1
|
Kang LI, Sarullo K, Marsh JN, Lu L, Khonde P, Ma C, Haritunians T, Mujukian A, Mengesha E, McGovern DPB, Stappenbeck TS, Swamidass SJ, Liu TC. Development of a deep learning algorithm for Paneth cell density quantification for inflammatory bowel disease. EBioMedicine 2024; 110:105440. [PMID: 39536395 PMCID: PMC11605460 DOI: 10.1016/j.ebiom.2024.105440] [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: 07/08/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Alterations in ileal Paneth cell (PC) density have been described in gut inflammatory diseases such as Crohn's disease (CD) and could be used as a biomarker for disease prognosis. However, quantifying PCs is time-intensive, a barrier for clinical workflow. Deep learning (DL) has transformed the development of robust and accurate tools for complex image evaluation. Our aim was to use DL to quantify PCs for use as a quantitative biomarker. METHODS A retrospective cohort of whole slide images (WSI) of ileal tissue samples from patients with/without inflammatory bowel disease (IBD) was used for the study. A pathologist-annotated training set of WSI were used to train a U-net two-stage DL model to quantify PC number, crypt number, and PC density. For validation, a cohort of 48 WSIs were manually quantified by study pathologists and compared to the DL algorithm, using root mean square error (RMSE) and the coefficient of determination (r2) as metrics. To test the value of PC quantification as a biomarker, resection specimens from patients with CD (n = 142) and without IBD (n = 48) patients were analysed with the DL model. Finally, we compared time to disease recurrence in patients with CD with low versus high DL-quantified PC density using Log-rank test. FINDINGS Initial one-stage DL model showed moderate accuracy in predicting PC density in cross-validation tests (RMSE = 1.880, r2 = 0.641), but adding a second stage significantly improved accuracy (RMSE = 0.802, r2 = 0.748). In the validation of the two-stage model compared to expert pathologists, the algorithm showed good performance up to RMSE = 1.148, r2 = 0.708. The retrospective cross-sectional cohort had mean ages of 62.1 years in the patients without IBD and 38.6 years for the patients with CD. In the non-IBD cohort, 43.75% of the patients were male, compared to 49.3% of the patients with CD. Analysis by the DL model showed significantly higher PC density in non-IBD controls compared to the patients with CD (4.04 versus 2.99 PC/crypt). Finally, the algorithm quantification of PCs density in patients with CD showed patients with the lowest 25% PC density (Quartile 1) have significantly shorter recurrence-free interval (p = 0.0399). INTERPRETATION The current model performance demonstrates the feasibility of developing a DL-based tool to measure PC density as a predictive biomarker for future clinical practice. FUNDING This study was funded by the National Institutes of Health (NIH).
Collapse
Affiliation(s)
- Liang-I Kang
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States
| | - Kathryn Sarullo
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States
| | - Jon N Marsh
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States
| | - Liang Lu
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States
| | - Pooja Khonde
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States
| | - Changqing Ma
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States
| | - Talin Haritunians
- The F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, United States
| | - Angela Mujukian
- The F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, United States
| | - Emebet Mengesha
- The F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, United States
| | - Dermot P B McGovern
- The F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, United States
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Mail Code NE30, 9500 Euclid Avenue, Cleveland, OH, 44195, United States
| | - S Joshua Swamidass
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States.
| | - Ta-Chiang Liu
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO, 63110, United States.
| |
Collapse
|
2
|
Hung CT, Ma C, Panda SK, Trsan T, Hodel M, Frein J, Foster A, Sun S, Wu HT, Kern J, Mishra R, Jain U, Ho YC, Colonna M, Stappenbeck TS, Liu TC. Western diet reduces small intestinal intraepithelial lymphocytes via FXR-Interferon pathway. Mucosal Immunol 2024; 17:1019-1028. [PMID: 38992433 DOI: 10.1016/j.mucimm.2024.07.001] [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: 05/02/2023] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
The prevalence of obesity in the United States has continued to increase over the past several decades. Understanding how diet-induced obesity modulates mucosal immunity is of clinical relevance. We previously showed that consumption of a high fat, high sugar "Western" diet (WD) reduces the density and function of small intestinal Paneth cells, a small intestinal epithelial cell type with innate immune function. We hypothesized that obesity could also result in repressed gut adaptive immunity. Using small intestinal intraepithelial lymphocytes (IEL) as a readout, we found that in non-inflammatory bowel disease (IBD) subjects, high body mass index correlated with reduced IEL density. We recapitulated this in wild type (WT) mice fed with WD. A 4-week WD consumption was able to reduce IEL but not splenic, blood, or bone marrow lymphocytes, and the effect was reversible after another 2 weeks of standard diet (SD) washout. Importantly, WD-associated IEL reduction was not dependent on the presence of gut microbiota, as WD-fed germ-free mice also showed IEL reduction. We further found that WD-mediated Farnesoid X Receptor (FXR) activation in the gut triggered IEL reduction, and this was partially mediated by intestinal phagocytes. Activated FXR signaling stimulated phagocytes to secrete type I IFN, and inhibition of either FXR or type I IFN signaling within the phagocytes prevented WD-mediated IEL loss. Therefore, WD consumption represses both innate and adaptive immunity in the gut. These findings have significant clinical implications in the understanding of how diet modulates mucosal immunity.
Collapse
Affiliation(s)
- Chen-Ting Hung
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Changqing Ma
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Santosh K Panda
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Miki Hodel
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Jennifer Frein
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Amanda Foster
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Shengxiang Sun
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Hung-Ting Wu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Justin Kern
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Richa Mishra
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Umang Jain
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Ya-Chi Ho
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, United States.
| |
Collapse
|
3
|
Zhang J, Zhang R, Chen Y, Guo X, Ren Y, Wang M, Li X, Huang Z, Zhu W, Yu K. Indole-3-aldehyde Alleviates High-Fat Diet-Induced Gut Barrier Disruption by Increasing Intestinal Stem Cell Expansion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18930-18941. [PMID: 39146439 DOI: 10.1021/acs.jafc.4c02381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
High-fat diet (HFD) feeding is known to cause intestinal barrier disruption, thereby triggering severe intestinal inflammatory disease. Indole-3-aldehyde (IAld) has emerged as a potential candidate for mitigating inflammatory responses and maintaining intestinal homeostasis. However, the role of IAld in the HFD-related intestinal disruption remains unclear. In this study, 48 7 week-old male C57BL/6J mice were assigned to four groups: the normal chow diet (NCD) group received a NCD; the HFD group was fed an HFD; the HFD + IAld200 group was supplemented with 200 mg/kg IAld in the HFD; and the HFD + IAld600 group was supplemented with 600 mg/kg IAld in the HFD. The results showed that dietary IAld supplementation ameliorated fat accumulation and metabolic disorders, which are associated with reduced intestinal permeability. This reduction potentially led to decreased systemic inflammation and enhanced intestinal barrier function in HFD-fed mice. Furthermore, we found that IAld promoted intestinal stem cell (ISC) proliferation by activating aryl hydrocarbon receptors (AHRs) in vivo and ex vivo. These findings suggest that IAld restores the HFD-induced intestinal barrier disruption by promoting AHR-mediated ISC proliferation.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruofan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
- Animal Health Inspection Institute of Suzhou, Wujiang District, Suzhou 215200, China
| | - Yahui Chen
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xin Guo
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuting Ren
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Mengting Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Zan Huang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
4
|
Ragab M, Schlichting H, Hicken M, Mester P, Hirose M, Almeida LN, Christiansen L, Ibrahim S, Tews HC, Divanovic S, Sina C, Derer S. Azathioprine promotes intestinal epithelial cell differentiation into Paneth cells and alleviates ileal Crohn's disease severity. Sci Rep 2024; 14:12879. [PMID: 38839896 PMCID: PMC11153537 DOI: 10.1038/s41598-024-63730-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/30/2024] [Indexed: 06/07/2024] Open
Abstract
Paneth cells (PCs), a subset of intestinal epithelial cells (IECs) found at the base of small intestinal crypts, play an essential role in maintaining intestinal homeostasis. Altered PCs function is associated with diverse intestinal pathologies, including ileal Crohn's disease (CD). CD patients with ileal involvement have been previously demonstrated to display impairment in PCs and decreased levels of anti-microbial peptides. Although the immunosuppressive drug Azathioprine (AZA) is widely used in CD therapy, the impact of AZA on IEC differentiation remains largely elusive. In the present study, we hypothesized that the orally administered drug AZA also exerts its effect through modulation of the intestinal epithelium and specifically via modulation of PC function. AZA-treated CD patients exhibited an ileal upregulation of AMPs on both mRNA and protein levels compared to non-AZA treated patients. Upon in vitro AZA stimulation, intestinal epithelial cell line MODE-K exhibited heightened expression levels of PC marker in concert with diminished cell proliferation but boosted mitochondrial OXPHOS activity. Moreover, differentiation of IECs, including PCs differentiation, was boosted in AZA-treated murine small intestinal organoids and was associated with decreased D-glucose consumption and decreased growth rates. Of note, AZA treatment strongly decreased Lgr5 mRNA expression as well as Ki67 positive cells. Further, AZA restored dysregulated PCs associated with mitochondrial dysfunction. AZA-dependent inhibition of IEC proliferation is accompanied by boosted mitochondria function and IEC differentiation into PC.
