1
|
Li H, Wang K, Hao M, Liu Y, Liang X, Yuan D, Ding L. Intestinal epithelial Cldn-7 regulates intestinal inflammation by altering the gut microbiota. Pathol Res Pract 2024; 260:155448. [PMID: 39004000 DOI: 10.1016/j.prp.2024.155448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/05/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND AND AIM Tight junctions maintain gut homeostasis by forming a physical barrier that protects the gut from invasion by microbiota. Cldn-7 is an important component involved in this protection, but the relationship between Cldn-7, intestinal inflammation, and gut microbiota has not been clarified. Here, we hypothesize that Cldn-7 depletion affects intestinal inflammation by altering the gut microbiota. METHODS Based on the induced intestinal condition of Cldn-7 knockout mice (Cldn7fl/fl;villin-CreaERT2), we established the intestinal flora depletion model and colitis model by antibiotic drinking and feeding with dextran sodium sulfate (DSS). The environment of Cldn-7 gene deletion mice was changed by co-housing experiment. AB-PAS staining and Muc2 were used to detect the effect of co-housing and Cldn-7 deficiency on the mucus layer after flora depletion. qRT-PCR was used to detect the expression of intestinal inflammatory factors and AMPs in mice. Feces were collected and proportions of microbiota were analyzed by 16 S rRNA amplicon sequencing. RESULTS Mice in the co-housing experiment had altered intestinal microbiota, including diversity, composition, and functional prediction, compared to controls. Intestinal inflammation was restored to some extent following altered intestinal microbiota. The intestinal inflammation caused by Cldn-7 deficiency and susceptibility to DSS could be reduced after antibiotic administration compared to controls, in terms of phenotype, pathological changes, inflammatory factors, mucus barrier, and expression of AMPs. CONCLUSIONS In analyses of intestinal tissues, colitis induction, and gut microbiota in mice with intestinal disruption of Cldn-7, we found this protein to prevent intestinal inflammation by regulating the gut microbiota. Cldn-7might therefore be an important mediator of host-microbiome interactions. Our research has revealed that Cldn-7 plays an indispensable role in maintaining intestinal homeostasis by regulating the gut microbiota and impacting intestinal inflammation. These findings provide new insights into the pathogenesis of ulcerative colitis.
Collapse
Affiliation(s)
- Huimin Li
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Kun Wang
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Mengdi Hao
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Yin Liu
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiaoqing Liang
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Dajin Yuan
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Lei Ding
- Department of Gastrointestinal Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
2
|
Jacobsen F, Pushpadevan R, Viehweger F, Freytag M, Schlichter R, Gorbokon N, Büscheck F, Luebke AM, Putri D, Kluth M, Hube-Magg C, Hinsch A, Höflmayer D, Fraune C, Bernreuther C, Lebok P, Sauter G, Minner S, Steurer S, Simon R, Burandt E, Dum D, Lutz F, Marx AH, Krech T, Clauditz TS. Cadherin-17 (CDH17) expression in human cancer: A tissue microarray study on 18,131 tumors. Pathol Res Pract 2024; 256:155175. [PMID: 38452580 DOI: 10.1016/j.prp.2024.155175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 03/09/2024]
Abstract
Cadherin-17 (CDH17) is a membranous cell adhesion protein predominantly expressed in intestinal epithelial cells. CDH17 is therefore considered a possible diagnostic and therapeutic target. This study was to comprehensively determine the expression of CDH17 in cancer and to further assess the diagnostic utility of CDH17 immunohistochemistry (IHC). A tissue microarray containing 14,948 interpretable samples from 150 different tumor types and subtypes as well as 76 different normal tissue types was analyzed by IHC. In normal tissues, a membranous CDH17 staining was predominantly seen in the epithelium of the intestine and pancreatic excretory ducts. In tumors, 53 of 150 analyzed categories showed CDH17 positivity including 26 categories with at least one strongly positive case. CDH17 positivity was most common in epithelial and neuroendocrine colorectal neoplasms (50.0%-100%), other gastrointestinal adenocarcinomas (42.7%-61.6%), mucinous ovarian cancer (61.1%), pancreatic acinar cell carcinoma (28.6%), cervical adenocarcinoma (52.6%), bilio-pancreatic adenocarcinomas (40.5-69.8%), and other neuroendocrine neoplasms (5.6%-100%). OnIy 9.9% of 182 pulmonary adenocarcinomas were CDH17 positive. In colorectal adenocarcinomas, reduced CDH17 staining was linked to high pT (p = 0.0147), nodal metastasis (p = 0.0041), V1 (p = 0.0025), L1 (p = 0.0054), location in the right colon (p = 0.0033), and microsatellite instability (p < 0.0001). The CDH17 expression level was unrelated to tumor phenotype in gastric and pancreatic cancer. In summary, our comprehensive overview on CDH17 expression in human tumors identified various tumor entities that might often benefit from anti-CDH17 therapies and suggest utility of CDH17 IHC for the distinction of metastatic gastrointestinal or bilio-pancreatic adenocarcinomas (often positive) from primary pulmonary adenocarcinomas (mostly negative).
