1
|
Jeong YE, Shea K, Ford KA. Unraveling Caco-2 cells through functional and transcriptomic assessments. Regul Toxicol Pharmacol 2025; 156:105771. [PMID: 39761805 DOI: 10.1016/j.yrtph.2025.105771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/20/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
The static Caco-2 monolayer is an extensively utilized model for predicting the permeability of small molecules during the drug development process. While these cells can differentiate and develop key functional and morphological features that emulate human enterocytes, they do not fully replicate the complexity of human intestinal physiology. In this study, we investigated functional and morphological aspects of Caco-2 cells, alongside their transcriptomic profiles, with a particular emphasis on genes encoding drug-metabolizing enzymes and drug transporters. We found that Caco-2 cells not only established a robust and bio-relevant permeable intestinal barrier but also demonstrated functional maturity and differentiation in the intestinal epithelium, substantiated by the activities of important enzymes and an efflux transporter. However, our targeted gene expression analyses revealed that substantial disparities were found in mRNA transcript levels among Caco-2 cells and human biopsy samples. These findings highlight that, although Caco-2 cells are valuable for assessing the passive transport of drugs, their accuracy for predicting active transport or small intestinal drug metabolism is constrained by their transcriptomic divergence from human intestinal tissues. This study highlights the importance of understanding the Caco-2 model's inherent limitations and provides insights that could inform its appropriate application in drug development and regulatory decision-making.
Collapse
Affiliation(s)
- Ye Eun Jeong
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| | - Katherine Shea
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| | - Kevin A Ford
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| |
Collapse
|
2
|
Lee J, Menon N, Lim CT. Dissecting Gut-Microbial Community Interactions using a Gut Microbiome-on-a-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302113. [PMID: 38414327 PMCID: PMC11132043 DOI: 10.1002/advs.202302113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/21/2023] [Indexed: 02/29/2024]
Abstract
While the human gut microbiota has a significant impact on gut health and disease, understanding of the roles of gut microbes, interactions, and collective impact of gut microbes on various aspects of human gut health is limited by the lack of suitable in vitro model system that can accurately replicate gut-like environment and enable the close visualization on causal and mechanistic relationships between microbial constitutents and the gut. , In this study, we present a scalable Gut Microbiome-on-a-Chip (GMoC) with great imaging capability and scalability, providing a physiologically relevant dynamic gut-microbes interfaces. This chip features a reproducible 3D stratified gut epithelium derived from Caco-2 cells (µGut), mimicking key intestinal architecture, functions, and cellular complexity, providing a physiolocially relevant gut environment for microbes residing in the gut. Incorporating tumorigenic bacteria, enterotoxigenic Bacteroides fragilis (ETBF), into the GMoC enable the observation of pathogenic behaviors of ETBF, leading to µGut disruption and pro-tumorigenic signaling activations. Pre-treating the µGut with a beneficial gut microbe Lactobacillus spp., effectively prevent ETBF-mediated gut pathogenesis, preserving the healthy state of the µGut through competition-mediated colonization resistance. The GMoC holds potential as a valuable tool for exploring unknown roles of gut microbes in microbe-induced pathogenesis and microbe-based therapeutic development.
Collapse
Affiliation(s)
- Jeeyeon Lee
- Institute for Health Innovation and Technology (iHealthtech)National University of SingaporeSingapore117599Singapore
| | - Nishanth Menon
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology (iHealthtech)National University of SingaporeSingapore117599Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Mechanobiology InstituteNational University of SingaporeSingapore117411Singapore
| |
Collapse
|
3
|
Pantalone MR, Almazan NM, Lattanzio R, Taher C, De Fabritiis S, Valentinuzzi S, Bishehsari F, Mahdavinia M, Verginelli F, Rahbar A, Mariani-Costantini R, Söderberg-Naucler C. Human cytomegalovirus infection enhances 5‑lipoxygenase and cycloxygenase‑2 expression in colorectal cancer. Int J Oncol 2023; 63:116. [PMID: 37654195 PMCID: PMC10546380 DOI: 10.3892/ijo.2023.5564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/07/2023] [Indexed: 09/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common and fatal types of cancer. Inflammation promotes CRC development, however, the underlying etiological factors are unknown. Human cytomegalovirus (HCMV), a virus that induces inflammation and other cancer hallmarks, has been detected in several types of malignancy, including CRC. The present study investigated whether HCMV infection was associated with expression of the pro‑inflammatory enzymes 5‑lipoxygenase (5‑LO) and cyclooxygenase‑2 (COX‑2) and other molecular, genetic and clinicopathological CRC features. The present study assessed 146 individual paraffin‑embedded CRC tissue microarray (TMA) cores already characterized for TP53 and KRAS mutations, microsatellite instability (MSI) status, Ki‑67 index and EGFR by immunohistochemistry (IHC). The cores were further analyzed by IHC for the expression of two HCMV proteins (Immediate Early, IE and pp65) and the inflammatory markers 5‑LO and COX‑2. The CRC cell lines Caco‑2 and LS‑174T were infected with HCMV strain VR1814, treated with antiviral drug ganciclovir (GCV) and/or anti‑inflammatory drug celecoxib (CCX) and analyzed by reverse transcription‑quantitative PCR and immunofluorescence for 5‑LO, COX‑2, IE and pp65 transcripts and proteins. HCMV IE and pp65 proteins were detected in ~90% of the CRC cases tested; this was correlated with COX‑2, 5‑LO and KI‑67 expression, but not with EGFR immunostaining, TP53 and KRAS mutations or MSI status. In vitro, HCMV infection upregulated 5‑LO and COX‑2 transcript and proteins in both Caco‑2 and LS‑174T cells and enhanced cell proliferation as determined by MTT assay. Treatment with GCV and CCX significantly decreased the transcript levels of COX‑2, 5‑LO, HCMV IE and pp65 in infected cells. HCMV was widely expressed in CRC and may promote inflammation and serve as a potential new target for CRC therapy.
Collapse
Affiliation(s)
- Mattia Russel Pantalone
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, 17164 Stockholm, Sweden
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
| | - Nerea Martin Almazan
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Laboratory Medicine, Unit of Microbial Pathogenesis, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rossano Lattanzio
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, G. d'Annunzio University, I-66100 Chieti, Italy
| | - Chato Taher
- Department of Basic Sciences, Hawler Medical University, Erbil 44001, Iraq
| | - Simone De Fabritiis
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, G. d'Annunzio University, I-66100 Chieti, Italy
| | - Silvia Valentinuzzi
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, I-66100 Chieti, Italy
| | - Faraz Bishehsari
- Division of Digestive Diseases, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA
- Digestive Disease Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran 14114, Iran
| | - Mahboobeh Mahdavinia
- Digestive Disease Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran 14114, Iran
- Department of Internal Medicine, Division of Allergy and Immunology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fabio Verginelli
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, I-66100 Chieti, Italy
| | - Afsar Rahbar
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, 17164 Stockholm, Sweden
| | | | - Cecilia Söderberg-Naucler
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, 17164 Stockholm, Sweden
- MediCity Research Laboratory, University of Turku, FI-20014 Turku, Finland
- Institute of Biomedicine, University of Turku, FI-20014 Turku, Finland
| |
Collapse
|
4
|
Donnaloja F, Izzo L, Campanile M, Perottoni S, Boeri L, Fanizza F, Sardelli L, Jacchetti E, Raimondi MT, Rito LD, Craparotta I, Bolis M, Giordano C, Albani D. Human gut epithelium features recapitulated in MINERVA 2.0 millifluidic organ-on-a-chip device. APL Bioeng 2023; 7:036117. [PMID: 37736017 PMCID: PMC10511260 DOI: 10.1063/5.0144862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023] Open
Abstract
We developed an innovative millifluidic organ-on-a-chip device, named MINERVA 2.0, that is optically accessible and suitable to serial connection. In the present work, we evaluated MINERVA 2.0 as millifluidic gut epithelium-on-a-chip by using computational modeling and biological assessment. We also tested MINERVA 2.0 in a serially connected configuration prodromal to address the complexity of multiorgan interaction. Once cultured under perfusion in our device, human gut immortalized Caco-2 epithelial cells were able to survive at least up to 7 days and form a three-dimensional layer with detectable tight junctions (occludin and zonulin-1 positive). Functional layer development was supported by measurable trans-epithelial resistance and FITC-dextran permeability regulation, together with mucin-2 expression. The dynamic culturing led to a specific transcriptomic profile, assessed by RNASeq, with a total of 524 dysregulated transcripts (191 upregulated and 333 downregulated) between static and dynamic condition. Overall, the collected results suggest that our gut-on-a-chip millifluidic model displays key gut epithelium features and, thanks to its modular design, may be the basis to build a customizable multiorgan-on-a-chip platform.
Collapse
Affiliation(s)
- Francesca Donnaloja
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Marzia Campanile
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Francesca Fanizza
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Lorenzo Sardelli
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Manuela T. Raimondi
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Laura Di Rito
- Department of Oncology, Computational Oncology Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Craparotta
- Department of Oncology, Computational Oncology Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Bolis
- Department of Oncology, Computational Oncology Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
5
|
Scheller AS, Philipp TM, Klotz LO, Steinbrenner H. Altered Capacity for H 2S Production during the Spontaneous Differentiation of Caco-2 Cells to Colonocytes Due to Reciprocal Regulation of CBS and SELENBP1. Antioxidants (Basel) 2022; 11:antiox11101957. [PMID: 36290680 PMCID: PMC9598602 DOI: 10.3390/antiox11101957] [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: 08/19/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Hydrogen sulfide (H2S) has been proposed to promote tumor growth. Elevated H2S levels have been detected in human colorectal cancer (CRC) biopsies, resulting from the selective upregulation of cystathionine β-synthase (CBS). In contrast, the recently identified novel H2S-generating enzyme, selenium-binding protein 1 (SELENBP1), is largely suppressed in tumors. Here, we provide the first comparative analysis of the four human H2S-producing enzymes and the key H2S-catabolizing enzyme, sulfide:quinone oxidoreductase (SQOR), in Caco-2 human colorectal adenocarcinoma cells. The gene expression pattern of proliferating Caco-2 cells parallels that of CRC, while confluent cells undergo spontaneous differentiation to a colonocyte-like phenotype. SELENBP1 and SQOR were strongly upregulated during spontaneous differentiation, whereas CBS was downregulated. Cystathionine γ-lyase and 3-mercaptopyruvate sulfurtransferase remained unaffected. Terminally differentiated cells showed an enhanced capacity to produce H2S from methanethiol and homocysteine. Differentiation induced by exposure to butyrate also resulted in the upregulation of SELENBP1, accompanied by increased SELENBP1 promoter activity. In contrast to spontaneous differentiation, however, butyrate did not cause downregulation of CBS. In summary, SELENBP1 and CBS are reciprocally regulated during the spontaneous differentiation of Caco-2 cells, thus paralleling their opposing regulation in CRC. Butyrate exposure, while imitating some aspects of spontaneous differentiation, does not elicit the same expression patterns of genes encoding H2S-modulating enzymes.
Collapse
|
6
|
El-Maadawy WH, Hafiz E, Okasha H, Osman NA, Ali GH, Hussein RA. Phycocyanin stimulates ulcerative colitis healing via selective activation of cannabinoid receptor-2, intestinal mucosal healing, Treg accumulation, and p38MAPK/MK2 signaling inhibition. Life Sci 2022; 305:120741. [PMID: 35777583 DOI: 10.1016/j.lfs.2022.120741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/16/2022] [Accepted: 06/24/2022] [Indexed: 11/19/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory condition that until this date, lacks curative treatments. Previously, synthetic selective CB2 receptor (CB2R) agonists demonstrated effective preclinical anti-inflammatory activities in UC. Phycocyanin (PC), photosynthetic assistant protein isolated from Microcystis aeruginosa Kützing blue green algae, has multiple pharmacological effects, however, it's effect against UC remains unexplored. Our study aimed at investigating the therapeutic effectiveness of PC against UC, and correlating its mechanisms with CB2R agonistic activities. In silico; PC showed structural similarity with endocannabinoid receptors' ligand "Δ9-tetrahydrocannabinol", target prediction studies suggested high affinity for G-coupled protein family-receptors, and molecular docking affirmed preferable affinity towards CB2R vs CB1R. In LPS-exposed-Caco-2 cell line; PC demonstrated comparable interaction with CB2R, and downregulation of CB2R, p38 and MK2 gene expressions with reference agonist "6d", and exhibited preferred selectivity towards CB2R over CB1R. In DSS-induced mice; PC-treatment ameliorated DSS-induced colon shortening, elevated disease activity index, and colonic pathological alterations. PC showed effective CB2R activation through potent anti-inflammatory activities, Treg-cell accumulation, suppression in p38MAPK/MK2 signaling, and tight junction barrier restoration as indicated by ultrastructural examinations, elevated ZO-1 and occludin protein expressions, and Ki67 immunohistochemical expression in colonic tissues. Additionally, PC alleviated intestinal dysbiosis via downregulating LPS/TLR4/NF-κB signaling and gut microbiota maintenance. Notably, PC-protective activities were abolished when co-administered with SR144528 (selective CB2 antagonist) except for gut microbiota maintenance, which was independent from CB2R activation. Our findings provide evidence of PC effectiveness against UC through acting as CB2R agonist, thus expanding its possible therapeutic application against other inflammatory diseases.