Collapse
Affiliation(s)
- Mohab Ragab
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Heidi Schlichting
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Maren Hicken
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Patricia Mester
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital, Regensburg, Germany
| | - Misa Hirose
- Lübeck Institute of Experimental Dermatology and Center for Research On Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Larissa N Almeida
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Lea Christiansen
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Saleh Ibrahim
- Lübeck Institute of Experimental Dermatology and Center for Research On Inflammation of the Skin, University of Lübeck, Lübeck, Germany
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Hauke Christian Tews
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital, Regensburg, Germany
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christian Sina
- Institute of Nutritional Medicine and 1st Department of Medicine, Division of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Stefanie Derer
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
| |
Collapse
|
5
|
Ventrello SW, McMurry NR, Edwards NM, Bain LJ. Chronic arsenic exposure affects stromal cells and signaling in the small intestine in a sex-specific manner. Toxicol Sci 2024; 198:303-315. [PMID: 38310360 PMCID: PMC10964740 DOI: 10.1093/toxsci/kfae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2024] Open
Abstract
Arsenic is a toxicant that is ingested through drinking water and food, exposing nearly 140 million people to levels above the 10 ppb guideline concentration. Studies have shown that arsenic affects intestinal stem cells (ISCs), but the mechanisms by which arsenic alters the formation of adult cells in the small intestine are not well understood. Signals derived from intestinal stromal cells initiate and maintain differentiation. The goal of this study is to evaluate arsenic's effect on intestinal stromal cells, including PdgfrαLo trophocytes, located proximal to the ISCs, and PdgfrαHi telocytes, located proximal to the transit-amplifying region and up the villi. Adult Sox9tm2Crm-EGFP mice were exposed to 0, 33, and 100 ppb sodium arsenite in their drinking water for 13 weeks, and sections of duodenum were examined. Flow cytometry indicated that arsenic exposure dose-responsively reduced Sox9+ epithelial cells and trended toward increased Pdgfrα+ cells. The trophocyte marker, CD81, was reduced by 10-fold and 9.0-fold in the 100 ppb exposure group in male and female mice, respectively. Additionally, a significant 2.2- to 3.1-fold increase in PdgfrαLo expression was found in male mice in trophocytes and Igfbp5+ cells. PdgfrαHi protein expression, a telocyte marker, was more prevalent along the villus/crypt structure in females, whereas Gli1 expression (telocytes) was reduced in male mice exposed to arsenic. Principle coordinate analysis confirmed the sex-dependent response to arsenic exposure, with an increase in trophocyte and decrease in telocyte marker expression observed in male mice. These results imply that arsenic alters intestinal mesenchymal cells in a sex-dependent manner.
Collapse
Affiliation(s)
- Scott W Ventrello
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas R McMurry
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas M Edwards
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| |
Collapse
|
6
|
Bautista GM, Cera AJ, Schoenauer RJ, Persiani M, Lakshminrusimha S, Chandrasekharan P, Gugino SF, Underwood MA, McElroy SJ. Paneth cell ontogeny in term and preterm ovine models. Front Vet Sci 2024; 11:1275293. [PMID: 38318150 PMCID: PMC10839032 DOI: 10.3389/fvets.2024.1275293] [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: 08/09/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Paneth cells are critically important to intestinal health, including protecting intestinal stem cells, shaping the intestinal microbiome, and regulating host immunity. Understanding Paneth cell biology in the immature intestine is often modeled in rodents with little information in larger mammals such as sheep. Previous studies have only established the distribution pattern of Paneth cells in healthy adult sheep. Our study aimed to examine the ontogeny, quantification, and localization of Paneth cells in fetal and newborn lambs at different gestational ages and with perinatal transient asphyxia. We hypothesized that ovine Paneth cell distribution at birth resembles the pattern seen in humans (highest concentrations in the ileum) and that ovine Paneth cell density is gestation-dependent. Methods Intestinal samples were obtained from 126-127 (preterm, with and without perinatal transient asphyxia) and 140-141 (term) days gestation sheep. Samples were quantified per crypt in at least 100 crypts per animal and confirmed as Paneth cells through in immunohistochemistry. Results Paneth cells had significantly higher density in the ileum compared to the jejunum and were absent in the colon. Discussion Exposure to perinatal transient asphyxia acutely decreased Paneth cell numbers. These novel data support the possibility of utilizing ovine models for understanding Paneth cell biology in the fetus and neonate.
Collapse
Affiliation(s)
- Geoanna M. Bautista
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | - Anjali J. Cera
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | - Rebecca J. Schoenauer
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Michele Persiani
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | - Satyan Lakshminrusimha
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | | | - Sylvia F Gugino
- Department of Pediatrics, University of Buffalo, Buffalo, NY, United States
| | - Mark A. Underwood
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | - Steven J. McElroy
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
7
|
Tao Q, Zhang J, Liang Q, Song S, Wang S, Yao X, Gao Q, Wang L. Puerarin alleviates sleep disorders in aged mice related to repairing intestinal mucosal barrier. NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:29. [PMID: 37698689 PMCID: PMC10497485 DOI: 10.1007/s13659-023-00390-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
More and more evidence suggests that puerarin, a potential remedy for gut inflammation, may have an ameliorative effect on sleep disturbances. However, the relationship between puerarin and sleep disruption has not been extensively researched. This study aims to explore the role and mechanisms of puerarin in improving sleep disorders. We established a light-induced sleep disorder model in mice and assessed the effects of puerarin on cognitive behavior using open field and water maze tests. Pathological detection demonstrated that sleep disturbances resulted in observable damage to the liver, lung, and kidney. Puerarin reversed multi-organ damage and inflammation. Further, puerarin activated paneth cells, resulting in increased lysozyme and TGF-β production, and stimulating intestinal stem cell proliferation. Puerarin also effectively inhibited the expression of F4/80, iNOS, TNF-α, and IL-1β in the small intestine, while it increased Chil3, CD206, and Arg-1 levels. Moreover, puerarin treatment significantly decreased P-P65, TLR4, Bcl-xl, and cleaved caspase-3 protein levels while increasing barrier protein levels, including ZO-1, Occludin, Claudin 1 and E-cadherin suggesting a reduction in inflammation and apoptosis in the gut. Overall, puerarin diminished systemic inflammation, particularly intestinal inflammation, and enhanced intestinal barrier integrity in mice with sleep disorders. Our findings suggest a potential new therapeutic pathway for sleep disorders.