Collapse
Affiliation(s)
- Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramesh Pushpadevan
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Morton Freytag
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ria Schlichter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Devita Putri
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Lutz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Varani J, McClintock SD, Nadeem DM, Harber I, Zeidan D, Aslam MN. A multi-mineral intervention to counter pro-inflammatory activity and to improve the barrier in human colon organoids. Front Cell Dev Biol 2023; 11:1132905. [PMID: 37476158 PMCID: PMC10354648 DOI: 10.3389/fcell.2023.1132905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Introduction: Ulcerative colitis is a chronic inflammatory condition, and continuous inflammatory stimulus may lead to barrier dysfunction. The goal of this study was to assess barrier proteomic expression by a red algae-derived multi-mineral intervention in the absence or presence of pro-inflammatory insult. Methods: Human colon organoids were maintained in a control culture medium alone or exposed to lipopolysaccharide with a combination of three pro-inflammatory cytokines [tumor necrosis factor-α, interleukin-1β and interferon-γ (LPS-cytokines)] to mimic the environment in the inflamed colon. Untreated organoids and those exposed to LPS-cytokines were concomitantly treated for 14 days with a multi-mineral product (Aquamin®) that has previously been shown to improve barrier structure/function. The colon organoids were subjected to proteomic analysis to obtain a broad view of the protein changes induced by the two interventions alone and in combination. In parallel, confocal fluorescence microscopy, tissue cohesion and transepithelial electrical resistance (TEER) measurements were used to assess barrier structure/function. Results: The LPS-cytokine mix altered the expression of multiple proteins that influence innate immunity and promote inflammation. Several of these were significantly decreased with Aquamin® alone but only a modest decrease in a subset of these proteins was detected by Aquamin® in the presence of LPS-cytokines. Among these, a subset of inflammation-related proteins including fibrinogen-β and -γ chains (FGB and FGG), phospholipase A2 (PLA2G2A) and SPARC was significantly downregulated in the presence of Aquamin® (alone and in combination with LPS-cytokines); another subset of proteins with anti-inflammatory, antioxidant or anti-microbial activity was upregulated by Aquamin® treatment. When provided alone, Aquamin® strongly upregulated proteins that contribute to barrier formation and tissue strength. Concomitant treatment with LPS-cytokines did not inhibit barrier formation in response to Aquamin®. Confocal microscopy also displayed increased expression of desmoglein-2 (DSG2) and cadherin-17 (CDH17) with Aquamin®, either alone or in the presence of the pro-inflammatory stimulus. Increased cohesion and TEER with Aquamin® (alone or in the presence of LPS-cytokines) indicates improved barrier function. Conclusion: Taken together, these findings suggest that multi-mineral intervention (Aquamin®) may provide a novel approach to combating inflammation in the colon by improving barrier structure/function as well as by directly altering the expression of certain pro-inflammatory proteins.
