammatory conditions. Various approaches and technologies are used in drug discoveries from NPs. This review comprehensively focuses on anti-inflammatory small molecules and describes the key strategies in identifying, extracting, fractionating and isolating small-molecule drug leads. This review also discusses the (i) most used approaches and recently available techniques, including artificial intelligence (AI), (ii) machine learning, and computational approaches in drug discovery; (iii) provides various animal models and cell lines used in in-vitro and in-vivo assessment of the anti-inflammatory potential of NPs.
Collapse
Affiliation(s)
- Tenzin Jamtsho
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia; (K.Y.); (M.J.P.)
- Australian Institute of Tropical Health, and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| | - Karma Yeshi
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia; (K.Y.); (M.J.P.)
- Australian Institute of Tropical Health, and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| | - Matthew J. Perry
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia; (K.Y.); (M.J.P.)
- Australian Institute of Tropical Health, and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| | - Alex Loukas
- Australian Institute of Tropical Health, and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| | - Phurpa Wangchuk
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia; (K.Y.); (M.J.P.)
- Australian Institute of Tropical Health, and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| |
Collapse
|
21
|
Minaychev VV, Smirnova PV, Kobyakova MI, Teterina AY, Smirnov IV, Skirda VD, Alexandrov AS, Gafurov MR, Shlykov MA, Pyatina KV, Senotov AS, Salynkin PS, Fadeev RS, Komlev VS, Fadeeva IS. Low-Temperature Calcium Phosphate Ceramics Can Modulate Monocytes and Macrophages Inflammatory Response In Vitro. Biomedicines 2024; 12:263. [PMID: 38397865 PMCID: PMC10887285 DOI: 10.3390/biomedicines12020263] [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: 12/04/2023] [Revised: 01/09/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
Creating bioactive materials for bone tissue regeneration and augmentation remains a pertinent challenge. One of the most promising and rapidly advancing approaches involves the use of low-temperature ceramics that closely mimic the natural composition of the extracellular matrix of native bone tissue, such as Hydroxyapatite (HAp) and its phase precursors (Dicalcium Phosphate Dihydrate-DCPD, Octacalcium Phosphate-OCP, etc.). However, despite significant scientific interest, the current knowledge and understanding remain limited regarding the impact of these ceramics not only on reparative histogenesis processes but also on the immunostimulation and initiation of local aseptic inflammation leading to material rejection. Using the stable cell models of monocyte-like (THP-1ATRA) and macrophage-like (THP-1PMA) cells under the conditions of LPS-induced model inflammation in vitro, the influence of DCPD, OCP, and HAp on cell viability, ROS and intracellular NO production, phagocytosis, and the secretion of pro-inflammatory cytokines was assessed. The results demonstrate that all investigated ceramic particles exhibit biological activity toward human macrophage and monocyte cells in vitro, potentially providing conditions necessary for bone tissue restoration/regeneration in the peri-implant environment in vivo. Among the studied ceramics, DCPD appears to be the most preferable for implantation in patients with latent inflammation or unpredictable immune status, as this ceramic had the most favorable overall impact on the investigated cellular models.
Collapse
Affiliation(s)
- Vladislav V. Minaychev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
| | - Polina V. Smirnova
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| | - Margarita I. Kobyakova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
| | - Anastasia Yu. Teterina
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| | - Igor V. Smirnov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| | - Vladimir D. Skirda
- Institute of Physics, Kazan Federal University, Kremlyovskaya St. 18, 420008 Kazan, Russia; (V.D.S.); (M.R.G.)
| | - Artem S. Alexandrov
- Institute of Physics, Kazan Federal University, Kremlyovskaya St. 18, 420008 Kazan, Russia; (V.D.S.); (M.R.G.)
| | - Marat R. Gafurov
- Institute of Physics, Kazan Federal University, Kremlyovskaya St. 18, 420008 Kazan, Russia; (V.D.S.); (M.R.G.)
| | - Mikhail A. Shlykov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| | - Kira V. Pyatina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
| | - Anatoliy S. Senotov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
| | - Pavel S. Salynkin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
| | - Roman S. Fadeev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| | - Vladimir S. Komlev
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| | - Irina S. Fadeeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (V.V.M.); (M.I.K.); (A.S.S.); (I.S.F.)