Collapse
Affiliation(s)
- Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Jinhua Zhang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Shuxia Wang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Xiaoming Yao
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China.
| | - Lei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China.
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
8
|
Salm F, Znalesniak EB, Laskou A, Harder S, Schlüter H, Hoffmann W. Expression Profiling along the Murine Intestine: Different Mucosal Protection Systems and Alterations in Tff1-Deficient Animals. Int J Mol Sci 2023; 24:12684. [PMID: 37628863 PMCID: PMC10454331 DOI: 10.3390/ijms241612684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Tff1 is a typical gastric peptide secreted together with the mucin, Muc5ac. Tff1-deficient (Tff1KO) mice are well known for their prominent gastric phenotype and represent a recognized model for antral tumorigenesis. Notably, intestinal abnormalities have also been reported in the past in these animals. Here, we have compared the expression of selected genes in Tff1KO mice and their corresponding wild-type littermates (RT-PCR analyses), focusing on different mucosal protection systems along the murine intestine. As hallmarks, genes were identified with maximum expression in the proximal colon and/or the duodenum: Agr2, Muc6/A4gnt/Tff2, Tff1, Fut2, Gkn2, Gkn3, Duox2/Lpo, Nox1. This is indicative of different protection systems such as Tff2/Muc6, Tff1-Fcgbp, gastrokines, fucosylation, and reactive oxygen species (ROS) in the proximal colon and/or duodenum. Few significant transcriptional changes were observed in the intestine of Tff1KO mice when compared with wild-type littermates, Clca1 (Gob5), Gkn1, Gkn2, Nox1, Tff2. We also analyzed the expression of Tff1, Tff2, and Tff3 in the pancreas, liver, and lung of Tff1KO and wild-type animals, indicating a cross-regulation of Tff gene expression. Furthermore, on the protein level, heteromeric Tff1-Fcgbp and various monomeric Tff1 forms were identified in the duodenum and a high-molecular-mass Tff2/Muc6 complex was identified in the proximal colon (FPLC, proteomics).
Collapse
Affiliation(s)
- Franz Salm
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Eva B. Znalesniak
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Aikaterini Laskou
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Sönke Harder
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
9
|
Barreto E Barreto L, Rattes IC, da Costa AV, Gama P. Paneth cells and their multiple functions. Cell Biol Int 2022; 46:701-710. [PMID: 35032139 DOI: 10.1002/cbin.11764] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 01/08/2023]
Abstract
The small intestine mucosa is lined by specialized cells that form the crypt-villus axis, which expands its surface. Among the six intestinal epithelial cell types, the Paneth cell is located at the base of the crypt, and it contains numerous granules in its cytoplasm, composed of antimicrobial peptides, such as defensins and lysozyme, and growth factors, such as EGF, TGF-alpha, and Wnt ligands. Together, these elements act in the defense against microorganisms, regulation of intestinal microbiota, maintenance, and regulation of stem cell identity. Pathologies that target Paneth cells can disturb such defense activity, but they also affect the maintenance of stem cell niche. In that way, Crohn's disease, necrotizing enterocolitis, and graft-versus-host disease promote a reduction of Paneth cell population, and consequently of secretion of their products into the lumen of the crypts, making the affected organism predisposed to infections and dysbiosis. Additionally, the emergence of new intestinal cells is also decreased. This review aims to address the main characteristics of Paneth cells, highlighting their multiple functions and the importance of their preservation to ensure bowel homeostasis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laylla Barreto E Barreto
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Isadora Campos Rattes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Aline Vasques da Costa
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Patrícia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
10
|
Nickerson KP, Llanos-Chea A, Ingano L, Serena G, Miranda-Ribera A, Perlman M, Lima R, Sztein MB, Fasano A, Senger S, Faherty CS. A Versatile Human Intestinal Organoid-Derived Epithelial Monolayer Model for the Study of Enteric Pathogens. Microbiol Spectr 2021; 9:e0000321. [PMID: 34106568 PMCID: PMC8552518 DOI: 10.1128/spectrum.00003-21] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/09/2023] Open
Abstract
Gastrointestinal infections cause significant morbidity and mortality worldwide. The complexity of human biology and limited insights into host-specific infection mechanisms are key barriers to current therapeutic development. Here, we demonstrate that two-dimensional epithelial monolayers derived from human intestinal organoids, combined with in vivo-like bacterial culturing conditions, provide significant advancements for the study of enteropathogens. Monolayers from the terminal ileum, cecum, and ascending colon recapitulated the composition of the gastrointestinal epithelium, in which several techniques were used to detect the presence of enterocytes, mucus-producing goblet cells, and other cell types following differentiation. Importantly, the addition of receptor activator of nuclear factor kappa-B ligand (RANKL) increased the presence of M cells, critical antigen-sampling cells often exploited by enteric pathogens. For infections, bacteria were grown under in vivo-like conditions known to induce virulence. Overall, interesting patterns of tissue tropism and clinical manifestations were observed. Shigella flexneri adhered efficiently to the cecum and colon; however, invasion in the colon was best following RANKL treatment. Both Salmonella enterica serovars Typhi and Typhimurium displayed different infection patterns, with S. Typhimurium causing more destruction of the terminal ileum and S. Typhi infecting the cecum more efficiently than the ileum, particularly with regard to adherence. Finally, various pathovars of Escherichia coli validated the model by confirming only adherence was observed with these strains. This work demonstrates that the combination of human-derived tissue with targeted bacterial growth conditions enables powerful analyses of human-specific infections that could lead to important insights into pathogenesis and accelerate future vaccine development. IMPORTANCE While traditional laboratory techniques and animal models have provided valuable knowledge in discerning virulence mechanisms of enteric pathogens, the complexity of the human gastrointestinal tract has hindered our understanding of physiologically relevant, human-specific interactions; and thus, has significantly delayed successful vaccine development. The human intestinal organoid-derived epithelial monolayer (HIODEM) model closely recapitulates the diverse cell populations of the intestine, allowing for the study of human-specific infections. Differentiation conditions permit the expansion of various cell populations, including M cells that are vital to immune recognition and the establishment of infection by some bacteria. We provide details of reproducible culture methods and infection conditions for the analyses of Shigella, Salmonella, and pathogenic Escherichia coli in which tissue tropism and pathogen-specific infection patterns were detected. This system will be vital for future studies that explore infection conditions, health status, or epigenetic differences and will serve as a novel screening platform for therapeutic development.
Collapse
Affiliation(s)
- Kourtney P. Nickerson
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alejandro Llanos-Chea
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Ingano
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gloria Serena
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alba Miranda-Ribera
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Meryl Perlman
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Rosiane Lima
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Juanes MA. Cytoskeletal Control and Wnt Signaling-APC's Dual Contributions in Stem Cell Division and Colorectal Cancer. Cancers (Basel) 2020; 12:E3811. [PMID: 33348689 PMCID: PMC7766042 DOI: 10.3390/cancers12123811] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal epithelium architecture is sustained by stem cell division. In principle, stem cells can divide symmetrically to generate two identical copies of themselves or asymmetrically to sustain tissue renewal in a balanced manner. The choice between the two helps preserve stem cell and progeny pools and is crucial for tissue homeostasis. Control of spindle orientation is a prime contributor to the specification of symmetric versus asymmetric cell division. Competition for space within the niche may be another factor limiting the stem cell pool. An integrative view of the multiple links between intracellular and extracellular signals and molecular determinants at play remains a challenge. One outstanding question is the precise molecular roles of the tumour suppressor Adenomatous polyposis coli (APC) for sustaining gut homeostasis through its respective functions as a cytoskeletal hub and a down regulator in Wnt signalling. Here, we review our current understanding of APC inherent activities and partners in order to explore novel avenues by which APC may act as a gatekeeper in colorectal cancer and as a therapeutic target.