Collapse
Affiliation(s)
| | | | | | | | | | - Muhammad N. Aslam
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
4
|
Li N, Xu S, Zhang S, Zhu Q, Meng X, An W, Fu B, Zhong M, Yang Y, Lin Z, Liu X, Xia J, Wang J, You T, Yan C, Tang H, Zhuang G, Peng Z. MSI2 deficiency in ILC3s attenuates DSS-induced colitis by affecting the intestinal microbiota. Front Immunol 2023; 13:963379. [PMID: 36713428 PMCID: PMC9877450 DOI: 10.3389/fimmu.2022.963379] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Background The etiology and pathogenesis of inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), are generally believed to be related to immune dysfunction and intestinal microbiota disorder. However, the exact mechanism is not yet fully understood. The pathological changes associated with dextran sodium sulfate (DSS)-induced colitis are similar to those in human UC. As a subgroup of the innate immune system, group 3 innate lymphoid cells (ILC3s) are widely distributed in the lamina propria of the intestinal mucosa, and their function can be regulated by a variety of molecules. Musashi2 (MSI2) is a type of evolutionarily conserved RNA-binding protein that maintains the function of various tissue stem cells and is essential for postintestinal epithelial regeneration. The effect of MSI2 deficiency in ILC3s on IBD has not been reported. Thus, mice with conditional MSI2 knockout in ILC3s were used to construct a DSS-induced colitis model and explore its effects on the pathogenesis of IBD and the species, quantity and function of the intestinal microbiota. Methods Msi2flox/flox mice (Msi2fl/fl ) and Msi2flox/floxRorcCre mice (Msi2ΔRorc ) were induced by DSS to establish the IBD model. The severity of colitis was evaluated by five measurements: body weight percentage, disease activity index, colon shortening degree, histopathological score and routine blood examination. The species, quantity and function of the intestinal microbiota were characterized by high-throughput 16S rRNA gene sequencing of DNA extracted from fecal samples. Results MSI2 was knocked out in the ILC3s of Msi2ΔRorc mice. The Msi2ΔRorc mice exhibited reductions in body weight loss, the disease activity index, degree of colon shortening, tissue histopathological score and immune cells in the peripheral blood compared to those of Msi2fl/fl mice after DSS administration. The 16S rRNA sequencing results showed that the diversity of the intestinal microbiota in DSS-treated Msi2ΔRorc mice changed, with the abundance of Firmicutes increasing and that of Bacteroidetes decreasing. The linear discriminant analysis effect size (LEfSe) approach revealed that Lactobacillaceae could be the key bacteria in the Msi2ΔRorc mouse during the improvement of colitis. Using PICRUST2 to predict the function of the intestinal microbiota, it was found that the functions of differential bacteria inferred by modeling were mainly enriched in infectious diseases, immune system and metabolic functions. Conclusions MSI2 deficiency in ILC3s attenuated DSS-induced colonic inflammation in mice and affected intestinal microbiota diversity, composition, and function, with Lactobacillaceae belonging to the phylum Firmicutes possibly representing the key bacteria. This finding could contribute to our understanding of the pathogenesis of IBD and provide new insights for its clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Nengneng Li
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shiquan Xu
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuaishuai Zhang
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qiang Zhu
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaole Meng
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenbin An
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Baoqing Fu
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Department of Laboratory Medicine, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China
| | - Mengya Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yan Yang
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zeyang Lin
- Department of Pathology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Xueni Liu
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junjie Xia
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jie Wang
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tingting You
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Changxiu Yan
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Huamei Tang
- Department of Pathology, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China,*Correspondence: Zhihai Peng, ; Guohong Zhuang, ; Huamei Tang,
| | - Guohong Zhuang
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,*Correspondence: Zhihai Peng, ; Guohong Zhuang, ; Huamei Tang,
| | - Zhihai Peng
- Department of Organ Transplantation, Xiang’an Hospital, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Organ Transplantation Clinical Medical Center of Xiamen University, Xiamen, Fujian, China,*Correspondence: Zhihai Peng, ; Guohong Zhuang, ; Huamei Tang,
| |
Collapse
|
5
|