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia; (P.V.S.); (A.Y.T.); (M.A.S.)
| |
Collapse
|
22
|
Tataru C, Livni M, Marean-Reardon C, Franco MC, David M. Cytokine induced inflammatory bowel disease model using organ-on-a-chip technology. PLoS One 2023; 18:e0289314. [PMID: 38091316 PMCID: PMC10718466 DOI: 10.1371/journal.pone.0289314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/14/2023] [Indexed: 12/18/2023] Open
Abstract
Over 2 million people in North America suffer from inflammatory bowel disease (IBD), a chronic and idiopathic inflammatory condition. While previous research has primarily focused on studying immune cells as a cause and therapeutic target for IBD, recent findings suggest that non-immune cells may also play a crucial role in mediating cytokine and chemokine signaling, and therefore IBD symptoms. In this study, we developed an organ-on-a-chip co-culture model of Caco2 epithelial and HUVEC endothelial cells and induced inflammation using pro-inflammatory cytokines TNF-α and IFN-γ. We tested different concentration ranges and delivery orientations (apical vs. basal) to develop a consistently inducible inflammatory response model. We then measured pro-inflammatory cytokines and chemokines IL-6, IL-8, and CXCL-10, as well as epithelial barrier integrity. Our results indicate that this model 1. induces IBD-like cytokine secretion in non-immune cells and 2. decreases barrier integrity, making it a feasible and reliable model to test the direct actions of potential anti-inflammatory therapeutics on epithelial and endothelial cells.
Collapse
Affiliation(s)
- Christine Tataru
- Oregon State University, College of Science, Microbiology, Corvallis, OR, United States of America
| | - Maya Livni
- Oregon State University, College of Science, Microbiology, Corvallis, OR, United States of America
| | - Carrie Marean-Reardon
- Oregon State University, College of Science, Biochemistry and Biophysics, Corvallis, OR, United States of America
| | - Maria Clara Franco
- Oregon State University, College of Science, Biochemistry and Biophysics, Corvallis, OR, United States of America
- Florida International University, Herbert Wertheim College of Medicine, Center for Translational Science, Port St. Lucie, FL, United States of America
| | - Maude David
- Oregon State University, College of Science, Microbiology, Corvallis, OR, United States of America
- Oregon State University, College of Pharmacy, Corvallis, OR, United States of America
| |
Collapse
|
23
|
Wright CW, Li N, Shaffer L, Hill A, Boyer N, Alves SE, Venkataraman S, Biswas K, Lieberman LA, Mohammadi S. Establishment of a 96-well transwell system using primary human gut organoids to capture multiple quantitative pathway readouts. Sci Rep 2023; 13:16357. [PMID: 37773535 PMCID: PMC10541891 DOI: 10.1038/s41598-023-43656-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 09/26/2023] [Indexed: 10/01/2023] Open
Abstract
Disruptions in the gut epithelial barrier can lead to the development of chronic indications such as inflammatory bowel disease (IBD). Historically, barrier function has been assessed in cancer cell lines, which do not contain all human intestinal cell types, leading to poor translatability. To bridge this gap, we adapted human primary gut organoids grown as monolayers to quantify transcription factor phosphorylation, gene expression, cytokine production, and barrier function. In this work we describe and characterize a novel 96-well human gut organoid-derived monolayer system that enables quantitative assessment of candidate therapeutics. Normal human intestine differentiation patterns and barrier function were characterized and confirmed to recapitulate key aspects of in vivo biology. Next, cellular response to TNF-α (a central driver of IBD) was determined using a diverse cadre of quantitative readouts. We showed that TNF-α pathway antagonists rescued damage caused by TNF-α in a dose-dependent manner, indicating that this system is suitable for quantitative assessment of barrier modulating factors. Taken together, we have established a robust primary cell-based 96-well system capable of interrogating questions around mucosal response. This system is well suited to provide pivotal functional data to support translational target and drug discovery efforts.