Collapse
Affiliation(s)
- Walaa H El-Maadawy
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt.
| | - Ehab Hafiz
- Electron Microscopy Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt
| | - Hend Okasha
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt
| | - Noha A Osman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Gamila H Ali
- Water Pollution Department, National Research Centre, 33 El Buhouth St., Dokki, Giza, P.O.12622, Egypt
| | - Rehab Ali Hussein
- Pharmacognosy Department, National Research Centre, 33 El Buhouth St., Dokki, Giza, P.O.12622, Egypt.
| |
Collapse
|
7
|
Lofft Z, Taibi A, Massara P, Tokar T, Paetau‐Robinson I, Khoo C, Comelli EM. Cranberry proanthocyanidin and its microbial metabolite 3,4‐dihydroxyphenylacetic acid, but not 3‐(4‐hydroxyphenyl)‐propionic acid, partially reverse pro‐inflammatory microRNA responses in human intestinal epithelial cells. Mol Nutr Food Res 2022; 66:e2100853. [DOI: 10.1002/mnfr.202100853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/13/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Zoe Lofft
- Department of Nutritional Sciences University of Toronto ON Canada
| | - Amel Taibi
- Department of Nutritional Sciences University of Toronto ON Canada
| | - Paraskevi Massara
- Department of Nutritional Sciences University of Toronto ON Canada
- Translational Medicine Program Hospital for Sick Children Toronto Canada
| | - Tomas Tokar
- Krembil Research Institute University Health Network Toronto ON M5T 0S8 Canada
| | | | - Christina Khoo
- Ocean Spray Cranberries, Inc. Lakeville‐Middleboro MA USA
| | - Elena M. Comelli
- Department of Nutritional Sciences University of Toronto ON Canada
- Joannah and Brian Lawson Centre for Child Nutrition University of Toronto ON Canada
| |
Collapse
|
8
|
Chong HB, Youn J, Shin W, Kim HJ, Kim DS. Multiplex recreation of human intestinal morphogenesis on a multi-well insert platform by basolateral convective flow. LAB ON A CHIP 2021; 21:3316-3327. [PMID: 34323906 DOI: 10.1039/d1lc00404b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Here, we report a multiplex culture system that enables simultaneous recreation of multiple replications of the three-dimensional (3D) microarchitecture of the human intestinal epithelium in vitro. The "basolateral convective flow-generating multi-well insert platform (BASIN)" contains 24 nano-porous inserts and an open basolateral chamber applying controllable convective flow in the basolateral compartment that recreates a biomimetic morphogen gradient using a conventional orbital shaker. The mechanistic approach by which the removal of morphogen inhibitors in the basolateral medium can induce intestinal morphogenesis was applied to manipulate the basolateral convective flow in space and time. In a multiplex BASIN, we successfully regenerated a 3D villi-like intestinal microstructure using the Caco-2 human intestinal epithelium that presents high barrier function with minimal insert-to-insert variations. The enhanced cytodifferentiation and proliferation of the 3D epithelial layers formed in the BASIN were visualized with markers of absorptive (villin) and proliferative cells (Ki67). The paracellular transport and efflux profiles of the microengineered 3D epithelial layers in the BASIN confirmed its reproducibility, robustness, and scalability for multiplex biochemical or pharmaceutical studies. Finally, the BASIN was used to investigate the effects of dextran sodium sulfate on the intestinal epithelial barrier and morphology to validate its practical applicability for investigating the effects of external chemicals on the intestinal epithelium and constructing a leaky-gut model. We envision that the BASIN may provide an improved multiplex, scalable, and physiological intestinal epithelial model that is readily accessible to researchers in both basic and applied sciences.
Collapse
Affiliation(s)
- Hyeon Beom Chong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea.
| | | | | | | | | |
Collapse
|
9
|
Zhou K, Amiri M, Salari A, Yu Y, Xu H, Seidler U, Nikolovska K. Functional characterization of the sodium/hydrogen exchanger 8 and its role in proliferation of colonic epithelial cells. Am J Physiol Cell Physiol 2021; 321:C471-C488. [PMID: 34288721 DOI: 10.1152/ajpcell.00582.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Intestinal NaCl, HCO3-, and fluid absorption are strongly dependent on apical Na+/H+ exchange. The intestine expresses three presumably apical sodium-hydrogen exchanger (NHE) isoforms: NHE2, NHE3, and NHE8. We addressed the role of NHE8 [solute carrier 9A8 (SLC9A8)] and its interplay with NHE2 (SLC9A2) in luminal proton extrusion during acute and chronic enterocyte acidosis and studied the differential effects of NHE8 and NHE2 on enterocyte proliferation. In contrast to NHE3, which was upregulated in differentiated versus undifferentiated colonoids, the expression of NHE2 and NHE8 remained constant during differentiation of colonoids and Caco2Bbe cells. Heterogeneously expressed Flag-tagged rat (r)Nhe8 and human (h)NHE8 translocated to the apical membrane of Caco2Bbe cells. rNhe8 and hNHE8, when expressed in NHE-deficient PS120 fibroblasts showed higher sensitivity to HOE642 compared to NHE2. Lentiviral shRNA knockdown of endogenous NHE2 in Caco2Bbe cells (C2Bbe/shNHE2) resulted in a decreased steady-state intracellular pH (pHi), an increased NHE8 mRNA expression, and augmented NHE8-mediated apical NHE activity. Lentiviral shRNA knockdown of endogenous NHE8 in Caco2Bbe cells (C2Bbe/shNHE8) resulted in a decreased steady-state pHi as well, accompanied by decreased NHE2 mRNA expression and activity, which together contributed to reduced apical NHE activity in the NHE8-knockdown cells. Chronic acidosis increased NHE8 but not NHE2 mRNA expression. Alterations in NHE2 and NHE8 expression/activity affected proliferation, with C2Bbe/shNHE2 cells having lower and C2Bbe/shNHE8 having higher proliferative capacity, accompanied by amplified ERK1/2 signaling pathway and increased EGFR expression in the latter cell line. Thus, both Na+/H+ exchangers have distinct functions during cellular homeostasis by triggering different signaling pathways to regulate cellular proliferation and pHi control.
Collapse
Affiliation(s)
- Kunyan Zhou
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Azam Salari
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Yan Yu
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hua Xu
- Department of Pediatrics, University of Arizona Health Science Center, Tucson, Arizona
| | - Ursula Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Katerina Nikolovska
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Fritsche E, Haarmann-Stemmann T, Kapr J, Galanjuk S, Hartmann J, Mertens PR, Kämpfer AAM, Schins RPF, Tigges J, Koch K. Stem Cells for Next Level Toxicity Testing in the 21st Century. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006252. [PMID: 33354870 DOI: 10.1002/smll.202006252] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Indexed: 06/12/2023]
Abstract
The call for a paradigm change in toxicology from the United States National Research Council in 2007 initiates awareness for the invention and use of human-relevant alternative methods for toxicological hazard assessment. Simple 2D in vitro systems may serve as first screening tools, however, recent developments infer the need for more complex, multicellular organotypic models, which are superior in mimicking the complexity of human organs. In this review article most critical organs for toxicity assessment, i.e., skin, brain, thyroid system, lung, heart, liver, kidney, and intestine are discussed with regards to their functions in health and disease. Embracing the manifold modes-of-action how xenobiotic compounds can interfere with physiological organ functions and cause toxicity, the need for translation of such multifaceted organ features into the dish seems obvious. Currently used in vitro methods for toxicological applications and ongoing developments not yet arrived in toxicity testing are discussed, especially highlighting the potential of models based on embryonic stem cells and induced pluripotent stem cells of human origin. Finally, the application of innovative technologies like organs-on-a-chip and genome editing point toward a toxicological paradigm change moves into action.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | | | - Julia Kapr
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Saskia Galanjuk
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Hartmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, Magdeburg, 39106, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| |
Collapse
|
11
|
Ribeiro DL, Machado ART, Machado C, Ferro Aissa A, Dos Santos PW, Barcelos GRM, Antunes LMG. p-synephrine induces transcriptional changes via the cAMP/PKA pathway but not cytotoxicity or mutagenicity in human gastrointestinal cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:196-212. [PMID: 33292089 DOI: 10.1080/15287394.2020.1855490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
p-Synephrine (SN) is an alkaloid added to thermogenic formulations for weight loss that is predominantly absorbed in the human gastrointestinal tract (GI). As the adverse effects of SN on GI cells remain unclear, the aim of present study was to examine whether SN affected cell viability, cell cycle kinetics, genomic stability, redox status, and expression of cAMP/PKA pathway genes related to metabolism/energy homeostasis in stomach mucosa (MNP01) and colon adenocarcinoma (Caco-2) human cells. p-Synephrine at 25-5000 μM was not cytotoxic to both cell lines. At 2-200 μM, SN increased the formation of reactive oxygen species (ROS) but also enhanced levels of antioxidant defense molecules glutathione (GSH) and catalase (CAT) activity, which may account for the absence of cytotoxicity/mutagenicity in both cell lines. SN induced expression of the cAMP/PKA pathway genes ADCY3 and MAPK1 in MNP01 cells and MAPK1, GNAS, PRKACA, and PRKAR2A in Caco-2 cells, as well as modulated the transcription of genes related to cell proliferation (JUN; AKT1) and inflammation (RELA; TNF) in both cell lines. Therefore, the improved antioxidant state mitigated pro-oxidative effects attributed to SN. Evidence indicates that SN does not appear to exhibit adverse potential but modulated the cAMP/PKA pathway in human GI cell lines.
Collapse
Affiliation(s)
- Diego Luis Ribeiro
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo ,Ribeirão Preto, Brazil
| | - Ana Rita Thomazela Machado
- Department Of Clinical Analyses, Toxicology, And Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo , : Ribeirão Preto, Brazil
| | - Carla Machado
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo ,Ribeirão Preto, Brazil
| | - Alexandre Ferro Aissa
- Department Of Clinical Analyses, Toxicology, And Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo , : Ribeirão Preto, Brazil
| | - Patrick Wellington Dos Santos
- Department Of Clinical Analyses, Toxicology, And Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo , : Ribeirão Preto, Brazil
| | | | - Lusânia Maria Greggi Antunes
- Department Of Clinical Analyses, Toxicology, And Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo , : Ribeirão Preto, Brazil
| |
Collapse
|
12
|
Differentiated Caco-2 cell models in food-intestine interaction study: Current applications and future trends. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2020.11.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
13
|
Li B, Wu RY, Horne RG, Ahmed A, Lee D, Robinson SC, Zhu H, Lee C, Cadete M, Johnson-Henry KC, Landberg E, Alganabi M, Abrahamsson T, Delgado-Olguin P, Pierro A, Sherman PM. Human Milk Oligosaccharides Protect against Necrotizing Enterocolitis by Activating Intestinal Cell Differentiation. Mol Nutr Food Res 2020; 64:e2000519. [PMID: 32926533 DOI: 10.1002/mnfr.202000519] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/09/2020] [Indexed: 02/06/2023]
Abstract
SCOPE Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency and currently the leading cause of mortality in preterm infants. Recent studies show that human milk oligosaccharides (HMOs) reduce the frequency and incidence of NEC; however, the molecular mechanisms for their protection are largely unexplored. METHODS AND RESULTS To address this gap, a genome-wide profiling of the intestinal epithelial transcriptome in response to HMOs using RNA-sequencing is performed. It is found that HMOs alter the host transcriptome in 225 unique target genes pertaining to cell proliferation and differentiation, including upregulation of stem cell differentiation marker HMGCS2. To validate these results, differentiation in Caco-2Bbe1 (Caco-2) intestinal cells is verified by Alcian Blue staining and transepithelial electrical resistance (TER) recordings. Furthermore, an in vivo model of NEC is also employed whereby neonatal pups are gavage fed HMOs. Interestingly, HMOs-fed pups show enhanced cell MUC2 differentiation and HMGCS2 expression. CONCLUSIONS These findings demonstrate HMOs protect against NEC in part by altering the differentiation of the crypt-villus axis. In addition, this study suggests that pooled HMOs directly induce a series of biological processes, which provide mechanistic insights to how HMOs protect the host intestine.