Collapse
Affiliation(s)
- M. Angeles Juanes
- School of Health and Life Science, Teesside University, Middlesbrough TS1 3BX, UK;
- National Horizons Centre, Teesside University, 38 John Dixon Lane, Darlington DL1 1HG, UK
| |
Collapse
|
12
|
Hartl K, Sigal M. Microbe-Driven Genotoxicity in Gastrointestinal Carcinogenesis. Int J Mol Sci 2020; 21:E7439. [PMID: 33050171 PMCID: PMC7587957 DOI: 10.3390/ijms21207439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium serves as a barrier to discriminate the outside from the inside and is in constant exchange with the luminal contents, including nutrients and the microbiota. Pathogens have evolved mechanisms to overcome the multiple ways of defense in the mucosa, while several members of the microbiota can exhibit pathogenic features once the healthy barrier integrity of the epithelium is disrupted. This not only leads to symptoms accompanying the acute infection but may also contribute to long-term injuries such as genomic instability, which is linked to mutations and cancer. While for Helicobacter pylori a link between infection and cancer is well established, many other bacteria and their virulence factors have only recently been linked to gastrointestinal malignancies through epidemiological as well as mechanistic studies. This review will focus on those pathogens and members of the microbiota that have been linked to genotoxicity in the context of gastric or colorectal cancer. We will address the mechanisms by which such bacteria establish contact with the gastrointestinal epithelium-either via an existing breach in the barrier or via their own virulence factors as well as the mechanisms by which they interfere with host genomic integrity.
Collapse
Affiliation(s)
- Kimberly Hartl
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, 10117 Berlin, Germany;
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Michael Sigal
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, 10117 Berlin, Germany;
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| |
Collapse
|
13
|
Li J, Li X, Song J, Yan B, Rock S, Jia J, Liu J, Wang C, Weiss T, Weiss HL, Gao T, Alam A, Evers BM. Absence of neurotensin attenuates intestinal dysbiosis and inflammation by maintaining Mmp7/α-defensin axis in diet-induced obese mice. FASEB J 2020; 34:8596-8610. [PMID: 32359121 PMCID: PMC7754978 DOI: 10.1096/fj.201902374rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 12/19/2022]
Abstract
We previously reported that high levels of plasma neurotensin (NT), a gut hormone released from enteroendocrine cells of the small bowel, contribute to obesity and comorbid conditions. Gut microbiota has been implicated in the obesity development. Paneth cells are critical in maintaining gut microbiota composition and homeostasis by releasing antimicrobial proteins including α-defensins. The purpose of our current study was to determine the possible role of NT in gut microbiota composition and α-defensin gene expression associated with obesity. Here we show that the ratio of Firmicutes/Bacteroidetes (F/B ratio) and intestinal proinflammatory cytokines is significantly increased in NT+/+ mice fed with a high-fat diet (HFD) which were improved in NT-deficient mice. HFD disrupted the intestinal Mmp7/α-defensin axis, which was completely prevented in NT-/- mice. In addition, NT treatment inhibited DEFA5 expression and concurrent NF-κB activity, which was blocked by a pan PKC inhibitor (Gö6983) or an inhibitor for atypical PKCs (CRT0066854). More importantly, the shRNA-mediated knockdown of atypical PKCτ reversed NT-attenuated DEFA5 expression and increased NF-κB activity. NT contributes to the HFD-induced disruption of gut microbiota composition and α-defensin expression. PKCτ/λ plays a central role in NT-mediated α-defensin gene expression which might be mediated through the inhibition of NF-κB signaling pathways in Paneth cells.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Xian Li
- Department of Surgery, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jun Song
- Department of Surgery, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Baoxiang Yan
- Department of Surgery, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Stephanie Rock
- Department of Surgery, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jianhang Jia
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Todd Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Heidi L. Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Tianyan Gao
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Ashfaqul Alam
- Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - B. Mark Evers
- Department of Surgery, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
14
|
Mikuda N, Schmidt-Ullrich R, Kärgel E, Golusda L, Wolf J, Höpken UE, Scheidereit C, Kühl AA, Kolesnichenko M. Deficiency in IκBα in the intestinal epithelium leads to spontaneous inflammation and mediates apoptosis in the gut. J Pathol 2020; 251:160-174. [PMID: 32222043 DOI: 10.1002/path.5437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/25/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022]
Abstract
The IκB kinase (IKK)-NF-κB signaling pathway plays a multifaceted role in inflammatory bowel disease (IBD): on the one hand, it protects from apoptosis; on the other, it activates transcription of numerous inflammatory cytokines and chemokines. Although several murine models of IBD rely on disruption of IKK-NF-κB signaling, these involve either knockouts of a single family member of NF-κB or of upstream kinases that are known to have additional, NF-κB-independent, functions. This has made the distinct contribution of NF-κB to homeostasis in intestinal epithelium cells difficult to assess. To examine the role of constitutive NF-κB activation in intestinal epithelial cells, we generated a mouse model with a tissue-specific knockout of the direct inhibitor of NF-κB, Nfkbia/IκBα. We demonstrate that constitutive activation of NF-κB in intestinal epithelial cells induces several hallmarks of IBD including increased apoptosis, mucosal inflammation in both the small intestine and the colon, crypt hyperplasia, and depletion of Paneth cells, concomitant with aberrant Wnt signaling. To determine which NF-κB-driven phenotypes are cell-intrinsic, and which are extrinsic and thus require the immune compartment, we established a long-term organoid culture. Constitutive NF-κB promoted stem-cell proliferation, mis-localization of Paneth cells, and sensitization of intestinal epithelial cells to apoptosis in a cell-intrinsic manner. Increased number of stem cells was accompanied by a net increase in Wnt activity in organoids. Because aberrant Wnt signaling is associated with increased risk of cancer in IBD patients and because NFKBIA has recently emerged as a risk locus for IBD, our findings have critical implications for the clinic. In a context of constitutive NF-κB, our findings imply that general anti-inflammatory or immunosuppressive therapies should be supplemented with direct targeting of NF-κB within the epithelial compartment in order to attenuate apoptosis, inflammation, and hyperproliferation. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Nadine Mikuda
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Ruth Schmidt-Ullrich
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Eva Kärgel
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Laura Golusda
- Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, iPATH.Berlin - Core Unit for Immunopathology, Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Anja A Kühl
- Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, iPATH.Berlin - Core Unit for Immunopathology, Berlin, Germany
| | - Marina Kolesnichenko
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| |
Collapse
|
15
|
Koeninger L, Armbruster NS, Brinch KS, Kjaerulf S, Andersen B, Langnau C, Autenrieth SE, Schneidawind D, Stange EF, Malek NP, Nordkild P, Jensen BAH, Wehkamp J. Human β-Defensin 2 Mediated Immune Modulation as Treatment for Experimental Colitis. Front Immunol 2020; 11:93. [PMID: 32076420 PMCID: PMC7006816 DOI: 10.3389/fimmu.2020.00093] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/14/2020] [Indexed: 12/18/2022] Open
Abstract
Defensins represents an integral part of the innate immune system serving to ward off potential pathogens and to protect the intestinal barrier from microbial encroachment. In addition to their antimicrobial activities, defensins in general, and human β-defensin 2 (hBD2) in particular, also exhibit immunomodulatory capabilities. In this report, we assessed the therapeutic efficacy of systemically administered recombinant hBD2 to ameliorate intestinal inflammation in three distinct animal models of inflammatory bowel disease; i.e., chemically induced mucosal injury (DSS), loss of mucosal tolerance (TNBS), and T-cell transfer into immunodeficient recipient mice. Treatment efficacy was confirmed in all tested models, where systemically administered hBD2 mitigated inflammation, improved disease activity index, and hindered colitis-induced body weight loss on par with anti-TNF-α and steroids. Treatment of lipopolysaccharide (LPS)-activated human peripheral blood mononuclear cells with rhBD2 confirmed the immunomodulatory capacity in the circulatory compartment. Subsequent analyzes revealed dendritic cells (DCs) as the main target population. Suppression of LPS-induced inflammation was dependent on chemokine receptor 2 (CCR2) expression. Mechanistically, hBD2 engaged with CCR2 on its DC target cell to decrease NF-κB, and increase CREB phosphorylation, hence curbing inflammation. To our knowledge, this is the first study showing in vivo efficacy of a systemically administered defensin in experimental disease.