Tournier B, Aucagne R, Truntzer C, Fournier C, Ghiringhelli F, Chapusot C, Martin L, Bouvier AM, Manfredi S, Jooste V, Callanan MB, Lepage C. Integrative Clinical and DNA Methylation Analyses in a Population-Based Cohort Identifies CDH17 and LRP2 as Risk Recurrence Factors in Stage II Colon Cancer. Cancers (Basel) 2022; 15:cancers15010158. [PMID: 36612154 PMCID: PMC9817957 DOI: 10.3390/cancers15010158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Stage II colon cancer (CC), although diagnosed early, accounts for 16% of CC deaths. Predictors of recurrence risk could mitigate this but are currently lacking. By using a DNA methylation-based clinical screening in real-world (n = 383) and in TCGA-derived cohorts of stage II CC (n = 134), we have devised a novel 40 CpG site-based classifier that can segregate stage II CC into four previously undescribed disease sub-classes that are characterised by distinct molecular features, including activation of MYC/E2F-dependant proliferation signatures. By multivariate analyses, hypermethylation of 2 CpG sites at genes CDH17 and LRP2, respectively, was found to independently confer either significantly increased (CDH17; p-value, 0.0203) or reduced (LRP2; p-value, 0.0047) risk of CC recurrence. Functional enrichment and immune cell infiltration analyses, on RNAseq data from the TCGA cohort, revealed cases with hypermethylation at CDH17 to be enriched for KRAS, epithelial-mesenchymal transition and inflammatory functions (via IL2/STAT5), associated with infiltration by 'exhausted' T cells. By contrast, LRP2 hypermethylated cases showed enrichment for mTORC1, DNA repair pathways and activated B cell signatures. These findings will be of value for improving personalised care paths and treatment in stage II CC patients.
Collapse
Affiliation(s)
- Benjamin Tournier
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Pathology, Dijon University Hospital, 21000 Dijon, France
| | - Romain Aucagne
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
| | - Caroline Truntzer
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Centre Georges-François Leclerc (CGFL), 21000 Dijon, France
| | - Cyril Fournier
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
| | - François Ghiringhelli
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
- Centre Georges-François Leclerc (CGFL), 21000 Dijon, France
| | - Caroline Chapusot
- Department of Pathology, Dijon University Hospital, 21000 Dijon, France
| | - Laurent Martin
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Pathology, Dijon University Hospital, 21000 Dijon, France
| | - Anne Marie Bouvier
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
| | - Sylvain Manfredi
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, 21000 Dijon, France
| | - Valérie Jooste
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, 21000 Dijon, France
| | - Mary B. Callanan
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
- Correspondence: (M.B.C.); (C.L.)
| | - Côme Lepage
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, 21000 Dijon, France
- Correspondence: (M.B.C.); (C.L.)
| |
Collapse
|
6
|
Orlova E, Dudding T, Chernus JM, Alotaibi RN, Haworth S, Crout RJ, Lee MK, Mukhopadhyay N, Feingold E, Levy SM, McNeil DW, Foxman B, Weyant RJ, Timpson NJ, Marazita ML, Shaffer JR. Association of Early Childhood Caries with Bitter Taste Receptors: A Meta-Analysis of Genome-Wide Association Studies and Transcriptome-Wide Association Study. Genes (Basel) 2022; 14:59. [PMID: 36672800 PMCID: PMC9858612 DOI: 10.3390/genes14010059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022] Open
Abstract
Although genetics affects early childhood caries (ECC) risk, few studies have focused on finding its specific genetic determinants. Here, we performed genome-wide association studies (GWAS) in five cohorts of children (aged up to 5 years, total N = 2974, cohorts: Center for Oral Health Research in Appalachia cohorts one and two [COHRA1, COHRA2], Iowa Fluoride Study, Iowa Head Start, Avon Longitudinal Study of Parents and Children [ALSPAC]) aiming to identify genes with potential roles in ECC biology. We meta-analyzed the GWASs testing ~3.9 million genetic variants and found suggestive evidence for association at genetic regions previously associated with caries in primary and permanent dentition, including the β-defensin anti-microbial proteins. We then integrated the meta-analysis results with gene expression data in a transcriptome-wide association study (TWAS). This approach identified four genes whose genetically predicted expression was associated with ECC (p-values < 3.09 × 10−6; CDH17, TAS2R43, SMIM10L1, TAS2R14). Some of the strongest associations were with genes encoding members of the bitter taste receptor family (TAS2R); other members of this family have previously been associated with caries. Of note, we identified the receptor encoded by TAS2R14, which stimulates innate immunity and anti-microbial defense in response to molecules released by the cariogenic bacteria, Streptococcus mutans and Staphylococcus aureus. These findings provide insight into ECC genetic architecture, underscore the importance of host-microbial interaction in caries risk, and identify novel risk genes.