Collapse
Affiliation(s)
- Bo Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Richard You Wu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Rachael G Horne
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Abdalla Ahmed
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A1, Canada
| | - Dorothy Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Shaiya C Robinson
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Haitao Zhu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Carol Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Marissa Cadete
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Kathene C Johnson-Henry
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Eva Landberg
- Department of Clinical Chemistry, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mashriq Alganabi
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Thomas Abrahamsson
- Biomedical and Clinical Science and Department of Pediatrics, Linköping University, Linköping, Sweden
| | - Paul Delgado-Olguin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A1, Canada.,Heart & Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, ON, M5S 3H2, Canada
| | - Agostino Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Philip M Sherman
- Cell Biology Program, The Hospital for Sick Children, Toronto, M5G 1X8, Canada.,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| |
Collapse
|
14
|
Proquin H, Jonkhout MCM, Jetten MJ, van Loveren H, de Kok TM, Briedé JJ. Transcriptome changes in undifferentiated Caco-2 cells exposed to food-grade titanium dioxide (E171): contribution of the nano- and micro- sized particles. Sci Rep 2019; 9:18287. [PMID: 31797963 PMCID: PMC6893026 DOI: 10.1038/s41598-019-54675-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
The food additive titanium dioxide (TiO2), or E171, is a white food colorant. Recent studies showed after E171 ingestion a significantly increased number of colorectal tumours in a colorectal cancer mouse model as well as inflammatory responses and dysregulation of the immune system in the intestine of rats. In the mouse colon, E171 induced gene expression changes related to oxidative stress, impairment of the immune system, activation of signalling and cancer-related processes. E171 comprises nanoparticles (NPs) and microparticles (MPs). Previous in vitro studies showed that E171, NPs and MPs induced oxidative stress responses, DNA damage and micronuclei formation. This study aimed to investigate the relative contribution of the NPs and MPs to effects of E171 at the transcriptome level in undifferentiated Caco-2 cells by genome wide microarray analysis. The results showed that E171, NPs, and MPs induce gene expression changes related to signalling, inflammation, immune system, transport and cancer. At the pathway level, metabolism of proteins with the insulin processing pathway and haemostasis were specific to E171 exposure. The gene expression changes associated with the immune system and inflammation induced by E171, MPs, and NPs suggest the creation of a favourable environment for colon cancer development.
Collapse
Affiliation(s)
- Héloïse Proquin
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Marloes C M Jonkhout
- Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, box 901 3000, Leuven, Belgium
| | - Marlon J Jetten
- Complex Tissue Regeneration (CTR), Institute for Technology-Inspired Regenerative Medicine (MERLN), Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Henk van Loveren
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Theo M de Kok
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Jacob J Briedé
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands.
| |
Collapse
|
15
|
Tartakover Matalon S, Ringel Y, Konikoff F, Drucker L, Pery S, Naftali T. Cannabinoid receptor 2 agonist promotes parameters implicated in mucosal healing in patients with inflammatory bowel disease. United European Gastroenterol J 2019; 8:271-283. [PMID: 32213014 DOI: 10.1177/2050640619889773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cannabis benefits patients with inflammatory bowel disease (IBD). Cannabinoid receptors are expressed in gut immune cells and in epithelial cells of inflamed guts. Mucosal healing (MH) requires epithelial layer restoration. OBJECTIVE To analyze the effects of CB2 agonist on parameters implicated in gut inflammation and MH. METHODS Mucosal samples from areas of inflamed/uninflamed colon from 16 patients with IBD were cultured without/with cannabinoid receptor 2 (CB2) agonist (JWH-133, 10 µM, 6 hours (hr)), and analyzed for epithelial/stromal cell proliferation, apoptosis (secretome matrix metalloproteinase 9 (MMP9) activity, which impairs epithelial permeability) and interleukin-8 (IL-8) levels (n = 5-9). In addition, Caco-2 (colon carcinoma epithelial cells) were cultured with biopsy secretomes (48 hr), and analyzed for phenotype and protein markers of proliferation (proliferating cell nuclear antigen), autophagy (LC3IIB) and permeability (Zonula occludens-1) (n = 4-6). RESULTS Uninflamed tissue had higher epithelial proliferation (Ki67: 50%↑, p < 0.05), and reduced secretome MMP9 activity and IL-8 levels (>50%↓, p < 0.05) compared to inflamed tissue. Treatment with CB2 agonist had no effect on epithelial apoptosis, but increased epithelial Ki67 expression (25%), and reduced secretome MMP9 and IL-8 levels in inflamed biopsies. Secretomes of CB2-treated biopsies increased Caco-2 number, migration, proliferating cell nuclear antigen and LC3IIB expression (all, p < 0.05), but had no effect on ZO-1. CONCLUSION Using ex vivo and in vitro human models, we demonstrated that manipulating the cannabinoid system affects colon cells and secretome characteristics that facilitate MH in IBD.
Collapse
Affiliation(s)
- Shelly Tartakover Matalon
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Ringel
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel.,Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, 4107 Bioinformatics Building, 130 Mason Farm Road, Chapel Hill, NC, 27599-7080, USA
| | - Fred Konikoff
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel
| | - Liat Drucker
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Oncogenetics Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Shaul Pery
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Timna Naftali
- Gastroenterology Laboratory, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel
| |
Collapse
|
16
|
A prognostic index based on an eleven gene signature to predict systemic recurrences in colorectal cancer. Exp Mol Med 2019; 51:1-12. [PMID: 31578316 PMCID: PMC6802642 DOI: 10.1038/s12276-019-0319-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/12/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023] Open
Abstract
Approximately half of colorectal cancer (CRC) patients experience disease recurrence and metastasis, and these individuals frequently fail to respond to treatment due to their clinical and biological diversity. Here, we aimed to identify a prognostic signature consisting of a small gene group for precisely predicting CRC heterogeneity. We performed transcriptomic profiling using RNA-seq data generated from the primary tissue samples of 130 CRC patients. A prognostic index (PI) based on recurrence-associated genes was developed and validated in two larger independent CRC patient cohorts (n = 795). The association between the PI and prognosis of CRC patients was evaluated using Kaplan–Meier plots, log-rank tests, a Cox regression analysis and a RT-PCR analysis. Transcriptomic profiling in 130 CRC patients identified two distinct subtypes associated with systemic recurrence. Pathway enrichment and RT-PCR analyses revealed an eleven gene signature incorporated into the PI system, which was a significant prognostic indicator of CRC. Multivariate and subset analyses showed that PI was an independent risk factor (HR = 1.812, 95% CI = 1.342–2.448, P < 0.001) with predictive value to identify low-risk stage II patients who responded the worst to adjuvant chemotherapy. Finally, a comparative analysis with previously reported Consensus Molecular Subgroup (CMS), high-risk patients classified by the PI revealed a distinct molecular property similar to CMS4, associated with a poor prognosis. This novel PI predictor based on an eleven gene signature likely represents a surrogate diagnostic tool for identifying high-risk CRC patients and for predicting the worst responding patients for adjuvant chemotherapy. A prognostic tool that searches for eleven genes associated with colorectal cancer recurrence shows promise in initial trials. The complexity of colorectal cancer (CRC) makes it challenging to treat. A significant number of patients relapse or experience metastasis even after surgical intervention, and fail to respond to post-surgical chemotherapy. Yong Sung Kim at the Korea Research Institute of Bioscience and Biotechnology, Daejeon, and Jin Cheon Kim from the University of Ulsan, Seoul, South Korea, and co-workers, conducted RNA-sequencing analysis on samples from 130 patients with CRC, 58 of whom had suffered relapse. They pinpointed eleven genes strongly associated with recurrence-free survival. They used these to develop a prognostic tool to identify high-risk patients and those more likely to respond poorly to chemotherapy. The tool was validated on a further 795 patients with CRC.
Collapse
|
17
|
Robinson JM, Turkington S, Abey SA, Kenea N, Henderson WA. Differential gene expression and gene-set enrichment analysis in Caco-2 monolayers during a 30-day timeline with Dexamethasone exposure. Tissue Barriers 2019; 7:e1651597. [PMID: 31438773 PMCID: PMC6748367 DOI: 10.1080/21688370.2019.1651597] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glucocorticoid hormones affect gene expression via activation of glucocorticoid receptor NR3C1, causing modulation of inflammation and autoimmune activation. The glucocorticoid Dexamethasone is an important pharmaceutical for the treatment of colitis and other inflammatory bowel diseases. While suppressive effects of glucocorticoids on activated immune cells is significant, their effects upon epithelial cells are less well studied. Previous research shows that the effects of Dexamethasone treatment on polarized Caco-2 cell layer permeability is delayed for >10 treatment days (as measured by transepithelial electrical resistance). In vivo intestinal epithelial cells turn over every 3–5 days; we therefore hypothesized that culture age may produce marked effects on gene expression, potentially acting as a confounding variable. To investigate this issue, we cultured polarized Caco-2 monolayers during a 30-day timecourse with ~15 days of continuous Dexamethasone exposure. We collected samples during the timecourse and tested differential expression using a 250-plex gene expression panel and Nanostring nCounter® system. Our custom panel was selectively enriched for KEGG annotations for tight-junction, actin cytoskeleton regulation, and colorectal cancer-associated genes, allowing for focused gene ontology-based pathway enrichment analyses. To test for confounding effects of time and Dexamethasone variables, we used the Nanostring nSolver differential expression data model which includes a mixturenegative binomial modelwith optimization. We identified a time-associated “EMT-like” signature with differential expression seen in important actomyosin cytoskeleton, tight junction, integrin, and cell cycle pathway genes. Dexamethasone treatment resulted in a subtle yet significant counter-signal showing suppression of actomyosin genes and differential expression of various growth factor receptors.
Collapse
Affiliation(s)
- J M Robinson
- Digestive Disorder Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research (NINR), NIH, DHHS , Bethesda , MD , USA
| | - S Turkington
- Digestive Disorder Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research (NINR), NIH, DHHS , Bethesda , MD , USA
| | - S A Abey
- Digestive Disorder Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research (NINR), NIH, DHHS , Bethesda , MD , USA
| | - N Kenea
- Digestive Disorder Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research (NINR), NIH, DHHS , Bethesda , MD , USA
| | - W A Henderson
- Digestive Disorder Unit, Biobehavioral Branch, Division of Intramural Research, National Institute of Nursing Research (NINR), NIH, DHHS , Bethesda , MD , USA
| |
Collapse
|
18
|
Shin W, Hinojosa CD, Ingber DE, Kim HJ. Human Intestinal Morphogenesis Controlled by Transepithelial Morphogen Gradient and Flow-Dependent Physical Cues in a Microengineered Gut-on-a-Chip. iScience 2019; 15:391-406. [PMID: 31108394 PMCID: PMC6526295 DOI: 10.1016/j.isci.2019.04.037] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/28/2018] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
We leveraged a human gut-on-a-chip (Gut Chip) microdevice that enables independent control of fluid flow and mechanical deformations to explore how physical cues and morphogen gradients influence intestinal morphogenesis. Both human intestinal Caco-2 and intestinal organoid-derived primary epithelial cells formed three-dimensional (3D) villi-like microarchitecture when exposed to apical and basal fluid flow; however, 3D morphogenesis did not occur and preformed villi-like structure involuted when basal flow was ceased. When cells were cultured in static Transwells, similar morphogenesis could be induced by removing or diluting the basal medium. Computational simulations and experimental studies revealed that the establishment of a transepithelial gradient of the Wnt antagonist Dickkopf-1 and flow-induced regulation of the Frizzled-9 receptor mediate the histogenesis. Computational simulations also predicted spatial growth patterns of 3D epithelial morphology observed experimentally in the Gut Chip. A microengineered Gut Chip may be useful for studies analyzing stem cell biology and tissue development.