Collapse
Affiliation(s)
- Louis Koeninger
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Nicole S Armbruster
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | | | | | | | - Carolin Langnau
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Stella E Autenrieth
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Dominik Schneidawind
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Eduard F Stange
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Nisar P Malek
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | | | - Benjamin A H Jensen
- Department of Medicine, Faculty of Medicine, Cardiology Axis, Quebec Heart and Lung Institute, Laval University, Quebec, QC, Canada.,Section for Human Genomics and Metagenomics in Metabolism, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jan Wehkamp
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Chen L, Strohmeier V, He Z, Deshpande M, Catalan-Dibene J, Durum SK, Moran TM, Kraus T, Xiong H, Faith JJ, Sodhi CP, Hackam DJ, Lira SA, Furtado GC. Interleukin 22 disrupts pancreatic function in newborn mice expressing IL-23. Nat Commun 2019; 10:4517. [PMID: 31586069 PMCID: PMC6778080 DOI: 10.1038/s41467-019-12540-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
Neonatal inflammatory diseases are associated with severe morbidity, but the inflammatory factors underlying them and their potential effector mechanisms are poorly defined. Here we show that necrotizing enterocolitis in neonate mice is accompanied by elevation of IL-23 and IL-22 and decreased production of pancreatic enzymes. These phenotypes are mirrored in neonate mice overexpressing IL-23 in CX3CR1+ myeloid cells or in keratinocytes. The mice fail to grow and die prematurely, displaying systemic inflammation, nutrient malabsorption and decreased expression of intestinal and pancreatic genes mediating digestion and absorption of carbohydrates, proteins, and lipids. Germ-free environment improves, and genetic ablation of IL-22 restores normal growth in mice overexpressing IL-23. Mechanistically, IL-22 acts directly at the level of pancreatic acinar cells to decrease expression of the pancreas associated transcription factor 1a (PTF1a). These results show that augmented production of IL-23 and IL-22 in early life has a negative impact on pancreatic enzyme secretion and food absorption.
Collapse
Affiliation(s)
- Lili Chen
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Valentina Strohmeier
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104, Freiburg, Germany
| | - Zhengxiang He
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Madhura Deshpande
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jovani Catalan-Dibene
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Scott K Durum
- Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Thomas M Moran
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Therapeutic Antibody Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Thomas Kraus
- Center for Therapeutic Antibody Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Huabao Xiong
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chhinder P Sodhi
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - David J Hackam
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - Sergio A Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Glaucia C Furtado
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
17
|
Wu Y, Tang L, Wang B, Sun Q, Zhao P, Li W. The role of autophagy in maintaining intestinal mucosal barrier. J Cell Physiol 2019; 234:19406-19419. [PMID: 31020664 DOI: 10.1002/jcp.28722] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/23/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
The intestinal mucosal barrier is the first line to defense against luminal content penetration and performs numerous biological functions. The intestinal epithelium contains a huge surface that is lined by a monolayer of intestinal epithelial cells (IECs). IECs are dominant mediators in maintaining intestinal homeostasis that drive diverse functions including nutrient absorption, physical segregation, secretion of antibacterial peptides, and modulation of immune responses. Autophagy is a cellular self-protection mechanism in response to various stresses, and accumulating studies have revealed its importance in participating physiological processes of IECs. The regulatory effects of autophagy depend on the specific IEC types. This review aims to elucidate the myriad roles of autophagy in regulating the functions of different IECs (stem cells, enterocytes, goblet cells, and Paneth cells), and present the progress of autophagy-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide new preventive and therapeutic strategies for gastrointestinal dysfunction and diseases.
Collapse
Affiliation(s)
- Yanping Wu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Li Tang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Baikui Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pengwei Zhao
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Alrajban WA, Khubrani RA, Almalki MS, Almassri A, Alrikabi AC. Extensive Paneth cell metaplasia in an ovarian Krukenberg tumor: report of an unusual case and literature review. J Surg Case Rep 2018; 2018:rjy323. [PMID: 30555671 PMCID: PMC6289214 DOI: 10.1093/jscr/rjy323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/07/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Paneth cells are classified as secretory cells which are normally found in the cecum and ascending colon. Their presence in other parts of the gastrointestinal tract is regarded as abnormal and indicates metaplasia. Paneth cells may also be rarely found in gastrointestinal, biliary and prostatic tumors. The presence of Paneth cells in ovarian Krukenberg tumors is rare and to the best of our knowledge is restricted to metastatic appendiceal goblet cells tumors. We report a rare case of unilateral metastatic gastric carcinoma to the ovary in a 23-year-old female. This tumor showed unusual extensive Paneth cells metaplasia with classical signet ring cell morphology.
Collapse
Affiliation(s)
- Waleed Abdullah Alrajban
- Department of Pathology, King Khalid University Hospital and King Saud University, Riyadh, Saudi Arabia
| | - Reham Ali Khubrani
- Department of Pathology, King Khalid University Hospital and King Saud University, Riyadh, Saudi Arabia
| | - Mamdouh Saad Almalki
- Department of Radiology, King Khalid University Hospital and King Saud University, Riyadh, Saudi Arabia
| | - Ashraf Almassri
- Department of Pathology, Al Hammadi Hospital, Riyadh, Saudi Arabia
| | - Ammar Cherkess Alrikabi
- Department of Pathology, King Khalid University Hospital and King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Holly MK, Smith JG. Paneth Cells during Viral Infection and Pathogenesis. Viruses 2018; 10:v10050225. [PMID: 29701691 PMCID: PMC5977218 DOI: 10.3390/v10050225] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023] Open
Abstract
Paneth cells are major secretory cells located in the crypts of Lieberkühn in the small intestine. Our understanding of the diverse roles that Paneth cells play in homeostasis and disease has grown substantially since their discovery over a hundred years ago. Classically, Paneth cells have been characterized as a significant source of antimicrobial peptides and proteins important in host defense and shaping the composition of the commensal microbiota. More recently, Paneth cells have been shown to supply key developmental and homeostatic signals to intestinal stem cells in the crypt base. Paneth cell dysfunction leading to dysbiosis and a compromised epithelial barrier have been implicated in the etiology of Crohn’s disease and susceptibility to enteric bacterial infection. Our understanding of the impact of Paneth cells on viral infection is incomplete. Enteric α-defensins, produced by Paneth cells, can directly alter viral infection. In addition, α-defensins and other antimicrobial Paneth cell products may modulate viral infection indirectly by impacting the microbiome. Here, we discuss recent insights into Paneth cell biology, models to study their function, and the impact, both direct and indirect, of Paneth cells on enteric viral infection.