Collapse
Affiliation(s)
- Charles W Wright
- Discovery Immunology, Merck & Co., Inc., Cambridge, MA, 02141, USA
| | - Naomi Li
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Lynsey Shaffer
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, 02115, USA
- Moderna, Inc., Cambridge, MA, USA
| | - Armetta Hill
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Nicolas Boyer
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Stephen E Alves
- Discovery Immunology, Merck & Co., Inc., Cambridge, MA, 02141, USA
| | | | - Kaustav Biswas
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, 02115, USA
| | | | - Sina Mohammadi
- Discovery Immunology, Merck & Co., Inc., Cambridge, MA, 02141, USA.
| |
Collapse
|
24
|
Chua KJ, Ling H, Hwang IY, Lee HL, March JC, Lee YS, Chang MW. An Engineered Probiotic Produces a Type III Interferon IFNL1 and Reduces Inflammations in in vitro Inflammatory Bowel Disease Models. ACS Biomater Sci Eng 2023; 9:5123-5135. [PMID: 36399014 PMCID: PMC10498420 DOI: 10.1021/acsbiomaterials.2c00202] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022]
Abstract
The etiology of inflammatory bowel diseases (IBDs) frequently results in the uncontrolled inflammation of intestinal epithelial linings and the local environment. Here, we hypothesized that interferon-driven immunomodulation could promote anti-inflammatory effects. To test this hypothesis, we engineered probiotic Escherichia coli Nissle 1917 (EcN) to produce and secrete a type III interferon, interferon lambda 1 (IFNL1), in response to nitric oxide (NO), a well-known colorectal inflammation marker. We then validated the anti-inflammatory effects of the engineered EcN strains in two in vitro models: a Caco-2/Jurkat T cell coculture model and a scaffold-based 3D coculture IBD model that comprises intestinal epithelial cells, myofibroblasts, and T cells. The IFNL1-expressing EcN strains upregulated Foxp3 expression in T cells and thereafter reduced the production of pro-inflammatory cytokines such as IL-13 and -33, significantly ameliorating inflammation. The engineered strains also rescued the integrity of the inflamed epithelial cell monolayer, protecting epithelial barrier integrity even under inflammation. In the 3D coculture model, IFNL1-expressing EcN treatment enhanced the population of regulatory T cells and increased anti-inflammatory cytokine IL-10. Taken together, our study showed the anti-inflammatory effects of IFNL1-expressing probiotics in two in vitro IBD models, demonstrating their potential as live biotherapeutics for IBD immunotherapy.
Collapse
Affiliation(s)
- Koon Jiew Chua
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore,117596, Singapore
- Wilmar-NUS
Corporate Laboratory, National University
of Singapore, 117599, Singapore
| | - Hua Ling
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore,117596, Singapore
- Wilmar-NUS
Corporate Laboratory, National University
of Singapore, 117599, Singapore
| | - In Young Hwang
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore,117596, Singapore
- Wilmar-NUS
Corporate Laboratory, National University
of Singapore, 117599, Singapore
| | - Hui Ling Lee
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore,117596, Singapore
- Wilmar-NUS
Corporate Laboratory, National University
of Singapore, 117599, Singapore
| | - John C. March
- Department
of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Yung Seng Lee
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Department
of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
| | - Matthew Wook Chang
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore,117596, Singapore
- Wilmar-NUS
Corporate Laboratory, National University
of Singapore, 117599, Singapore
| |
Collapse
|
25
|
Kapoor S, Padwad YS. Phloretin suppresses intestinal inflammation and maintained epithelial tight junction integrity by modulating cytokines secretion in in vitro model of gut inflammation. Cell Immunol 2023; 391-392:104754. [PMID: 37506521 DOI: 10.1016/j.cellimm.2023.104754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Ulcerative colitis is a type of inflammatory bowel disease which in long run can lead to colorectal cancer (CRC). Chronic inflammation can be a key factor for the occurrence of CRC thus mitigating an inflammation can be a preventive strategy for the occurrence of CRC. In this study we have explored the anti-inflammatory potential of phloretin, in in vitro gut inflammation model, developed by co-culture of Caco2 (intestinal epithelial) cells and RAW264.7 macrophages (immune cells). Phloretin is a dihydrochalcone present in apple, pear and strawberries. An anti-inflammatory effect of phloretin in reducing LPS induced inflammation and maintenance of transepithelial electric resistance (TEER) in Caco2 cells was examined. Paracellular permeability assay was performed using Lucifer yellow dye to evaluate the effect of phloretin in inhibiting gut leakiness caused by inflammatory mediators secreted by activated macrophages. Phloretin