Collapse
Affiliation(s)
- Woojung Shin
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Christopher D Hinojosa
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Hyun Jung Kim
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
19
|
Yu Y, Seidler A, Zhou K, Yuan Z, Yeruva S, Amiri M, Yun CC, Nikolovska K, Seidler U. Expression, Localization and Functional Activity of the Major Na⁺/H⁺ Exchange Isoforms Expressed in the Intestinal Cell Line Caco-2BBe. Cell Physiol Biochem 2019; 52:1017-1038. [PMID: 30977986 PMCID: PMC7104346 DOI: 10.33594/000000070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background/Aims Enterocytes express a number of NHE isoforms with presumed localization in the apical (NHE2, 3 and 8) or basolateral (NHE1) membrane. Functional activity and localization of enterocyte NHE isoforms were assessed using fully differentiated Caco-2BBe cells, whose genetic expression profile closely resembles mature enterocytes. Methods The activity of the different NHEs was analyzed by fluorometric pHi-metry in a perfusion chamber with separate apical and basolateral perfusion, using specific inhibitors and shRNA knockdown of NHE2. The expression of the NHEs and of other relevant acid extrusion transporters was quantified by qPCR. Results Quantitative comparison of the mRNA expression levels of the different NHE isoforms in 14 day-differentiated Caco-2BBe cells showed the following order: NHE2>NHE8>NHE3>NHE1. Acid-activated NHE exchange rates in the basolateral membrane were >6-fold higher than in the apical membrane. 79 ± 3 % of the acid-activated basolateral Na+/H+ exchange rate displayed a NHE1-typical inhibitor profile, and no NHE2/3/8 typical activity could be observed. Analysis of the apical Na+/H+ exchange rates revealed that approximately 51 ± 3 % of the total apical activity displayed a NHE2/8-typical inhibitor profile and 31 ± 6 % a NHE3-typical inhibitor profile. Because no selective NHE2 inhibitor is available, a stable NHE2 knockdown cell line (C2NHE2KD) was generated. C2NHE2KD displayed a reduced NHE2-typical apical Na+/H+ exchange rate and maintained a lower steady-state pHi, despite high expression levels of other acid extruders, in particular NBCn1 (Slc4a7). Conclusion Differentiated Caco-2BBe cells display particularly high mRNA expression levels of NHE2, which can be functionally identified in the apical membrane. Although at low intracellular pH, NHE2 transport rate was far lower than that of NHE1. NHE2 activity was nevertheless essential for the maintenance of the steady-state pHi of these cells.
Collapse
Affiliation(s)
- Yan Yu
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Anna Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Kunyan Zhou
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Zhenglin Yuan
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Sunil Yeruva
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Chris C Yun
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | | | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany,
| |
Collapse
|
20
|
Nandagopal N, Santat LA, Elowitz MB. Cis-activation in the Notch signaling pathway. eLife 2019; 8:37880. [PMID: 30628888 PMCID: PMC6345567 DOI: 10.7554/elife.37880] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 01/09/2019] [Indexed: 12/31/2022] Open
Abstract
The Notch signaling pathway consists of transmembrane ligands and receptors that can interact both within the same cell (cis) and across cell boundaries (trans). Previous work has shown that cis-interactions act to inhibit productive signaling. Here, by analyzing Notch activation in single cells while controlling cell density and ligand expression level, we show that cis-ligands can also activate Notch receptors. This cis-activation process resembles trans-activation in its ligand level dependence, susceptibility to cis-inhibition, and sensitivity to Fringe modification. Cis-activation occurred for multiple ligand-receptor pairs, in diverse cell types, and affected survival in neural stem cells. Finally, mathematical modeling shows how cis-activation could potentially expand the capabilities of Notch signaling, for example enabling ‘negative’ (repressive) signaling. These results establish cis-activation as an additional mode of signaling in the Notch pathway, and should contribute to a more complete understanding of how Notch signaling functions in developmental, physiological, and biomedical contexts.
Collapse
Affiliation(s)
- Nagarajan Nandagopal
- Division of Biology and Biological Engineering, California Institute of Technology, Howard Hughes Medical Institute, Pasadena, United States
| | - Leah A Santat
- Division of Biology and Biological Engineering, California Institute of Technology, Howard Hughes Medical Institute, Pasadena, United States
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Howard Hughes Medical Institute, Pasadena, United States
| |
Collapse
|
21
|
Sivakumar A, Kurpios NA. Transcriptional regulation of cell shape during organ morphogenesis. J Cell Biol 2018; 217:2987-3005. [PMID: 30061107 PMCID: PMC6122985 DOI: 10.1083/jcb.201612115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
The emerging field of transcriptional regulation of cell shape changes aims to address the critical question of how gene expression programs produce a change in cell shape. Together with cell growth, division, and death, changes in cell shape are essential for organ morphogenesis. Whereas most studies of cell shape focus on posttranslational events involved in protein organization and distribution, cell shape changes can be genetically programmed. This review highlights the essential role of transcriptional regulation of cell shape during morphogenesis of the heart, lungs, gastrointestinal tract, and kidneys. We emphasize the evolutionary conservation of these processes across different model organisms and discuss perspectives on open questions and research avenues that may provide mechanistic insights toward understanding birth defects.
Collapse
Affiliation(s)
- Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
22
|
Gamo K, Okuzono Y, Yabuki M, Ochi T, Sugimura K, Sato Y, Sagara M, Hayashi H, Ishimura Y, Nishimoto Y, Murakawa Y, Shiokawa Z, Gotoh M, Miyazaki T, Ebisuno Y. Gene Signature-Based Approach Identified MEK1/2 as a Potential Target Associated With Relapse After Anti-TNFα Treatment for Crohn's Disease. Inflamm Bowel Dis 2018; 24:1251-1265. [PMID: 29669006 PMCID: PMC6176896 DOI: 10.1093/ibd/izy079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Anti-tumor necrosis factor alpha (anti-TNFα) therapy has become the mainstay of therapy for Crohn's disease (CD). However, post-therapy, the recurrence rate is still high. The aim of this study was to dissect the molecular mechanism for recurrence of CD treated with anti-TNFα therapy and investigate novel therapeutic options that could induce complete remission. METHODS We re-analyzed publicly available mucosal gene expression data from CD patients pre- and post-infliximab therapy to extract the transcriptional differences between responders and healthy controls. We used a systematic computational approach based on identified differences to discover novel therapies and validated this prediction through in vitro and in vivo experimentation. RESULTS We identified a set of 3545 anti-TNFα therapy-untreatable genes (TUGs) that are significantly regulated in intestinal epithelial cells, which remain altered during remission. Pathway enrichment analysis of these genes clearly showed excessive growth state and suppressed terminal differentiation, whereas immune components were clearly resolved. Through in silico screening strategy, we observed that MEK inhibitors were predicted to revert expression of genes dysregulated in infliximab responders. In vitro transcriptome analysis demonstrated that selective MEK1/2 inhibitor significantly normalized reference genes from TUGs. In addition, in vitro functional study proved that MEK1/2 inhibitor facilitated intestinal epithelial differentiation. Finally, using murine colitis model, administration of MEK1/2 inhibitor significantly improved diarrhea and histological score. CONCLUSIONS Our data revealed the abnormalities in anti-TNFα responders' CD colons that would be cause of recurrence of CD. Also, we provided evidence regarding MEK1/2 inhibitor as a potential treatment against CD to achieve sustainable remission.
Collapse
Affiliation(s)
- Kanae Gamo
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuumi Okuzono
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masato Yabuki
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takashi Ochi
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kyoko Sugimura
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yosuke Sato
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masaki Sagara
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroki Hayashi
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yoshimasa Ishimura
- Drug Safety Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yutaka Nishimoto
- Pharmaceutical Technology Research and Development Laboratories, CMC Center, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Osaka, Japan
| | - Yusuke Murakawa
- Pharmaceutical Technology Research and Development Laboratories, CMC Center, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Osaka, Japan
| | - Zenyu Shiokawa
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masayuki Gotoh
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takahiro Miyazaki
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yukihiko Ebisuno
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Japan,Address correspondence to: Yukihiko Ebisuno, PhD, Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa 251–8555, Japan ()
| |
Collapse
|
23
|
Wang Y, Jiang M, Du C, Yu Y, Liu Y, Li M, Luo F. Utilization of lung cancer cell lines for the study of lung cancer stem cells. Oncol Lett 2018; 15:6791-6798. [PMID: 29731860 PMCID: PMC5920960 DOI: 10.3892/ol.2018.8265] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/30/2017] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is one of the most lethal types of cancer, and its poor prognosis is primarily due to drug resistance and cancer recurrence. As it is associated with a low five-year survival rate, lung cancer stem cells (LCSCs) have been the subject of numerous recent studies. For these studies of LCSCs, lung cancer cell lines are more commonly used than lung cancer tissues obtained from patients, as they are easier to acquire. The methods utilized for the identification of LCSCs from lung cancer cell lines include fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), sphere-forming assay and bacterial surface display library screening. As LCSCs have certain proteins expressed on the surface (CD133, CD44 and CD24) or in the cytoplasm (ALDH and ABCG2), which may act as specific markers, the most frequently used technique to identify and obtain LCSCs is FACS. The current lack of recognized biomarkers in LCSCs makes the identification of LCSCs problematic. Furthermore, the various proportions of LCSCs in specific cell lines, as revealed by numerous previous studies, may cause the LCSC model to be questioned with regard to whether the utilization of certain lung cancer cell lines is dependable for LCSC studies. The current review focuses on lung cancer cell lines that are used for the study of LCSCs and the methods available to identify LCSCs with various markers. The present study also aimed to determine the proportion of LCSCs present in specific cell lines reported by various studies, and to discuss the suitability of specific lung cancer cell lines for the study of LCSCs.
Collapse
Affiliation(s)
- Yuyi Wang
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ming Jiang
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chi Du
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Oncology, The Second People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Yang Yu
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanyang Liu
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mei Li
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, Lung Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
24
|
The miR-196b miRNA inhibits the GATA6 intestinal transcription factor and is upregulated in colon cancer patients. Oncotarget 2018; 8:4747-4759. [PMID: 27902469 PMCID: PMC5354868 DOI: 10.18632/oncotarget.13580] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 11/11/2016] [Indexed: 12/22/2022] Open
Abstract
Objective To explore the possible misexpression of the microRNA miR-196b in colorectal cancer (CRC) and its role in controlling the expression of GATA6, a putative target gene crucial to intestinal cell homeostasis and tumorigenesis. Design The expression of miR-196b was analysed by qRT-PCR in surgical resection samples from a cohort of sporadic colon cancer patients. Manipulations of miR-196b expression were performed to demonstrate its inhibition of GATA6 protein levels. Results We found that miR-196b is significantly upregulated in pre-treatment surgical resection samples from a cohort of sporadic colon cancer patients. The upregulation of miR-196b correlates with less severe clinicopathological characteristics, such as early tumor stage and absence of lymph node metastases. We show that in CRC cells, miR-196b targets the mRNA of GATA6, a transcription factor involved in the homeostasis and differentiation of intestinal epithelial cells, and a positive regulator of the Wnt/β-catenin pathway. We moreover found that the increase of miR-196b correlates with a reduced GATA6 protein expression in colon cancer patients. Conclusion Our results establish miR-196b as a post-transcriptional inhibitor of GATA6 in CRC cells, implicating miR-196b function in gene regulatory pathways crucial to intestinal cell homeostasis and tumorigenesis. Our results furthermore suggest a role of miR-196b expression in CRC, as an antagonist of GATA6 function in tumor cells, thus providing the basis for a potential targeting strategy for the treatment of CRC.