Collapse
Affiliation(s)
- Mayumi K Holly
- Department of Microbiology, University of Washington, Box 357735, 1705 NE Pacific St., Seattle, WA 98195, USA.
| | - Jason G Smith
- Department of Microbiology, University of Washington, Box 357735, 1705 NE Pacific St., Seattle, WA 98195, USA.
| |
Collapse
|
20
|
Wang SL, Shao BZ, Zhao SB, Fang J, Gu L, Miao CY, Li ZS, Bai Y. Impact of Paneth Cell Autophagy on Inflammatory Bowel Disease. Front Immunol 2018; 9:693. [PMID: 29675025 PMCID: PMC5895641 DOI: 10.3389/fimmu.2018.00693] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/21/2018] [Indexed: 12/19/2022] Open
Abstract
Intestinal mucosal barrier, mainly consisting of the mucus layer and epithelium, functions in absorbing nutrition as well as prevention of the invasion of pathogenic microorganisms. Paneth cell, an important component of mucosal barrier, plays a vital role in maintaining the intestinal homeostasis by producing antimicrobial materials and controlling the host-commensal balance. Current evidence shows that the dysfunction of intestinal mucosal barrier, especially Paneth cell, participates in the onset and progression of inflammatory bowel disease (IBD). Autophagy, a cellular stress response, involves various physiological processes, such as secretion of proteins, production of antimicrobial peptides, and degradation of aberrant organelles or proteins. In the recent years, the roles of autophagy in the pathogenesis of IBD have been increasingly studied. Here in this review, we mainly focus on describing the roles of Paneth cell autophagy in IBD as well as several popular autophagy-related genetic variants in Penath cell and the related therapeutic strategies against IBD.
Collapse
Affiliation(s)
- Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Bo-Zong Shao
- Department of Pharmocology, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Jun Fang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lun Gu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmocology, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Walsh DI, Dydek EV, Lock JY, Carlson TL, Carrier RL, Kong DS, Cabrera CR, Thorsen T. Emulation of Colonic Oxygen Gradients in a Microdevice. SLAS Technol 2017; 23:164-171. [PMID: 29186668 DOI: 10.1177/2472630317743425] [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] [Indexed: 11/15/2022]
Abstract
Gut-on-a-chip in vitro modeling is an emerging field, as the human gut epithelium and gut microbiome have been recently identified as novel drug targets for a wide variety of diseases. Realistic in vitro gut models require a variety of precise environmental cues, such as chemical and gas gradients, in combination with substrates like mucus that support the growth of microbial communities. This technical brief describes a microfluidic architecture capable of developing a physiologically relevant oxygen gradient that emulates the oxygen profile proximal to the epithelial inner lining of the human colon. The device generates stable and repeatable defined oxygen gradients from 0% to 4 % partial pressure O2 over a length scale of hundreds of microns, and was applied to study the effects of oxygenation on the structure of native mucus that lines the colon wall. Using simulation as a design tool for hybrid gas-liquid microfluidic devices enables on-chip creation of defined, physiologically oxygen gradients. These microfluidic architectures have powerful potential applications for gut physiology, including providing optimal oxygenation conditions for the culture of mammalian epithelial cells in the gut lining, as well as creating a realistic mimic of the oxygen gradient found in the intestinal lumen for complex microbiome cultures.
Collapse
Affiliation(s)
| | | | - Jaclyn Y Lock
- 2 Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Taylor L Carlson
- 3 Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Rebecca L Carrier
- 2 Department of Bioengineering, Northeastern University, Boston, MA, USA.,3 Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - David S Kong
- 1 MIT-Lincoln Laboratory, Lexington, MA, USA.,4 MIT Media Laboratory, Cambridge, MA, USA
| | | | | |
Collapse
|
22
|
Paneth cells in the developing gut: when do they arise and when are they immune competent? Pediatr Res 2016; 80:306-10. [PMID: 27049291 DOI: 10.1038/pr.2016.67] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Little is known about the perinatal development of Paneth cells (PCs) during gestation and the relation with necrotizing enterocolitis (NEC). We aimed to investigate when PCs arise and when they become immune competent during gestation. METHODS We included 57 samples of ileum tissue of fetuses/infants with a gestional age (GA) between 9 and 40 wk taken as part of a standard autopsy procedure. Hematoxylin-eosin staining and anti-human defensin 5 immunohistochemistry were performed. We performed a semi-quantitative assessment of (immune-competent) PC numbers per 10 crypts per tissue section per GA. RESULTS The number of PCs and the number of immune-competent PCs increased with increasing GA (Spearman's ρ = 0.41, P = 0.002 and ρ = 0.61, P < 0.001, respectively). Whereas significantly higher PC numbers were observed after 37 wk gestation (median 7, range 0-12) compared to preterm infants (median 0, range 0-15; P = 0.002), we counted higher numbers of immune-competent PCs already in infants with GA above 29 wk (median 6, range 0-18) compared to infants with GA under 29 wk (median 2, range 0-9; P < 0.001). CONCLUSION The significant increase of immune-competent PCs starting from a GA of 29 wk mimics the rise in incidence of NEC during a similar postmenstrual age in preterm infants.
Collapse
|
23
|
Nakamura K, Sakuragi N, Takakuwa A, Ayabe T. Paneth cell α-defensins and enteric microbiota in health and disease. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2015; 35:57-67. [PMID: 27200259 PMCID: PMC4858879 DOI: 10.12938/bmfh.2015-019] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/25/2015] [Indexed: 12/12/2022]
Abstract
Antimicrobial peptides are major effectors of innate immunity of multicellular organisms including humans and play a critical role in host defense, and their importance is widely recognized. The epithelium of the intestine is the largest surface area exposed to the outer environment, including pathogens, toxins and foods. The Paneth cell lineage of intestinal epithelial cells produces and secretes α-defensin antimicrobial peptides and functions in innate enteric immunity by removing pathogens and living symbiotically with commensal microbiota to contribute to intestinal homeostasis. Paneth cells secrete α-defensins, HD5 and HD6 in humans and cryptdins in mice, in response to bacterial, cholinergic and other stimuli. The α-defensins have selective activities against bacteria, eliciting potent microbicidal activities against pathogenic bacteria but minimal or no bactericidal activity against commensal bacteria. Therefore, α-defensins regulate the composition of the intestinal microbiota in vivo and play a role in homeostasis of the entire intestine. Recently, relationships between dysbiosis, or abnormal composition of the intestinal microbiota, and diseases such as inflammatory bowel disease and lifestyle diseases including obesity and atherosclerosis have been reported. Because α-defensins regulate the composition of the intestinal microbiota, Paneth cells and their α-defensins may have a key role as one mechanism linking the microbiota and disease.
Collapse
Affiliation(s)
- Kiminori Nakamura
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Naoya Sakuragi
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Akiko Takakuwa
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan; Department of Nutrition, Faculty of Nursing and Nutrition, Tenshi College, 3-1-30 Higashi, Kita-13, Higashi-ku, Sapporo, Hokkaido 065-0013, Japan
| | - Tokiyoshi Ayabe
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| |
Collapse
|
24
|
VanDussen KL, Marinshaw JM, Shaikh N, Miyoshi H, Moon C, Tarr PI, Ciorba MA, Stappenbeck TS. Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient-based assays. Gut 2015; 64:911-20. [PMID: 25007816 PMCID: PMC4305344 DOI: 10.1136/gutjnl-2013-306651] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 06/06/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The technology for the growth of human intestinal epithelial cells is rapidly progressing. An exciting possibility is that this system could serve as a platform for individualised medicine and research. However, to achieve this goal, human epithelial culture must be enhanced so that biopsies from individuals can be used to reproducibly generate cell lines in a short time frame so that multiple, functional assays can be performed (ie, barrier function and host-microbial interactions). DESIGN We created a large panel of human gastrointestinal epithelial cell lines (n=65) from patient biopsies taken during routine upper and lower endoscopy procedures. Proliferative stem/progenitor cells were rapidly expanded using a high concentration of conditioned media containing the factors critical for growth (Wnt3a, R-spondin and Noggin). A combination of lower conditioned media concentration and Notch inhibition was used to differentiate these cells for additional assays. RESULTS We obtained epithelial lines from all accessible tissue sites within 2 weeks of culture. The intestinal cell lines were enriched for stem cell markers and rapidly grew as spheroids that required passage at 1:3-1:4 every 3 days. Under differentiation conditions, intestinal epithelial spheroids showed region-specific development of mature epithelial lineages. These cells formed functional, polarised monolayers covered by a secreted mucus layer when grown on Transwell membranes. Using two-dimensional culture, these cells also demonstrated novel adherence phenotypes with various strains of pathogenic Escherichia coli. CONCLUSIONS This culture system will facilitate the study of interindividual, functional studies of human intestinal epithelial cells, including host-microbial interactions.