attenuated LPS induced nitric oxide levels, oxidative stress, depolarization of mitochondrial membrane potential in Caco2 cells as evidenced by reduction in reactive oxygen species (ROS), and enhancement of MMP, and decrease in inflammatory cytokines IL8, TNFα, IL1β and IL6. It exhibited anti-inflammatory activity by inhibiting the expression of NFκB, iNOS and Cox2. Phloretin maintained the epithelial integrity by regulating the expression of tight junction proteins ZO1, occludin, Claudin1 and JAM. Phloretin reduced LPS induced levels of Cox2 along with the reduction in Src expression which further regulated an expression of tight junction protein occludin. Phloretin in combination to sodium pyruvate exhibited potential anti-inflammatory activity via targeting NFkB signaling. Our findings paved a way to position phloretin as nutraceutical in preventing the occurrence of colitis and culmination of disease into colitis associated colorectal cancer.
Collapse
Affiliation(s)
- Smita Kapoor
- Pharmacology and Toxicology Lab, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Yogendra S Padwad
- Pharmacology and Toxicology Lab, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
26
|
Dufrusine B, Di Lisio C, Maurizio A, Sallese M, De Laurenzi V, Dainese E. Influence of food emulsifiers on cellular function and inflammation, a preliminary study. Front Nutr 2023; 10:1197686. [PMID: 37599692 PMCID: PMC10434242 DOI: 10.3389/fnut.2023.1197686] [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: 03/31/2023] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
Emulsifiers are extensively used as food additives and their consumption is increasing in Western countries. However, so far only few studies examined their potential effects on intestinal cellular functions and gut inflammation. The aim of this preliminary analysis was to study the emulsifiers and their concentrations capable of causing cellular damage compared to extra virgin olive oil (EVOO). We tested two commonly used emulsifiers (EMI, EMII) and EVOO on Caco-2 cells, derived from a colon carcinoma and widely used as a model of the intestinal inflammation. The diphenyltetrazolium bromide test MTT and clonogenic assay were used to study the effect of emulsifiers on cell viability. Cell migration was determined by the wound-healing assay. The inflammation was studied by measuring the levels of interleukin 6 (IL-6) and monocyte chemoattractant protein-1/C-C motif chemokine ligand 2 (CCL2), multifunctional cytokines with a major role in the acute-phase response. Furthermore, we analyzed the effect of conditioned media of Caco-2 cells treated with EMs on macrophages activation. In conclusion, our preliminary data provide evidence that EMs increase the proliferation and migration rate of Caco-2 cells. Moreover, Caco-2 cells treated with EMs enhance the IL-6 and CCL2 release and activated macrophages, supporting their role as proinflammatory molecules.
Collapse
Affiliation(s)
- Beatrice Dufrusine
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | | | - Michele Sallese
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
27
|
Gabriel-Segard T, Rontard J, Miny L, Dubuisson L, Batut A, Debis D, Gleyzes M, François F, Larramendy F, Soriano A, Honegger T, Paul S. Proof-of-Concept Human Organ-on-Chip Study: First Step of Platform to Assess Neuro-Immunological Communication Involved in Inflammatory Bowel Diseases. Int J Mol Sci 2023; 24:10568. [PMID: 37445748 DOI: 10.3390/ijms241310568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are complex chronic inflammatory disorders of the gastrointestinal (GI) tract. Recent evidence suggests that the gut-brain axis may be pivotal in gastrointestinal and neurological diseases, especially IBD. Here, we present the first proof of concept for a microfluidic technology to model bilateral neuro-immunological communication. We designed a device composed of three compartments with an asymmetric channel that allows the isolation of soma and neurites thanks to microchannels and creates an in vitro synaptic compartment. Human-induced pluripotent stem cell-derived cortical glutamatergic neurons were maintained in soma compartments for up to 21 days. We performed a localized addition of dendritic cells (MoDCs) to either the soma or synaptic compartment. The microfluidic device was coupled with microelectrode arrays (MEAs) to assess the impact on the electrophysiological activity of neurons while adding dendritic cells. Our data highlight that an electrophysiologic signal is transmitted between two compartments of glutamatergic neurons linked by synapses in a bottom-up way when soma is exposed to primed dendritic cells. In conclusion, our study authenticates communication between dendritic cells and neurons in inflammatory conditions such as IBD. This platform opens the way to complexification with gut components to reach a device for pharmacological compound screening by blocking the gut-brain axis at a mucosal level and may help patients.