Collapse
|
25
|
Riccio G, Bottone S, La Regina G, Badolati N, Passacantilli S, Rossi GB, Accardo A, Dentice M, Silvestri R, Novellino E, Stornaiuolo M. A Negative Allosteric Modulator of WNT Receptor Frizzled 4 Switches into an Allosteric Agonist. Biochemistry 2018; 57:839-851. [DOI: 10.1021/acs.biochem.7b01087] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Gennaro Riccio
- Department
of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Sara Bottone
- Department
of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Giuseppe La Regina
- Istituto
Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie
del Farmaco, Sapienza University of Rome, Rome, Italy
| | - Nadia Badolati
- Department
of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Sara Passacantilli
- Istituto
Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie
del Farmaco, Sapienza University of Rome, Rome, Italy
| | - Giovanni Battista Rossi
- Gastroenterology
and gastrointestinal endoscopy unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Antonella Accardo
- Department
of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Monica Dentice
- Department
of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Romano Silvestri
- Istituto
Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie
del Farmaco, Sapienza University of Rome, Rome, Italy
| | - Ettore Novellino
- Department
of Pharmacy, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
26
|
Lipopolysaccharide from Crypt-Specific Core Microbiota Modulates the Colonic Epithelial Proliferation-to-Differentiation Balance. mBio 2017; 8:mBio.01680-17. [PMID: 29042502 PMCID: PMC5646255 DOI: 10.1128/mbio.01680-17] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We identified a crypt-specific core microbiota (CSCM) dominated by strictly aerobic, nonfermentative bacteria in murine cecal and proximal colonic (PC) crypts and hypothesized that, among its possible functions, it may affect epithelial regeneration. In the present work, we isolated representative CSCM strains using selective media based upon our initial 16S rRNA-based molecular identification (i.e., Acinetobacter, Delftia, and Stenotrophomonas). Their tropism for the crypt was confirmed, and their influence on epithelial regeneration was demonstrated in vivo by monocolonization of germfree mice. We also showed that lipopolysaccharide (LPS), through its endotoxin activity, was the dominant bacterial agonist controlling proliferation. The relevant molecular mechanisms were analyzed using colonic crypt-derived organoids exposed to bacterial sonicates or highly purified LPS as agonists. We identified a Toll-like receptor 4 (TLR4)-dependent program affecting crypts at different stages of epithelial differentiation. LPS played a dual role: it repressed cell proliferation through RIPK3-mediated necroptosis of stem cells and cells of the transit-amplifying compartment and concurrently enhanced cell differentiation, particularly the goblet cell lineage. The LPS from crypt-specific core microbiota controls intestinal epithelium proliferation through necroptosis of stem cells and enhances cell differentiation, mainly the goblet cell lineage.
Collapse
|
27
|
Zhang K, Griffiths G, Repnik U, Hornef M. Seeing is understanding: Salmonella's way to penetrate the intestinal epithelium. Int J Med Microbiol 2017; 308:97-106. [PMID: 28939439 DOI: 10.1016/j.ijmm.2017.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
The molecular processes that govern host-microbial interaction illustrate not only the sophisticated and multifaceted mechanisms that protect the host from infection, but also the elaborated features of microbial pathogens that have evolved to overcome or evade the host's immune system. Here we focus on Salmonella that like other enteric pathogens must overcome the intestinal mucosal immune system, a surface constantly on alert and evolved to restrict the enteric microbiota. We discuss the initial step of Salmonella infection, the penetration of the intestinal epithelial barrier and the models used to study this fascinating aspect of microbial pathogenesis.
Collapse
Affiliation(s)
- Kaiyi Zhang
- Institute of Medical Microbiology, RWTH University Hospital Aachen, Aachen, Germany
| | | | - Urska Repnik
- Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Mathias Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
28
|
Abstract
The high success of silver nanoparticles (AgNPs), mainly associated with their proved antimicrobial properties, has led to an increasing spread in our close environment. Although many studies have been carried out to detect potential toxicity of AgNPs, most of them have been developed under unrealistic exposure conditions. In terms of human risk, the evaluation of long-term exposures to subtoxic doses of NPs remains a challenge. Here, we have determined different transformation-related end points under a scenario of 6 weeks long-term exposure to low noncytotoxic AgNPs concentrations (0.5 and 1 μg/mL) in Caco-2 cells. A significant uptake of AgNPs was demonstrated by using confocal microscopy showing a high presence of AgNPs in both the cytoplasm and the nucleus. As for the assayed parameters of cell transformation such as ability to growth without requiring adherence to a surface (soft-agar assay), the secretion of extracellular matrix metalloproteinase to the medium (zymography), migration capacity and ability of the secretome of exposed cells to promote tumor growth, significant effects were detected in all cases, with the exception of the extracellular matrix metalloproteinases (MMP2 and MMP9) secretion. Our results point out the potential carcinogenic risk associated with AgNPs exposure under long-term exposure conditions, as well as the importance of using realistic exposure scenarios to test nanomaterials.
Collapse
Affiliation(s)
- Laura Vila
- a Grup de Mutagènesi, Departament de Genètica i de Microbiologia , Facultat de Biociències, Universitat Autònoma de Barcelona , Bellaterra , Spain
| | - Ricard Marcos
- a Grup de Mutagènesi, Departament de Genètica i de Microbiologia , Facultat de Biociències, Universitat Autònoma de Barcelona , Bellaterra , Spain.,b CIBER Epidemiología y Salud Pública , ISCIII , Madrid , Spain
| | - Alba Hernández
- a Grup de Mutagènesi, Departament de Genètica i de Microbiologia , Facultat de Biociències, Universitat Autònoma de Barcelona , Bellaterra , Spain.,b CIBER Epidemiología y Salud Pública , ISCIII , Madrid , Spain
| |
Collapse
|
29
|
Kucki M, Diener L, Bohmer N, Hirsch C, Krug HF, Palermo V, Wick P. Uptake of label-free graphene oxide by Caco-2 cells is dependent on the cell differentiation status. J Nanobiotechnology 2017; 15:46. [PMID: 28637475 PMCID: PMC5480125 DOI: 10.1186/s12951-017-0280-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/13/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Understanding the interaction of graphene-related materials (GRM) with human cells is a key to the assessment of their potential risks for human health. There is a knowledge gap regarding the potential uptake of GRM by human intestinal cells after unintended ingestion. Therefore the aim of our study was to investigate the interaction of label-free graphene oxide (GO) with the intestinal cell line Caco-2 in vitro and to shed light on the influence of the cell phenotype given by the differentiation status on cellular uptake behaviour. RESULTS Internalisation of two label-free GOs with different lateral size and thickness by undifferentiated and differentiated Caco-2 cells was analysed by scanning electron microscopy and transmission electron microscopy. Semi-quantification of cells associated with GRM was performed by flow cytometry. Undifferentiated Caco-2 cells showed significant amounts of cell-associated GRM, whereas differentiated Caco-2 cells exhibited low adhesion of GO sheets. Transmission electron microscopy analysis revealed internalisation of both applied GO (small and large) by undifferentiated Caco-2 cells. Even large GO sheets with lateral dimensions up to 10 µm, were found internalised by undifferentiated cells, presumably by macropinocytosis. In contrast, no GO uptake could be found for differentiated Caco-2 cells exhibiting an enterocyte-like morphology with apical brush border. CONCLUSIONS Our results show that the internalisation of GO is highly dependent on the cell differentiation status of human intestinal cells. During differentiation Caco-2 cells undergo intense phenotypic changes which lead to a dramatic decrease in GRM internalisation. The results support the hypothesis that the cell surface topography of differentiated Caco-2 cells given by the brush border leads to low adhesion of GO sheets and sterical hindrance for material uptake. In addition, the mechanical properties of GRM, especially flexibility of the sheets, seem to be an important factor for internalisation of large GO sheets by epithelial cells. Our results highlight the importance of the choice of the in vitro model to enable better in vitro-in vivo translation.
Collapse
Affiliation(s)
- Melanie Kucki
- Laboratory for Particles-Biology Interactions, Empa-Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Liliane Diener
- Laboratory for Particles-Biology Interactions, Empa-Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Nils Bohmer
- Laboratory for Particles-Biology Interactions, Empa-Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Cordula Hirsch
- Laboratory for Particles-Biology Interactions, Empa-Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Harald F. Krug
- International Research Cooperations Manager, Empa-Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Vincenzo Palermo
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Richerche (CNR), Via P. Gobetti 101, 40129 Bologna, Italy
| | - Peter Wick
- Laboratory for Particles-Biology Interactions, Empa-Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
30
|
Xu X, Pan M, Gasiewicz AE, Li R, Kuo SM. Human and mouse microarrays-guided expression analysis of membrane protein trafficking-related genes in MDCK cells, a canine epithelial model for apical and basolateral differential protein targeting. BIOCHIMIE OPEN 2017; 4:119-126. [PMID: 29450149 PMCID: PMC5801818 DOI: 10.1016/j.biopen.2017.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/21/2017] [Indexed: 12/24/2022]
Abstract
MDCK cells are widely used to study the differential targeting of membrane transporters to apical and basolateral membrane but its canine origin limited the commercial tools available for the analysis of protein trafficking machinery. Because apical and basolateral membranes are only found in differentiated epithelial cells, genes critical for differential targeting may be specifically up-regulated upon MDCK cell differentiation. To search for these genes, a cross-species screening strategy was used. We first analyzed the human microarray data for protein trafficking-related genes that were up-regulated in colon carcinoma Caco2 cells upon differentiation. The results of mouse 44K gene expression microarray analysis were then used to extract additional candidate genes that showed higher expression in normal colon epithelium compared to primary embryonic fibroblasts. Finally, NCBI genomic sequence information was used to design RT-PCR primers for 13 candidate and 10 negative control genes and used to analyze MDCK cells at 2, 13 and 17 days after seeding. To determine whether the gene up-regulation was specific in epithelial differentiation, we also performed RT-PCR on rat non-differentiating intestinal IEC-6 cells and mouse C2C12 cells, a differentiating myoblast model. Of the 13 candidate genes, 3 genes, SDCBP2, KIF12, KIF27, met all criteria of specific up-regulation in differentiated MDCK cells. In addition, KIF13A showed up-regulation in differentiated MDCK and C2C12 cells but not in IEC-6 cells cultured for the same duration. The functions of these genes need to be analyzed in the future. This cross-species screening strategy may be useful for other non-human, non-rodent cell models.
Collapse
Affiliation(s)
- Xiaofan Xu
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Mingming Pan
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Alexis E Gasiewicz
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Rongzi Li
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Shiu-Ming Kuo
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
31
|
Aggarwal A, Schulz H, Manhardt T, Bilban M, Thakker RV, Kallay E. Expression profiling of colorectal cancer cells reveals inhibition of DNA replication licensing by extracellular calcium. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:987-996. [PMID: 28161520 PMCID: PMC5424886 DOI: 10.1016/j.bbamcr.2017.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/27/2017] [Accepted: 01/28/2017] [Indexed: 12/14/2022]
Abstract
Colorectal cancer is one of the most common cancers in industrialised societies. Epidemiological studies, animal experiments, and randomized clinical trials have shown that dietary factors can influence all stages of colorectal carcinogenesis, from initiation through promotion to progression. Calcium is one of the factors with a chemoprophylactic effect in colorectal cancer. The aim of this study was to understand the molecular mechanisms of the anti-tumorigenic effects of extracellular calcium ([Ca2+]o) in colon cancer cells. Gene expression microarray analysis of colon cancer cells treated for 1, 4, and 24 h with 2 mM [Ca2+]o identified significant changes in expression of 1571 probe sets (ANOVA, p < 10− 5). The main biological processes affected by [Ca2+]o were DNA replication, cell division, and regulation of transcription. All factors involved in DNA replication-licensing were significantly downregulated by [Ca2+]o. Furthermore, we show that the calcium-sensing receptor (CaSR), a G protein-coupled receptor is a mediator involved in this process. To test whether these results were physiologically relevant, we fed mice with a standard diet containing low (0.04%), intermediate (0.1%), or high (0.9%) levels of dietary calcium. The main molecules regulating replication licensing were inhibited also in vivo, in the colon of mice fed high calcium diet. We show that among the mechanisms behind the chemopreventive effect of [Ca2+]o is inhibition of replication licensing, a process often deregulated in neoplastic transformation. Our data suggest that dietary calcium is effective in preventing replicative stress, one of the main drivers of cancer and this process is mediated by the calcium-sensing receptor. Extracellular calcium inhibits cell division, DNA replication, in colon cancer cells. Extracellular calcium inhibits replication licensing in vitro and in vivo. The calcium-sensing receptor is a critical mediator of this process.