Collapse
Affiliation(s)
- Ekaterina Orlova
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Tom Dudding
- Bristol Dental School, University of Bristol, Bristol BS1 2LY, UK
- Medical Research Council Integrative Epidemiology Unit, Department of Population Health Sciences, University of Bristol, Bristol BS8 1QU, UK
| | - Jonathan M. Chernus
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Rasha N. Alotaibi
- Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Simon Haworth
- Bristol Dental School, University of Bristol, Bristol BS1 2LY, UK
- Medical Research Council Integrative Epidemiology Unit, Department of Population Health Sciences, University of Bristol, Bristol BS8 1QU, UK
| | - Richard J. Crout
- Department of Periodontics, School of Dentistry, West Virginia University, Morgantown, WV 26505, USA
| | - Myoung Keun Lee
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nandita Mukhopadhyay
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Eleanor Feingold
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Steven M. Levy
- Department of Preventive & Community Dentistry, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Daniel W. McNeil
- Department of Psychology & Department of Dental Public Health and Professional Practice, West Virginia University, Morgantown, WV 26505, USA
| | - Betsy Foxman
- Center for Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robert J. Weyant
- Dental Public Health, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nicholas J. Timpson
- Medical Research Council Integrative Epidemiology Unit, Department of Population Health Sciences, University of Bristol, Bristol BS8 1QU, UK
- Avon Longitudinal Study of Parents and Children, University of Bristol, Bristol BS8 1QU, UK
| | - Mary L. Marazita
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John R. Shaffer
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
7
|
Chemically Induced Colitis-Associated Cancer Models in Rodents for Pharmacological Modulation: A Systematic Review. J Clin Med 2022; 11:jcm11102739. [PMID: 35628865 PMCID: PMC9146029 DOI: 10.3390/jcm11102739] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
Animal models for colitis-associated colorectal cancer (CACC) represent an important tool to explore the mechanistic basis of cancer-related inflammation, providing important evidence that several inflammatory mediators play specific roles in the initiation and perpetuation of colitis and CACC. Although several original articles have been published describing the CACC model in rodents, there is no consensus about the induction method. This review aims to identify, summarize, compare, and discuss the chemical methods for the induction of CACC through the PRISMA methodology. METHODS We searched MEDLINE via the Pubmed platform for studies published through March 2021, using a highly sensitive search expression. The inclusion criteria were only original articles, articles where a chemically-induced animal model of CACC is described, preclinical studies in vivo with rodents, and articles published in English. RESULTS Chemically inducible models typically begin with the administration of a carcinogenic compound (as azoxymethane (AOM) or 1,2-dimethylhydrazine (DMH)), and inflammation is caused by repeated cycles of colitis-inducing agents (such as 2,4,6-trinitrobenzenesulfonic acid (TNBS) or dextran sulfate sodium (DSS)). The strains mostly used are C57BL/6 and Balb/c with 5-6 weeks. To characterize the preclinical model, the parameters more used include body weight, stool consistency and morbidity, inflammatory biomarkers such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, angiogenesis markers such as proliferating cell nuclear antigen (PCNA), marker of proliferation Ki-67, and caspase 3, the presence of ulcers, thickness or hyperemia in the colon, and histological evaluation of inflammation. CONCLUSION The AOM administration seems to be important to the CACC induction method, since the carcinogenic effect is achieved with just one administration. DSS has been the more used inflammatory agent; however, the TNBS contribution should be more studied, since it allows a reliable, robust, and a highly reproducible animal model of intestinal inflammation.