Collapse
Affiliation(s)
- Tristan Gabriel-Segard
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, 42023 Saint-Etienne, France
- Service de Psychiatrie Transversale, Centre Hospitalo-Universitaire de Saint Etienne, Hôpital Nord, 42055 Saint-Etienne, France
| | | | | | | | | | | | | | - Fabien François
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, 42023 Saint-Etienne, France
| | | | - Alessandra Soriano
- Internal Medicine Department, Gastroenterology Division and IBD Center, Azienda Unità Sanitaria Locale-IRCCS, 42122 Reggio Emilia, Italy
| | | | - Stéphane Paul
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, 42023 Saint-Etienne, France
| |
Collapse
|
28
|
Jang JY, Im E, Kim ND. Therapeutic Potential of Bioactive Components from Scutellaria baicalensis Georgi in Inflammatory Bowel Disease and Colorectal Cancer: A Review. Int J Mol Sci 2023; 24:1954. [PMID: 36768278 PMCID: PMC9916177 DOI: 10.3390/ijms24031954] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Scutellaria baicalensis Georgi (SBG), an herbal medicine with various biological activities, including anti-inflammatory, anticancer, antiviral, antibacterial, and antioxidant activities, is effective in treatment of colitis, hepatitis, pneumonia, respiratory infections, and allergic diseases. This herbal medicine consists of major active substances, such as baicalin, baicalein, wogonoside, and wogonin. Inflammatory bowel disease (IBD) comprises a group of inflammatory conditions of the colon and small intestine, with Crohn's disease and ulcerative colitis being the main types. IBD can lead to serious complications, such as increased risk of colorectal cancer (CRC), one of the most common cancers worldwide. Currently, there is no cure for IBD, and its incidence has been increasing over the past few decades. This review comprehensively summarizes the efficacy of SBG in IBD and CRC and may serve as a reference for future research and development of drugs for IBD and cancer treatment.
Collapse
Affiliation(s)
| | - Eunok Im
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| | - Nam Deuk Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
29
|
Kakni P, Truckenmüller R, Habibović P, van Griensven M, Giselbrecht S. A Microwell-Based Intestinal Organoid-Macrophage Co-Culture System to Study Intestinal Inflammation. Int J Mol Sci 2022; 23:15364. [PMID: 36499691 PMCID: PMC9736416 DOI: 10.3390/ijms232315364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/21/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The mammalian intestinal epithelium contains more immune cells than any other tissue, and this is largely because of its constant exposure to pathogens. Macrophages are crucial for maintaining intestinal homeostasis, but they also play a central role in chronic pathologies of the digestive system. We developed a versatile microwell-based intestinal organoid-macrophage co-culture system that enables us to recapitulate features of intestinal inflammation. This microwell-based platform facilitates the controlled positioning of cells in different configurations, continuous in situ monitoring of cell interactions, and high-throughput downstream applications. Using this novel system, we compared the inflammatory response when intestinal organoids were co-cultured with macrophages versus when intestinal organoids were treated with the pro-inflammatory cytokine TNF-α. Furthermore, we demonstrated that the tissue-specific response differs according to the physical distance between the organoids and the macrophages and that the intestinal organoids show an immunomodulatory competence. Our novel microwell-based intestinal organoid model incorporating acellular and cellular components of the immune system can pave the way to unravel unknown mechanisms related to intestinal homeostasis and disorders.