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Austria
| | | | - Teresa Manhardt
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Austria
| | - Martin Bilban
- Department of Medical and Chemical Laboratory Diagnostics, Medical University Vienna, Austria
| | | | - Enikö Kallay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Austria; Radcliffe Department of Medicine, University of Oxford, UK.
| |
Collapse
|
32
|
Akam EA, Tomat E. Targeting Iron in Colon Cancer via Glycoconjugation of Thiosemicarbazone Prochelators. Bioconjug Chem 2016; 27:1807-12. [PMID: 27471913 DOI: 10.1021/acs.bioconjchem.6b00332] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The implication of iron in the pathophysiology of colorectal cancer is documented at both the biochemical and epidemiological levels. Iron chelators are therefore useful molecular tools for the study and potential treatment of this type of cancer characterized by high incidence and mortality rates. We report a novel prochelation strategy that utilizes a disulfide redox switch to connect a thiosemicarbazone iron-binding unit with carbohydrate moieties targeting the increased expression of glucose transporters in colorectal cancer cells. We synthesized three glycoconjugates (GA2TC4, G6TC4, and M6TC4) with different connectivity and/or carbohydrate moieties, as well as an aglycone analog (ATC4). The sugar conjugates present increased solubility in neutral aqueous solutions, and the ester-linked conjugates M6TC4 and G6TC4 compete as effectively as d-glucose for transporter-mediated cellular uptake. The glycoconjugates show improved selectivity compared to the aglycone analog and are 6-11 times more toxic in Caco-2 colorectal adenocarcinoma cells than in normal CCD18-co colon fibroblasts.
Collapse
Affiliation(s)
- Eman A Akam
- Department of Chemistry and Biochemistry, The University of Arizona , 1306 East University Boulevard, Tucson, Arizona 85721-0041, United States
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, The University of Arizona , 1306 East University Boulevard, Tucson, Arizona 85721-0041, United States
| |
Collapse
|
33
|
Briceño MP, Nascimento LAC, Nogueira NP, Barenco PVC, Ferro EAV, Rezende-Oliveira K, Goulart LR, Alves PT, Barbosa BDF, Lima WR, Silva NM. Toxoplasma gondii Infection Promotes Epithelial Barrier Dysfunction of Caco-2 Cells. J Histochem Cytochem 2016; 64:459-69. [PMID: 27370796 DOI: 10.1369/0022155416656349] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022] Open
Abstract
After oral infection, Toxoplasma gondii invades intestinal cells, induces breakdown of intestinal physiology and barrier functions, and causes intestinal pathology in some animal species. Although parasites' invasion into host cells is a known phenomenon, the effects of T. gondii infection in the intestinal barrier are still not well established. To evaluate morphological and physiological modifications on the colorectal adenocarcinoma-derived Caco-2 cell line during T. gondii infection, microvilli, tight junction integrity, and transepithelial electrical resistance (TEER) were investigated under infection. It was observed that the dextran uptake (endocytosis) and distribution were smaller in infected than in noninfected Caco-2 cells. The infection leads to the partial loss of microvilli at the cell surface. Claudin-1, zonula occludens-1 (ZO-1), and occludin expressions were colocalized by immunofluorescence and presented discontinuous net patterns in infected cells. Immunoblotting analysis at 24 hr postinfection revealed decreasing expression of occludin and ZO-1 proteins, whereas claudin-1 presented similar expression level compared with noninfected cells. T. gondii decreased TEER in Caco-2 cells 24 hr after infection. Our results suggest that T. gondii infection may lead to the loss of integrity of intestinal mucosa, resulting in impaired barrier function.
Collapse
Affiliation(s)
- Marisol Pallete Briceño
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS)
| | | | - Nathalia Pires Nogueira
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS)
| | | | - Eloisa Amália Vieira Ferro
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS),Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (BFB, EAVF)
| | - Karine Rezende-Oliveira
- Laboratory of Biomedical Sciences, Federal University of Uberlândia, Ituiutaba, Minas Gerais, Brazil (KR-O)
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (LRG, PTA)
| | - Patrícia Terra Alves
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS),Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (LRG, PTA)
| | - Bellisa de Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (BFB, EAVF)
| | - Wânia Rezende Lima
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS),Institute of Exact and Natural Sciences, Federal University of Mato Grosso, Rondonópolis, Mato Grosso, Brazil (WRL)
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS)
| |
Collapse
|
34
|
Farkas AE, Hilgarth RS, Capaldo CT, Gerner-Smidt C, Powell DR, Vertino PM, Koval M, Parkos CA, Nusrat A. HNF4α regulates claudin-7 protein expression during intestinal epithelial differentiation. THE AMERICAN JOURNAL OF PATHOLOGY 2016. [PMID: 26216285 DOI: 10.1016/j.ajpath.2015.04.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The intestinal epithelium is a dynamic barrier that maintains the distinct environments of intestinal tissue and lumen. Epithelial barrier function is defined principally by tight junctions, which, in turn, depend on the regulated expression of claudin family proteins. Claudins are expressed differentially during intestinal epithelial cell (IEC) differentiation. However, regulatory mechanisms governing claudin expression during epithelial differentiation are incompletely understood. We investigated the molecular mechanisms regulating claudin-7 during IEC differentiation. Claudin-7 expression is increased as epithelial cells differentiate along the intestinal crypt-luminal axis. By using model IECs we observed increased claudin-7 mRNA and nascent heteronuclear RNA levels during differentiation. A screen for potential regulators of the CLDN7 gene during IEC differentiation was performed using a transcription factor/DNA binding array, CLDN7 luciferase reporters, and in silico promoter analysis. We identified hepatocyte nuclear factor 4α as a regulatory factor that bound endogenous CLDN7 promoter in differentiating IECs and stimulated CLDN7 promoter activity. These findings support a role of hepatocyte nuclear factor 4α in controlling claudin-7 expression during IEC differentiation.
Collapse
Affiliation(s)
- Attila E Farkas
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Roland S Hilgarth
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Christopher T Capaldo
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Christian Gerner-Smidt
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Doris R Powell
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Paula M Vertino
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Michael Koval
- Department of Radiation Oncology and the Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Charles A Parkos
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Pathology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
35
|
Jesus S, Soares E, Costa J, Borchard G, Borges O. Immune response elicited by an intranasally delivered HBsAg low-dose adsorbed to poly-ε-caprolactone based nanoparticles. Int J Pharm 2016; 504:59-69. [PMID: 26976502 DOI: 10.1016/j.ijpharm.2016.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/06/2016] [Accepted: 03/09/2016] [Indexed: 11/18/2022]
Abstract
Among new strategies to increase hepatitis B virus (HBV) vaccination, especially in developing countries, the development of self-administered vaccines is considered one of the most valuable. Nasal vaccination using polymeric nanoparticles (NPs) constitutes a valid approach to this issue. In detail, poly-ε-caprolactone (PCL)/chitosan NPs present advantages as a mucosal vaccine delivery system: the high resistance of PCL against degradation in biological fluids and the mucoadhesive and immunostimulatory properties of chitosan. In vitro studies revealed these NPs were retained in a mucus-secreting pulmonary epithelial cell line and were capable of entering into differentiated epithelial cells. The intranasal (IN) administration of 3 different doses of HBsAg (1.5 μg, 5 μg and 10 μg) adsorbed on a fixed amount of PCL/chitosan NPs (1614 μg) generated identical titers of serum anti-HBsAg IgG and anti-HBsAg sIgA in mice nasal secretions. Besides other factors, the NP surface characteristics, particularly, zeta potential differences among the administered formulations are believed to be implicated in the outcome of the immune response generated.
Collapse
Affiliation(s)
- Sandra Jesus
- Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Edna Soares
- Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - João Costa
- Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, quai Ernest-Ansermet, CH-1211 Geneva 4, Switzerland
| | - Olga Borges
- Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
36
|
Hepatocyte nuclear factor 1 coordinates multiple processes in a model of intestinal epithelial cell function. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:591-8. [PMID: 26855178 DOI: 10.1016/j.bbagrm.2016.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 12/26/2022]
Abstract
Mutations in hepatocyte nuclear factor 1 transcription factors (HNF1α/β) are associated with diabetes. These factors are well studied in the liver, pancreas and kidney, where they direct tissue-specific gene regulation. However, they also have an important role in the biology of many other tissues, including the intestine. We investigated the transcriptional network governed by HNF1 in an intestinal epithelial cell line (Caco2). We used chromatin immunoprecipitation followed by direct sequencing (ChIP-seq) to identify HNF1 binding sites genome-wide. Direct targets of HNF1 were validated using conventional ChIP assays and confirmed by siRNA-mediated depletion of HNF1, followed by RT-qPCR. Gene ontology process enrichment analysis of the HNF1 targets identified multiple processes with a role in intestinal epithelial cell function, including properties of the cell membrane, cellular response to hormones, and regulation of biosynthetic processes. Approximately 50% of HNF1 binding sites were also occupied by other members of the intestinal transcriptional network, including hepatocyte nuclear factor 4A (HNF4A), caudal type homeobox 2 (CDX2), and forkhead box A2 (FOXA2). Depletion of HNF1 in Caco2 cells increases FOXA2 abundance and decreases levels of CDX2, illustrating the coordinated activities of the network. These data suggest that HNF1 plays an important role in regulating intestinal epithelial cell function, both directly and through interactions with other intestinal transcription factors.
Collapse
|
37
|
Liu R, Huang S, Lei Y, Zhang T, Wang K, Liu B, Nice EC, Xiang R, Xie K, Li J, Huang C. FGF8 promotes colorectal cancer growth and metastasis by activating YAP1. Oncotarget 2015; 6:935-52. [PMID: 25473897 PMCID: PMC4359266 DOI: 10.18632/oncotarget.2822] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/25/2014] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related death worldwide. The poor prognosis of CRC is mainly due to uncontrolled tumor growth and distant metastases. In this study, we found that the level of FGF8 was elevated in the great majority of CRC cases and high FGF8 expression was significantly correlated with lymph nodes metastasis and worse overall survival. Functional studies showed that FGF8 can induce a more aggressive phenotype displaying epithelial-to-mesenchymal transition (EMT) and enhanced invasion and growth in CRC cells. Consistent with this, FGF8 can also promote tumor growth and metastasis in mouse models. Bioinformatics and pathological analysis suggested that YAP1 is a potential downstream target of FGF8 in CRC cells. Molecular validation demonstrated that FGF8 fully induced nuclear localization of YAP1 and enhanced transcriptional outcomes such as the expression of CTGF and CYR61, while decreasing YAP1 expression impeded FGF-8–induced cell growth, EMT, migration and invasion, revealing that YAP1 is required for FGF8-mediated CRC growth and metastasis. Taken together, these results demonstrate that FGF8 contributes to the proliferative and metastatic capacity of CRC cells and may represent a novel candidate for intervention in tumor growth and metastasis formation.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Shan Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, P. R. China
| | - Tao Zhang
- The School of Biomedical Sciences, Chengdu Medical College, Chengdu, P. R. China
| | - Kui Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Bo Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Rong Xiang
- School of Medicine, Nankai University, Tianjin, P.R. China
| | - Ke Xie
- Department of Oncology, Sichuan Provincial People's Hospital, Chengdu, P. R. China
| | - Jingyi Li
- The School of Biomedical Sciences, Chengdu Medical College, Chengdu, P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
38
|
Chen W, Hu Z, Li XZ, Li JL, Xu XK, Li HG, Liu Y, Liu BH, Jia WH, Li FC. CPEB4 interacts with Vimentin and involves in progressive features and poor prognosis of patients with astrocytic tumors. Tumour Biol 2015; 37:5075-87. [PMID: 26546435 DOI: 10.1007/s13277-015-3975-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 08/21/2015] [Indexed: 01/16/2023] Open
Abstract
Cytoplasmic polyadenylation element binding protein 4 (CPEB4) is a regulator of gene transcription and has been reported to be associated with biological malignancy in cancers. However, it is unclear whether CPEB4 has any clinical significance in patients with astrocytic tumors, and mechanisms that CPEB4 contribute to progression of astrocytic tumors remain largely unknown. Here, correlation between CPEB4 expression and prognosis of patients with astrocytic tumors were explored by using qPCR, WB and IHC, and X-tile, SPSS software. Cell lines U251 MG and A172 were used to study CPEB4's function and mechanisms. Co-immunoprecipitation, mass spectrometry, immunofluorescent assay, and western blot were performed to observe the interaction between CPEB4 and Vimentin. CPEB4 mRNA and protein levels were markedly elevated in 12/12 astrocytic tumors in comparison to paratumor. High expression of CPEB4 was significantly correlated with clinical progressive futures and work as an independent adverse prognostic factor for overall survival of patients with astrocytic tumors (relative risk 4.5, 95 % CI 2.1-11.2, p = 0.001). Moreover, knockdown of CPEB4 in astrocytic tumor cells inhibited their proliferation ability , clonogenicity, and invasiveness. Five candidate proteins, GRP78, Mortalin, Keratin, Vimentin, and β-actin, were identified, and the interaction between CPEB4 and Vimentin was finally confirmed. Downregulation of CPEB4 could reduce the protein expression of Vimentin. Our studies first validated that CPEB4 interacts with Vimentin and indicated that high CPEB4 expression in astrocytic tumors correlates closely with a clinically aggressive future, and that CPEB4 might represent a valuable prognostic marker for patients with astrocytic tumors.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Road of Riverside, Guangzhou, 510120, China
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China
| | - Zhen Hu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Road of Riverside, Guangzhou, 510120, China
| | - Xi-Zhao Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China
| | - Jun-Liang Li
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Road of Riverside, Guangzhou, 510120, China
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Xin-Ke Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Hai-Gang Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Road of Riverside, Guangzhou, 510120, China
| | - Yeqing Liu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Road of Riverside, Guangzhou, 510120, China
| | - Bai-Hui Liu
- Department of General Surgery, The Second Affiliated Hospital of GuangDong Pharmaceutical University, No. 167 Xingang Xi Road, Guangzhou, 510275, China
| | - Wei-Hua Jia
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China.
| | - Fang-Cheng Li
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Road of Riverside, Guangzhou, 510120, China.