Collapse
Affiliation(s)
- Kelli L VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey M Marinshaw
- Department of Internal Medicine (Division of Gastroenterology), Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics (Division of Gastroenterology, Hepatology and Nutrition), Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hiroyuki Miyoshi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Clara Moon
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Phillip I Tarr
- Department of Pediatrics (Division of Gastroenterology, Hepatology and Nutrition), Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew A Ciorba
- Department of Internal Medicine (Division of Gastroenterology), Washington University School of Medicine, St. Louis, MO 63110, USA,Corresponding Authors: Thaddeus S. Stappenbeck, 660 S. Euclid, Box 8118, St. Louis, MO 63110, Phone: 314-362-4214, . Matthew A. Ciorba, 660 S. Euclid, Box 8124, St. Louis, MO 63110, Phone: 314-362-9054,
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA,Corresponding Authors: Thaddeus S. Stappenbeck, 660 S. Euclid, Box 8118, St. Louis, MO 63110, Phone: 314-362-4214, . Matthew A. Ciorba, 660 S. Euclid, Box 8124, St. Louis, MO 63110, Phone: 314-362-9054,
| |
Collapse
|
25
|
Abstract
Recurrence of Crohn's disease (CD) after ileal or colonic resection is common. Many studies have tried to identify predictors of postoperative recurrence (POR) in CD. A wide range of histologic features have been identified, but for most of them, the literature provided conflicting data. In last years, several studies have suggested that histologic findings including inflammatory changes within the enteric nervous system of the resection margin may be associated with CD recurrence. Herein, after briefly summarizing pathophysiology of POR, we review all histological features that have been studied so far: granulomas, histologic appearance at the margin of resection, plexitis, lymphatic vessel density in proximal margin of resection, and morphological analysis of Paneth cells. Granulomas and chronic inflammation at the margin of resection do not seem to predict POR in CD. Active disease at the margin of resection, plexitis, lymphatic vessels density, morphological analysis of Paneth cells may predict POR. Most of these histological features await replication in independent studies. Available evidence indicates that histological findings may be taken into account when developing strategies aimed at preventing postoperative CD recurrence.
Collapse
|
26
|
Bowcutt R, Forman R, Glymenaki M, Carding SR, Else KJ, Cruickshank SM. Heterogeneity across the murine small and large intestine. World J Gastroenterol 2014; 20:15216-15232. [PMID: 25386070 PMCID: PMC4223255 DOI: 10.3748/wjg.v20.i41.15216] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/18/2014] [Accepted: 06/17/2014] [Indexed: 02/06/2023] Open
Abstract
The small and large intestine of the gastrointestinal tract (GIT) have evolved to have discrete functions with distinct anatomies and immune cell composition. The importance of these differences is underlined when considering that different pathogens have uniquely adapted to live in each region of the gut. Furthermore, different regions of the GIT are also associated with differences in susceptibility to diseases such as cancer and chronic inflammation. The large and small intestine, given their anatomical and functional differences, should be seen as two separate immunological sites. However, this distinction is often ignored with findings from one area of the GIT being inappropriately extrapolated to the other. Focussing largely on the murine small and large intestine, this review addresses the literature relating to the immunology and biology of the two sites, drawing comparisons between them and clarifying similarities and differences. We also highlight the gaps in our understanding and where further research is needed.
Collapse
|
27
|
Rigoli L, Caruso RA. Inflammatory bowel disease in pediatric and adolescent patients: A biomolecular and histopathological review. World J Gastroenterol 2014; 20:10262-10278. [PMID: 25132743 PMCID: PMC4130834 DOI: 10.3748/wjg.v20.i30.10262] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/13/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Crohn’s disease (CD) and ulcerative colitis (UC) are the two main forms of inflammatory bowel disease (IBD) with both overlapping and distinct clinical, pathological and biomolecular features. It has been suggested that pediatric IBD is a distinct disease entity, with probably different disease subtypes.The aim of this study is to review and summarize the evolution of the current concept of pediatric IBD. The results of this review reinforce the idea that pediatric CD and UC may be further classified in various clinicopathologic entities. For clinicians and pathologists convenience, practical algorithms for the distinction of the various subphenotypes of pediatric IBD are also provided.
Collapse
|
28
|
Loss of Sonic hedgehog leads to alterations in intestinal secretory cell maturation and autophagy. PLoS One 2014; 9:e98751. [PMID: 24887421 PMCID: PMC4041759 DOI: 10.1371/journal.pone.0098751] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 05/07/2014] [Indexed: 12/22/2022] Open
Abstract
Background Intestinal epithelial cells express the Sonic and Indian hedgehog ligands. Despite the strong interest in gut hedgehog signaling in GI diseases, no studies have specifically addressed the singular role of intestinal epithelial cell Sonic hedgehog signaling. The aim of this study was to investigate the specific role of Sonic hedgehog in adult ileal epithelial homeostasis. Methodology/Principal Findings A Sonic hedgehog intestinal epithelial conditional knockout mouse model was generated. Assessment of ileal histological abnormalities, crypt epithelial cell proliferation, epithelial cell fate, junctional proteins, signaling pathways, as well as ultrastructural analysis of intracellular organelles were performed in control and mutant mice. Mice lacking intestinal epithelial Sonic Hedgehog displayed decreased ileal crypt/villus length, decreased crypt proliferation as well as a decrease in the number of ileal mucin-secreting goblet cells and antimicrobial peptide-secreting Paneth cells during adult life. These secretory cells also exhibited disruption of their secretory products in mutant mice. Ultrastructural microscopy analysis revealed a dilated ER lumen in secretory cells. This phenotype was also associated with a decrease in autophagy. Conclusions/Significance Altogether, these findings indicate that the loss of Sonic hedgehog can lead to ileal secretory cell modifications indicative of endoplasmic reticulum stress, accompanied by a significant reduction in autophagy.
Collapse
|
29
|
Wang X, Pierre JF, Heneghan AF, Busch RA, Kudsk KA. Glutamine Improves Innate Immunity and Prevents Bacterial Enteroinvasion During Parenteral Nutrition. JPEN J Parenter Enteral Nutr 2014; 39:688-97. [PMID: 24836948 DOI: 10.1177/0148607114535265] [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: 03/10/2014] [Accepted: 04/20/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Patients receiving parenteral nutrition (PN) are at increased risk of infectious complications compared with enteral feeding, which is in part explained by impaired mucosal immune function during PN. Adding glutamine (GLN) to PN has improved outcome in some clinical patient groups. Although GLN improves acquired mucosal immunity, its effect on innate mucosal immunity (defensins, mucus, lysozymes) has not been investigated. METHODS Forty-eight hours following venous cannulation, male Institute of Cancer Research mice were randomized to chow (n = 10), PN (n = 12), or PN + GLN (n = 13) for 5 days. Small intestine tissue and luminal fluid were collected for mucin 2 (MUC2), lysozyme, cryptdin 4 analysis, and luminal interleukin (IL)-4, IL-10, and IL-13 level measurement. Tissue was also harvested for ex vivo intestinal segment culture to assess tissue susceptibility to enteroinvasive Escherichia coli. RESULTS In both luminal and tissue samples, PN reduced MUC2 and lysozyme (P < .0001, respectively) compared with chow, whereas GLN addition increased MUC2 and lysozyme (luminal, P < .05; tissue, P < .0001, respectively) compared with PN alone. PN significantly suppressed cryptdin 4 expression, while GLN supplementation significantly enhanced expression. IL-4, IL-10, and IL-13 decreased significantly with PN compared with chow, whereas GLN significantly increased these cytokines compared with PN. Functionally, bacterial invasion increased with PN compared with chow (P < .05), while GLN significantly decreased enteroinvasion to chow levels (P < .05). CONCLUSIONS GLN-supplemented PN improves innate immunity and resistance to bacterial mucosal invasion lost with PN alone. This work confirms a clinical rationale for providing glutamine for the protection of the intestinal mucosa.