Collapse
|
8
|
Ding Y, Wang K, Xu C, Hao M, Li H, Ding L. Intestinal Claudin-7 deficiency impacts the intestinal microbiota in mice with colitis. BMC Gastroenterol 2022; 22:24. [PMID: 35039003 PMCID: PMC8762895 DOI: 10.1186/s12876-022-02100-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Background Intestinal epithelial cells form a physical barrier that protects the intestine against the intestinal microbiota through tight junctions (TJs) and adhesive junctions, while barrier disruption may lead to inflammatory bowel disease (IBD). Claudin-7 (Cldn7) has been implicated in this protection as an important member of TJs. Here, we experimentally study the effect of Cldn7 deletion on intestinal microbiota in colitis. Methods Colitis model was established based on inducible intestinal conditional Cldn7 gene knockout mice (Cldn7fl/fl; villin-CreERT2), by feeding with dextran sodium sulfate (DSS). AB-PAS staining and immunohistochemical staining of Muc2 mucin were used to detect the effect of Cldn7 deficiency on the mucus layer of mice with colitis, and fluorescence in situ hybridization was used to detect how Cldn7 promotes spatial separation of the gut microbiota from the host. The microbiota population was characterized by high-throughput 16S rRNA gene sequencing of DNA extracted from fecal samples. Results Compared with the controls, Cldn7 knockout increased susceptibility to colitis, including greater degree of weight loss, colon shortening, and a significantly higher disease activity index score. DSS-treated Cldn7 knockout mice promoted the migration of bacteria to the intestinal epithelium to some extent by damaging the intestinal mucus layer. Sequencing of 16S rRNA showed that DSS-treated Cldn7 knockout mice reduced the gut microbiota diversity and had greater relative abundance of Escherichia coli. LEfSe analysis indicated that Escherichia coli may be the key bacteria in Cldn7 knockout mice during DSS-induced colitis. Furthermore, the Tax4Fun analysis predicted that DSS-treated Cldn7 knockout mice enriched for microbiota impacting infectious diseases, immune system and metabolic functions. Conclusions Our data suggests an association between intestinal Cldn7 knockout and microbiota dysbiosis during inflammatory events.
Collapse
Affiliation(s)
- Yuhan Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, China
| | - Kun Wang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, China
| | - Chang Xu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, China.,Department of Hepato-Pancreato-Biliary Surgery, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Mengdi Hao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, China
| | - Huimin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, China
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, China.
| |
Collapse
|
9
|
Tanca A, Abbondio M, Fiorito G, Pira G, Sau R, Manca A, Muroni MR, Porcu A, Scanu AM, Cossu-Rocca P, De Miglio MR, Uzzau S. Metaproteomic Profile of the Colonic Luminal Microbiota From Patients With Colon Cancer. Front Microbiol 2022; 13:869523. [PMID: 35495697 PMCID: PMC9048685 DOI: 10.3389/fmicb.2022.869523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Recent studies have provided evidence of interactions among the gut microbiota (GM), local host immune cells, and intestinal tissues in colon carcinogenesis. However, little is known regarding the functions exerted by the GM in colon cancer (CC), particularly with respect to tumor clinical classification and lymphocyte infiltration. In addition, stool, usually employed as a proxy of the GM, cannot fully represent the original complexity of CC microenvironment. Here, we present a pilot study aimed at characterizing the metaproteome of CC-associated colonic luminal contents and identifying its possible associations with CC clinicopathological features. Colonic luminal contents were collected from 24 CC tissue specimens immediately after surgery. Samples were analyzed by shotgun metaproteomics. Almost 30,000 microbial peptides were quantified in the samples, enabling the achievement of the taxonomic and functional profile of the tumor-associated colonic luminal metaproteome. Upon sample aggregation based on tumor stage, grade, or tumor-infiltrating lymphocytes (TILs), peptide sets enabling discrimination of sample groups were identified through discriminant analysis (DA). As a result, Bifidobacterium and Bacteroides fragilis were significantly enriched in high-stage and high-grade CC, respectively. Among metabolic functions, formate-tetrahydrofolate ligase was significantly associated with high-stage CC. Finally, based on the results of this pilot study, we assessed the optimal sample size for differential metaproteomic studies analyzing colonic luminal contents. In conclusion, we provide a detailed picture of the microbial and host components of the colonic luminal proteome and propose promising associations between GM taxonomic/functional features and CC clinicopathological features. Future studies will be needed to verify the prognostic value of these data and to fully exploit the potential of metaproteomics in enhancing our knowledge concerning CC progression.