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, No. 9 Jinsui Road, Guangzhou, 510623, China.
| |
Collapse
|
39
|
Halamoda-Kenzaoui B, Ceridono M, Colpo P, Valsesia A, Urbán P, Ojea-Jiménez I, Gioria S, Gilliland D, Rossi F, Kinsner-Ovaskainen A. Dispersion Behaviour of Silica Nanoparticles in Biological Media and Its Influence on Cellular Uptake. PLoS One 2015; 10:e0141593. [PMID: 26517371 PMCID: PMC4627765 DOI: 10.1371/journal.pone.0141593] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/09/2015] [Indexed: 11/19/2022] Open
Abstract
Given the increasing variety of manufactured nanomaterials, suitable, robust, standardized in vitro screening methods are needed to study the mechanisms by which they can interact with biological systems. The in vitro evaluation of interactions of nanoparticles (NPs) with living cells is challenging due to the complex behaviour of NPs, which may involve dissolution, aggregation, sedimentation and formation of a protein corona. These variable parameters have an influence on the surface properties and the stability of NPs in the biological environment and therefore also on the interaction of NPs with cells. We present here a study using 30 nm and 80 nm fluorescently-labelled silicon dioxide NPs (Rubipy-SiO2 NPs) to evaluate the NPs dispersion behaviour up to 48 hours in two different cellular media either supplemented with 10% of serum or in serum-free conditions. Size-dependent differences in dispersion behaviour were observed and the influence of the living cells on NPs stability and deposition was determined. Using flow cytometry and fluorescence microscopy techniques we studied the kinetics of the cellular uptake of Rubipy-SiO2 NPs by A549 and CaCo-2 cells and we found a correlation between the NPs characteristics in cell media and the amount of cellular uptake. Our results emphasize how relevant and important it is to evaluate and to monitor the size and agglomeration state of nanoparticles in the biological medium, in order to interpret correctly the results of the in vitro toxicological assays.
Collapse
Affiliation(s)
- Blanka Halamoda-Kenzaoui
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Mara Ceridono
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Pascal Colpo
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Andrea Valsesia
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Patricia Urbán
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Isaac Ojea-Jiménez
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Sabrina Gioria
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Douglas Gilliland
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - François Rossi
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
| | - Agnieszka Kinsner-Ovaskainen
- Nanobiosciences Unit, Institute for Health and Consumer Protection, European Commission Joint Research Centre (JRC), via E. Fermi 2749, 21027 Ispra, (VA), Italy
- * E-mail:
| |
Collapse
|
40
|
Thomas SS, Makar KW, Li L, Zheng Y, Yang P, Levy L, Rudolph RY, Lampe PD, Yan M, Markowitz SD, Bigler J, Lampe JW, Potter JD. Tissue-specific patterns of gene expression in the epithelium and stroma of normal colon in healthy individuals in an aspirin intervention trial. BMC MEDICAL GENETICS 2015; 16:18. [PMID: 25927723 PMCID: PMC4422425 DOI: 10.1186/s12881-015-0161-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/26/2015] [Indexed: 12/21/2022]
Abstract
Background Regular aspirin use reduces colon adenoma and carcinoma incidence. UDP-glucuronosyltransferases (UGT) are involved in aspirin metabolism and clearance, and variant alleles in UGT1A6 have been shown to alter salicylic acid metabolism and risk of colon neoplasia. Methods In a randomized, cross-over, placebo-controlled trial of 44 healthy men and women, homozygous for UGT1A6*1 or UGT1A6*2, we explored differences between global epithelial and stromal expression, using Affymetrix U133 + 2.0 microarrays and tested effects of 60-day aspirin supplementation (325 mg/d) on epithelial and stromal gene expression and colon prostaglandin E2 (PGE2) levels. Results No statistically significant differences in gene expression were observed in response to aspirin or UGT1A6 genotype, but tissue PGE2 levels were lower with aspirin compared to placebo (p <0.001). Transcripts differentially expressed between epithelium and stroma (N = 4916, P <0.01, false discovery rate <0.001), included a high proportion of genes involved in cell signaling, cellular movement, and cancer. Genes preferentially expressed in epithelium were involved in drug and xenobiotic metabolism, fatty acid and lipid metabolism, apoptosis signaling, and ion transport. Genes preferentially expressed in stroma included those involved in inflammation, cellular adhesion, and extracellular matrix production. Wnt-Tcf4 pathway genes were expressed in both epithelium and stroma but differed by subcellular location. Conclusions These results suggest that, in healthy individuals, subtle effects of aspirin on gene expression in normal colon tissue are likely overwhelmed by inter-individual variability in microarray analyses. Differential expression of critical genes between colonic epithelium and stroma suggest that these tissue types need to be considered separately. Electronic supplementary material The online version of this article (doi:10.1186/s12881-015-0161-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sushma S Thomas
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Karen W Makar
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Lin Li
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Yingye Zheng
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Peiying Yang
- M.D. Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Lisa Levy
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | | | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Min Yan
- Case Western Reserve University School of Medicine, Cincinnati, OH, 44106, USA.
| | - Sanford D Markowitz
- Case Western Reserve University School of Medicine, Cincinnati, OH, 44106, USA.
| | | | - Johanna W Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - John D Potter
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|
41
|
Tan G, Li RH, Li C, Wu F, Zhao XM, Ma JY, Lei S, Zhang WD, Zhi FC. Down-regulation of human enteric antimicrobial peptides by NOD2 during differentiation of the paneth cell lineage. Sci Rep 2015; 5:8383. [PMID: 25670499 PMCID: PMC4323657 DOI: 10.1038/srep08383] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/19/2015] [Indexed: 01/07/2023] Open
Abstract
Ileal Crohn's disease (CD) arising from the alteration of intestinal homeostasis is characterized by two features, namely a decrease in Paneth cell-produced antimicrobial peptides that play a key role in maintaining this balance and an increase in NOD2, an intracellular sensor. Although mutations in NOD2 are highly correlated with the incidence of CD, the physiological role of NOD2 in intestinal immunity remains elusive. Here, we show that NOD2 can down-regulate the expression of human enteric antimicrobial peptides during differentiation of the Paneth cell lineage. This finding, which links the decrease of human enteric antimicrobial peptides to increased NOD2 in ileal CD patients, provides a new view into the pathogenesis of ileal CD.
Collapse
Affiliation(s)
- Gao Tan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Run-hua Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chen Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fang Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin-mei Zhao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-yi Ma
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shan Lei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wen-di Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fa-chao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
42
|
Karagiannis GS, Musrap N, Saraon P, Treacy A, Schaeffer DF, Kirsch R, Riddell RH, Diamandis EP. Bone morphogenetic protein antagonist gremlin-1 regulates colon cancer progression. Biol Chem 2015; 396:163-83. [DOI: 10.1515/hsz-2014-0221] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/01/2014] [Indexed: 11/15/2022]
Abstract
Abstract
Bone morphogenetic proteins (BMP) are phylogenetically conserved signaling molecules of the transforming growth factor-beta (TGF-beta) superfamily of proteins, involved in developmental and (patho)physiological processes, including cancer. BMP signaling has been regarded as tumor-suppressive in colorectal cancer (CRC) by reducing cancer cell proliferation and invasion, and by impairing epithelial-to-mesenchymal transition (EMT). Here, we mined existing proteomic repositories to explore the expression of BMPs in CRC. We found that the BMP antagonist gremlin-1 (GREM1) is secreted from heterotypic tumor-host cell interactions. We then sought to investigate whether GREM1 is contextually and mechanistically associated with EMT in CRC. Using immunohistochemistry, we showed that GREM1-expressing stromal cells harbor prominent features of myofibroblasts (i.e., cancer-associated fibroblasts), such as expression of α-smooth muscle actin and laminin-beta-1, and were in contextual proximity to invasion fronts with loss of the tight junction protein occludin and parallel nuclear accumulation of β-catenin, two prominent EMT hallmarks. Furthermore, in vitro assays demonstrated that GREM1-dependent suppression of BMP signaling results in EMT induction, characterized by cadherin switching (loss of E-cadherin-upregulation of N-cadherin) and overexpression of Snail. Collectively, our data support that GREM1 promotes the loss of cancer cell differentiation at the cancer invasion front, a mechanism that may facilitate tumor progression.
Collapse
|
43
|
Lin M, Wang Z, Han X. B Cells with Regulatory Function in Animal Models of Autoimmune and Non-Autoimmune Diseases. ACTA ACUST UNITED AC 2015; 5:9-17. [PMID: 26236565 PMCID: PMC4517676 DOI: 10.4236/oji.2015.51002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although the identification of B cell subsets with negative regulatory functions and the definition of their mechanisms of action are recent events, the important negative regulatory roles of B cells in immune responses are now broadly recognized. There is an emerging appreciation for the pivotal role played by B cells in several areas of human diseases including autoimmune diseases and non-autoimmune diseases such as parasite infections and cancer. The recent research advancement of regulatory B cells in human disease coincides with the vastly accelerated pace of research on the bridging of innate and adaptive immune system. Current study and our continued research may provide better understanding of the mechanisms that promote regulatory B10 cell function to counteract exaggerated immune activation in autoimmune as well as non-autoimmune conditions. This review is focused on the current knowledge of BREG functions studied in animal models of autoimmune and non-autoimmune diseases.
Collapse
Affiliation(s)
- Mei Lin
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, USA ; Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, USA
| |
Collapse
|
44
|
Tan G, Zeng B, Zhi FC. Regulation of human enteric α-defensins by NOD2 in the Paneth cell lineage. Eur J Cell Biol 2014; 94:60-6. [PMID: 25433720 DOI: 10.1016/j.ejcb.2014.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/25/2014] [Accepted: 10/31/2014] [Indexed: 12/19/2022] Open
Abstract
Human enteric α-defensins (HD5 and HD6), major antimicrobial peptides produced by Paneth cells in the intestine, play important roles in intestinal innate immunity. Since their expression is decreased in Crohn's disease (CD), with decreased expression being more pronounced in the presence of NOD2 mutations, it would be extremely interesting to investigate the mechanism by which NOD2 may regulate expression of human enteric α-defensins. Here we show that although NOD2 by itself can slightly up-regulate expression of enteric α-defensins mainly through activation of the NF-κB pathway, it can strongly down-regulates their expression during differentiation of the Paneth cell lineage mainly by inhibiting activation of the MAPK pathway. Since NOD2 is over-expressed in CD and mutant NOD2 cannot result in NF-κB activity, our finding can provide an explanation of the previous observation showing decreased expression of human enteric α-defensin in CD and even more so in the presence of NOD2 mutations. In addition, this finding provides a new view on the function of NOD2 in regulating intestinal innate immunity.