Collapse
Affiliation(s)
- Panagiota Kakni
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
30
|
Boros É, Hegedűs Z, Kellermayer Z, Balogh P, Nagy I. Global alteration of colonic microRNAome landscape associated with inflammatory bowel disease. Front Immunol 2022; 13:991346. [PMID: 36177008 PMCID: PMC9513375 DOI: 10.3389/fimmu.2022.991346] [Citation(s) in RCA: 4] [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: 07/11/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) is characterized by chronic inflammation of the gastrointestinal tract that associates with, among others, increased risk of colorectal cancer. There is a growing evidence that miRNAs have important roles in pathological processes, such as inflammation or carcinogenesis. Understanding the molecular mechanisms such as alterations in microRNAome upon chronic intestinal inflammation is critical for understanding the exact pathomechanism of IBD. Hence, we conducted a genome wide microRNAome analysis by applying miRNA-Seq in a rat model of experimental colitis, validated the data by QPCR, examined the expression of a selection of precursor and mature miRNAs, performed in depth biological interpretation using Ingenuity Pathway Analysis and tested the obtained results on samples derived from human patients. We identified specific, interdependent expression pattern of activator/repressor transcription factors, miRNAs and their direct targets in the inflamed colon samples. Particularly, decreased expression of the miR-200 family members (miR-200a/b/c,-141, and -429) and miR-27b correlates with the reduced level of their enhancers (HNF1B, E2F1), elevated expression of their repressors (ZEB2, NFKB1) and increased expression of their target genes (ZEB2, RUNX1). Moreover, the marked upregulation of six miR-27b target genes (IFI16, GCA, CYP1B1, RUNX1, MEF2C and MMP13) in the inflamed colon tissues is a possible direct consequence of the lack of repression due to the downregulated miRNA-27b expression. Our data indicate that changes in microRNAome are associated with the pathophysiology of IBD, consequently, microRNAs offer potential targets for the diagnosis, prognosis and treatment of IBD.
Collapse
Affiliation(s)
- Éva Boros
- Seqomics Biotechnology Ltd., Mórahalom, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Zoltán Hegedűs
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, University of Pécs, Pécs, Hungary
- Lymphoid Organogenesis Research Group, Szentágothai János Research Center, University of Pécs, Pécs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, University of Pécs, Pécs, Hungary
- Lymphoid Organogenesis Research Group, Szentágothai János Research Center, University of Pécs, Pécs, Hungary
| | - István Nagy
- Seqomics Biotechnology Ltd., Mórahalom, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| |
Collapse
|
31
|
Zongo AWS, Zogona D, Youssef M, Ye S, Zhan F, Li J, Li B. Senegalia macrostachya seed polysaccharides attenuate inflammation-induced intestinal epithelial barrier dysfunction in a Caco-2 and RAW264.7 macrophage co-culture model by inhibiting the NF-κB/MLCK pathway. Food Funct 2022; 13:11676-11689. [DOI: 10.1039/d2fo02377f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Senegalia macrostachya seed polysaccharides improved the Caco-2 cell monolayer integrity from the inflammatory insult. SMSP2 treatment lowered the inflammatory cytokine release, increased TJ proteins, and downregulated the NF-κB/MLCK pathway.
Collapse
Affiliation(s)
- Abel Wend-Soo Zongo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
- Center for Research in Biological Sciences, Food and Nutrition, Department of Biochemistry and Microbiology, University Joseph Ki-Zerbo, BP 7021 Ouagadougou 03, Burkina Faso
| | - Daniel Zogona
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
- Center for Research in Biological Sciences, Food and Nutrition, Department of Biochemistry and Microbiology, University Joseph Ki-Zerbo, BP 7021 Ouagadougou 03, Burkina Faso
| | - Mahmoud Youssef
- Food Science and Technology Department, Faculty of Agriculture, Al-Azhar University, Cairo, Egypt
| | - Shuxin Ye
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Fuchao Zhan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| |
Collapse
|