Collapse
Affiliation(s)
- Alessandro Tanca
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Marcello Abbondio
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giovanni Fiorito
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,Medical Research Council (MRC), Centre for Environment and Health, Imperial College London, London, United Kingdom
| | - Giovanna Pira
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Rosangela Sau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandra Manca
- Department of Pathology, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy
| | - Maria Rosaria Muroni
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alberto Porcu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Antonio Mario Scanu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Paolo Cossu-Rocca
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy.,Surgical Pathology Unit, Department of Diagnostic Services, "Giovanni Paolo II" Hospital, Area Socio-Sanitaria Locale (ASSL) Olbia-Azienda per la Tutela della Salute (ATS) Sardegna, Olbia, Italy
| | - Maria Rosaria De Miglio
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Sergio Uzzau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
10
|
Wang K, Ding Y, Xu C, Hao M, Li H, Ding L. Cldn-7 deficiency promotes experimental colitis and associated carcinogenesis by regulating intestinal epithelial integrity. Oncoimmunology 2021; 10:1923910. [PMID: 34026335 PMCID: PMC8118418 DOI: 10.1080/2162402x.2021.1923910] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Intestinal epithelial barrier protects intestine from infection and injury, while chronic inflammation is a trigger for tumorigenesis. As a member of tight junctions (TJs) family, Claudin-7 (Cldn-7) is dedicated to maintaining cell polarity and TJs barrier integrity, and closely related to the development of inflammation and tumors. However, potential roles of Cldn-7 in intestinal inflammation and colitis-associated colorectal cancer (CAC) have not been well characterized in vivo. Here, we analyzed the expression profile of Cldn-7 in inflammatory bowel disease (IBD) and CAC. Colitis and colitis-cancer transformation models were established based on inducible intestinal conditional Cldn-7 gene knockout mice (Cldn7fl/fl;villin-CreERT2), by intraperitoneal injection of azomethane (AOM) and dextran sodium sulfate (DSS) feeding. Cldn-7 knockout promoted susceptibility to colitis and CAC, aggravated clinical symptoms, severely damaged intestinal epithelium, increased mucosal inflammation accompanied dysregulated cell proliferation-apoptosis. Epithelial barrier integrity was destroyed, and intercellular permeability was increased. After AOM/DSS induction, tumor burden and volume were increased, characterized by enhanced proliferation and activation of Wnt/β-catenin signaling pathway. Mechanistically, Cldn-7 deficiency promoted colitis and subsequently malignant transformation by destroying TJs integrity and increasing inflammatory cascade. Overall, based on Cldn-7 knockout mouse model, we have first demonstrated the key roles of Cldn-7 in maintaining intestinal homeostasis and preventing IBD and consequent CAC. Abbreviations: AJs: adherens junctions; AOM: azomethane; Cldn-7: Claudin-7; CRC: colorectal cancer; CAC: colitis-associated colorectal cancer; CD: Crohn's disease; DSS: dextran sodium sulfate; DAI: disease activity index; EMT: epithelial-mesenchymal transition; FITC: fluorescence isothiocyanate; HB: hemoglobin; IBD: inflammatory bowel disease; IECs: intestinal epithelial cells; ISCs: intestinal stem cells; PLT: platelet; RBC: red blood cell; ROS: reactive oxygen species; TAM: tamoxifen; TJs: tight junctions; TCF/LEF: T-cell factor/lymphoid enhancer factor; UC: ulcerative colitis; WBC: white blood cell.
Collapse
Affiliation(s)
- Kun Wang
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Yuhan Ding
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Chang Xu
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Hepato-Pancreato-Biliary Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Mengdi Hao
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Huimin Li
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Lei Ding
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|