Collapse
Affiliation(s)
- Gao Tan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Zeng
- Department of Gastroenterology, the First Affiliated Hospital of South China University, Hengyang 421001, China
| | - Fa-Chao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
45
|
Klaus C, Schneider U, Hedberg C, Schütz AK, Bernhagen J, Waldmann H, Gassler N, Kaemmerer E. Modulating effects of acyl-CoA synthetase 5-derived mitochondrial Wnt2B palmitoylation on intestinal Wnt activity. World J Gastroenterol 2014; 20:14855-14864. [PMID: 25356045 PMCID: PMC4209548 DOI: 10.3748/wjg.v20.i40.14855] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/10/2014] [Accepted: 06/23/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of acyl-CoA synthetase 5 (ACSL5) activity in Wnt signaling in intestinal surface epithelia.
METHODS: Several cell lines were used to investigate the ACSL5-dependent expression and synthesis of Wnt2B, a mitochondrially expressed protein of the Wnt signaling family. Wnt activity was functionally assessed with a luciferase reporter assay. ACSL5-related biochemical Wnt2B modifications were investigated with a modified acyl-exchange assay. The findings from the cell culture models were verified using an Apcmin/+ mouse model as well as normal and neoplastic diseased human intestinal tissues.
RESULTS: In the presence of ACSL5, Wnt2B was unable to translocate into the nucleus and was enriched in mitochondria, which was paralleled by a significant decrease in Wnt activity. ACSL5-dependent S-palmitoylation of Wnt2B was identified as a molecular reason for mitochondrial Wnt2B accumulation. In cell culture systems, a strong relation of ACSL5 expression, Wnt2B palmitoylation, and degree of malignancy were found. Using normal mucosa, the association of ACSL5 and Wnt2B was seen, but in intestinal neoplasias the mechanism was only rudimentarily observed.
CONCLUSION: ACSL5 mediates antiproliferative activities via Wnt2B palmitoylation with diminished Wnt activity. The molecular pathway is probably relevant for intestinal homeostasis, overwhelmed by other pathways in carcinogenesis.
Collapse
|
46
|
Yilmaz-Ozcan S, Sade A, Kucukkaraduman B, Kaygusuz Y, Senses KM, Banerjee S, Gure AO. Epigenetic mechanisms underlying the dynamic expression of cancer-testis genes, PAGE2, -2B and SPANX-B, during mesenchymal-to-epithelial transition. PLoS One 2014; 9:e107905. [PMID: 25229454 PMCID: PMC4168264 DOI: 10.1371/journal.pone.0107905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 08/22/2014] [Indexed: 12/14/2022] Open
Abstract
Cancer-testis (CT) genes are expressed in various cancers but not in normal tissues other than in cells of the germline. Although DNA demethylation of promoter-proximal CpGs of CT genes is linked to their expression in cancer, the mechanisms leading to demethylation are unknown. To elucidate such mechanisms we chose to study the Caco-2 colorectal cancer cell line during the course of its spontaneous differentiation in vitro, as we found CT genes, in particular PAGE2, -2B and SPANX-B, to be up-regulated during this process. Differentiation of these cells resulted in a mesenchymal-to-epithelial transition (MET) as evidenced by the gain of epithelial markers CDX2, Claudin-4 and E-cadherin, and a concomitant loss of mesenchymal markers Vimentin, Fibronectin-1 and Transgelin. PAGE2 and SPAN-X up-regulation was accompanied by an increase in Ten-eleven translocation-2 (TET2) expression and cytosine 5-hydroxymethylation as well as the disassociation of heterochromatin protein 1 and the polycomb repressive complex 2 protein EZH2 from promoter-proximal regions of these genes. Reversal of differentiation resulted in down-regulation of PAGE2, -2B and SPANX-B, and induction of epithelial-to-mesenchymal transition (EMT) markers, demonstrating the dynamic nature of CT gene regulation in this model.
Collapse
Affiliation(s)
- Sinem Yilmaz-Ozcan
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Asli Sade
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Baris Kucukkaraduman
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Yasemin Kaygusuz
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Kerem Mert Senses
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Ali Osmay Gure
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- * E-mail:
| |
Collapse
|
47
|
Klaus C, Kaemmerer E, Reinartz A, Schneider U, Plum P, Jeon MK, Hose J, Hartmann F, Schnölzer M, Wagner N, Kopitz J, Gassler N. TP53 status regulates ACSL5-induced expression of mitochondrial mortalin in enterocytes and colorectal adenocarcinomas. Cell Tissue Res 2014; 357:267-78. [PMID: 24770931 DOI: 10.1007/s00441-014-1826-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/23/2014] [Indexed: 12/25/2022]
Abstract
Acyl-CoA synthetase 5 (ACSL5), a mitochondrially localized enzyme, catalyzes the synthesis of long-chain fatty acid thioesters and is physiologically involved in pro-apoptotic sensing of enterocytes. The aim of the present study is to identify an ACSL5-dependent regulation of mitochondrially expressed proteins and the characterization of related pathways in normal and diseased human intestinal mucosa. Proteomics of isolated mitochondria from ACSL5 transfectants and CaCo2 controls were performed. ACSL5-dependent protein synthesis was verified with quantitative reverse transcription plus the polymerase chain reaction, Western blotting, short-interfering-RNA-mediated gene silencing and additional cell culture experiments. Lipid changes were analyzed with tandem mass spectrometry. ACSL5-related pathways were characterized in normal mucosa and sporadic adenocarcinomas of the human intestine. In CaCo2 cells transfected with ACSL5, mortalin (HSPA9) was about two-fold increased in mitochondria, whereas cytoplasmic mortalin levels were unchanged. Disturbance of acyl-CoA/sphingolipid metabolism, induced by ACSL5 over-expression, was characterized as crucial. ACSL5-related over-expression of mitochondrial mortalin was found in HEK293 and Lovo (wild-type TP53 [tumor protein p53]) and CaCo2 (p53-negative; TP53 mutated) cells but not in Colo320DM cells (mutated TP53). In normal human intestinal mucosa, an increasing gradient of both ACSL5 and mortalin from bottom to top was observed, whereas p53 (wild-type TP53) decreased. In sporadic intestinal adenocarcinomas with strong p53 immunostaining (mutated TP53), ACSL5-related mortalin expression was heterogeneous. ACSL5-induced mitochondrial mortalin expression is assumed to be a stress response to ACSL5-related changes in lipid metabolism and is regulated by the TP53 status. Uncoupling of ACSL5 and mitochondrial mortalin by mutated TP53 could be important in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Christina Klaus
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Karagiannis GS, Treacy A, Messenger D, Grin A, Kirsch R, Riddell RH, Diamandis EP. Expression patterns of bone morphogenetic protein antagonists in colorectal cancer desmoplastic invasion fronts. Mol Oncol 2014; 8:1240-52. [PMID: 24812030 DOI: 10.1016/j.molonc.2014.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 02/24/2014] [Accepted: 04/09/2014] [Indexed: 01/31/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are a group of growth factors with dual functions in cancer development and progression. Recently, certain tumor-promoting roles have been identified for selected antagonists/inhibitors (BMPIs) of this developmental pathway. A recent focus on the implication of BMP in colorectal cancer progression has emerged, mainly due to the presence of inactivating mutations in several members of the canonical signaling cascade. However, the detailed expression profiles of BMPIs remain largely unknown. Based on our previous work, whereby three specific BMPIs, gremlin-1 (GREM1), high-temperature requirement A3 (HTRA3) and follistatin (FST) were collectively overexpressed in desmoplastic cocultures of colorectal cancer (CRC), here, we undertook an immunohistochemical approach to describe the patterns of their expression in CRC patients. Two major characteristics described the BMPI expression signature: First, the synchronous and coordinated stromal and epithelial overexpression of individual BMPIs in desmoplastic lesions, which demonstrated that all three of them contribute to increasing levels of BMP antagonism in such areas. Second, the presence of microenvironmental polarity in the BMPI pattern of expression, which was indicated through the preferential expression of HTRA3 in the stromal, and the parallel FST/GREM1 expression in the epithelial component of the investigated sections. In addition, expression of HTRA3 in the epithelial compartment of the tumors demonstrated a significant predictive power to discriminate between tumor-budding-bearing and tumor-budding-free desmoplastic microenvironments. Together, these findings contribute to the understanding of signaling dynamics of BMP antagonism in the colorectal cancer desmoplastic invasion front.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ann Treacy
- MC Pathology, The Laboratory, Charlemont Clinic, Charlemont Mall, Dublin, Ireland
| | - David Messenger
- Zane Cohen Clinical Research Centre, Mount Sinai Hospital, Toronto, Canada; Division of General Surgery, Mount Sinai Hospital, Toronto, Canada
| | - Andrea Grin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, St Michael's Hospital, Toronto, Canada
| | - Richard Kirsch
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Robert H Riddell
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Natoli M, Christensen J, El-Gebali S, Felsani A, Anderle P. The role of CDX2 in Caco-2 cell differentiation. Eur J Pharm Biopharm 2014; 85:20-5. [PMID: 23958315 DOI: 10.1016/j.ejpb.2013.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/20/2013] [Accepted: 03/22/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND CDX2 plays a key part in the differentiation of Caco-2 cells, a colon carcinoma derived cell line that undergoes spontaneous differentiation. The effect of CDX2 expression in Caco-2 cells over time in culture has not been studied yet on a genome-wide level. METHODS The impact of CDX2 expression on the genomic profile of Caco-2 cells was studied by transducing cells with CDX2 targeting shRNAs. Knockdown efficiency was assessed on mRNA level and protein level by RTPCR, microarrays, and Western blots. Gene set enrichment analysis was performed to assess regulation of specific gene sets. RESULTS CDX2 expression had an inhibitory effect on the transcriptional activity of β-catenin/TCF at early stages of culturing, while at later stages, its role in the trans-activation of target genes specific for small intestinal enterocytes seemed more dominant. CONCLUSIONS The unique induction of a small intestinal signature upon differentiation in Caco-2 cells seems to be at least partially under the control of CDX2.
Collapse
Affiliation(s)
- Manuela Natoli
- Istituto di Biologia Cellulare e Neurobiologia, CNR, Rome, Italy
| | | | | | | | | |
Collapse
|
50
|
Fischer A, Gluth M, Weege F, Pape UF, Wiedenmann B, Baumgart DC, Theuring F. Glucocorticoids regulate barrier function and claudin expression in intestinal epithelial cells via MKP-1. Am J Physiol Gastrointest Liver Physiol 2014; 306:G218-28. [PMID: 24309183 DOI: 10.1152/ajpgi.00095.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Barrier dysfunction is pivotal to the pathogenesis of inflammatory bowel diseases (IBD) and collagenous colitis. Glucocorticoids restore barrier function in Crohn's disease, but whether this reflects attenuated inflammation or an epithelial-specific action has not yet been addressed. Using filter-grown Caco-2 monolayers as an in vitro model of the intestinal epithelial barrier, we observed that glucocorticoids induced a time- and dose-dependent increase in transepithelial electrical resistance (TEER) in a glucocorticoid receptor-dependent manner without altering flux of larger solutes or changing principal tight junction architecture. This was accompanied by reduced paracellular cation flux, reduced expression of the pore-forming tight junction component claudin-2, and upregulation of the sealing tight junction protein claudin-4. In contrast, expression of occludin, claudin-1, -7, or -8 was not altered. Dexamethasone increased expression and activity of MAPK phosphatase-1 and inhibition of this phosphatase prevented the glucocorticoid-induced changes in TEER and claudin expression, whereas inhibiting p38 or MEK1/2 was not sufficient to replicate the glucocorticoid effects. Upon exposure to IFN-γ, TNF-α, or IL-1β, TEERs declined in dexamethasone-treated cells but remained consistently higher than in cells not receiving glucocorticoids. Treatment with IFN/TNF resulted in an upregulation of claudin-2 that was significantly attenuated by dexamethasone, whereas increased claudin-2 expression upon IL-1β stimulation was not affected by glucocorticoids. Taken together, barrier augmentation might represent a previously unrecognized mechanism of action, potentially contributing to the therapeutic efficacy of glucocorticoids in IBD and collagenous colitis.
Collapse
Affiliation(s)
- Andreas Fischer
- Department of Medicine, Division of Gastroenterology and Hepatology Campus Virchow Klinikum and
| | | | | | | | | | | | | |
Collapse
|