Collapse
Affiliation(s)
- Xinying Wang
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Department of Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Joseph F Pierre
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Aaron F Heneghan
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Rebecca A Busch
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kenneth A Kudsk
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
30
|
VanDussen KL, Liu TC, Li D, Towfic F, Modiano N, Winter R, Haritunians T, Taylor KD, Dhall D, Targan SR, Xavier RJ, McGovern DPB, Stappenbeck TS. Genetic variants synthesize to produce paneth cell phenotypes that define subtypes of Crohn's disease. Gastroenterology 2014; 146:200-9. [PMID: 24076061 PMCID: PMC3899786 DOI: 10.1053/j.gastro.2013.09.048] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 09/11/2013] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Genetic susceptibility loci for Crohn's disease (CD) are numerous, complex, and likely interact with undefined components of the environment. It has been a challenge to link the effects of particular loci to phenotypes of cells associated with pathogenesis of CD, such as Paneth cells. We investigated whether specific phenotypes of Paneth cells associated with particular genetic susceptibility loci can be used to define specific subtypes of CD. METHODS We performed a retrospective analysis of 119 resection specimens collected from patients with CD at 2 separate medical centers. Paneth cell phenotypes were classified as normal or abnormal (with disordered, diminished, diffuse, or excluded granule phenotypes) based on lysozyme-positive secretory granule morphology. To uncover the molecular basis of the Paneth cell phenotypes, we developed methods to determine transcriptional profiles from whole-thickness and laser-capture microdissected, formalin-fixed, paraffin-embedded tissue sections. RESULTS The proportion of abnormal Paneth cells was associated with the number of CD-associated NOD2 risk alleles. The cumulative number of NOD2 and ATG16L1 risk alleles had an additive effect on the proportion of abnormal Paneth cells. Unsupervised clustering analysis of demographic and Paneth cell data divided patients into 2 principal subgroups, defined by high and low proportions of abnormal Paneth cells. The disordered and diffuse abnormal Paneth cell phenotypes were associated with an altered transcriptional signature of immune system activation. We observed an inverse correlation between abnormal Paneth cells and presence of granuloma. In addition, high proportions of abnormal Paneth cells were associated with shorter time to disease recurrence after surgery. CONCLUSIONS Histologic analysis of Paneth cell phenotypes can be used to divide patients with CD into subgroups with distinct pathognomonic and clinical features.
Collapse
Affiliation(s)
- Kelli L. VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dalin Li
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Fadi Towfic
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School and Broad Institute, Boston, MA 02115, USA
| | - Nir Modiano
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rachel Winter
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Talin Haritunians
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kent D. Taylor
- Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Deepti Dhall
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephan R. Targan
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School and Broad Institute, Boston, MA 02115, USA
| | - Dermot P. B. McGovern
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Corresponding Authors: Thaddeus S. Stappenbeck, 660 S. Euclid Ave, Box 8118, St. Louis, MO 63110, Phone: 314-362-4214, . Dermot P. B. McGovern, 8797 Beverly Blvd, Suite 300, Los Angeles, CA, 90048,
| | - Thaddeus S. Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA,Corresponding Authors: Thaddeus S. Stappenbeck, 660 S. Euclid Ave, Box 8118, St. Louis, MO 63110, Phone: 314-362-4214, . Dermot P. B. McGovern, 8797 Beverly Blvd, Suite 300, Los Angeles, CA, 90048,
| |
Collapse
|
31
|
Abstract
Paneth cells are highly specialized epithelial cells of the small intestine, where they coordinate many physiological functions. First identified more than a century ago on the basis of their readily discernible secretory granules by routine histology, these cells are located at the base of the crypts of Lieberkühn, tiny invaginations that line the mucosal surface all along the small intestine. Investigations over the past several decades determined that these cells synthesize and secrete substantial quantities of antimicrobial peptides and proteins. More recent studies have determined that these antimicrobial molecules are key mediators of host-microbe interactions, including homeostatic balance with colonizing microbiota and innate immune protection from enteric pathogens. Perhaps more intriguing, Paneth cells secrete factors that help sustain and modulate the epithelial stem and progenitor cells that cohabitate in the crypts and rejuvenate the small intestinal epithelium. Dysfunction of Paneth cell biology contributes to the pathogenesis of chronic inflammatory bowel disease.
Collapse
Affiliation(s)
- Hans C Clevers
- Hubrecht Institute-KNAW, University Medical Center Utrecht, Uppsalalaan, Utrecht 3584CT, The Netherlands.
| | | |
Collapse
|
32
|
Rossi O, van Baarlen P, Wells JM. Host-recognition of pathogens and commensals in the mammalian intestine. Curr Top Microbiol Immunol 2013; 358:291-321. [PMID: 22179258 DOI: 10.1007/82_2011_191] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To peacefully coexist with the microbial inhabitants of the intestine, mammals have evolved elaborate and interconnected regulatory mechanisms to maintain immune homeostasis in the face of potential infection and tissue damage by pathogenic microorganisms. Physical barriers, antimicrobial factors and secretory antibodies act in concert to keep microbes at a distance from the epithelium and initiate repair mechanisms in the event of damage. Commensal bacteria are not ignored but dynamically controlled via many complex overlapping and intertwined mechanisms involving intestinal epithelial cells (IECs) and signals from the microbiota. Polarized IECs play a decisive role in homeostasis by regulating the expression and activity of the pattern-recognition receptors (PRRs), in different compartments of the intestine. The differential signaling and expression of receptors on apical and basal membranes of the epithelium also plays its part in distinguishing commensals from harmful invaders. In steady state conditions macrophages and dendritic cells (DCs) in the lamina propria (LP) are conditioned by environmental factors to induce immune tolerance. The distinction between pathogen and non-pathogen is linked to the ability of pathogens to invade and cause damage to the host cells and tissues. This induces local inflammatory responses and the attraction of capillary leukocytes by chemokines released from colonized and invaded epithelial cells. This bypasses the tolerogenic mechanisms controlling the responses of resident DCs and macrophages leading to pathogen killing and adaptive immune responses. Research on this topic has important implications for the development of novel therapeutic approaches to treat or prevent inflammatory bowel disease (IBD), inflammation-related cancer and other gut-related diseases and disorders.
Collapse
Affiliation(s)
- Oriana Rossi
- Wageningen University and Research Centre, Wageningen, The Netherlands
| | | | | |
Collapse
|
33
|
Xing XF, Li H, Zhong XY, Zhang LH, Wang XH, Liu YQ, Jia SQ, Shi T, Niu ZJ, Peng Y, Du H, Zhang GG, Hu Y, Lu AP, Li JY, Chen S, Ji JF. Phospholipase A2 group IIA expression correlates with prolonged survival in gastric cancer. Histopathology 2011; 59:198-206. [DOI: 10.1111/j.1365-2559.2011.03913.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
34
|
Ouellette AJ. Paneth cell α-defensins in enteric innate immunity. Cell Mol Life Sci 2011; 68:2215-29. [PMID: 21560070 PMCID: PMC4073591 DOI: 10.1007/s00018-011-0714-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 04/26/2011] [Accepted: 04/26/2011] [Indexed: 12/18/2022]
Abstract
Paneth cells at the base of small intestinal crypts of Lieberkühn secrete high levels of α-defensins in response to cholinergic and microbial stimuli. Paneth cell α-defensins are broad spectrum microbicides that function in the extracellular environment of the intestinal lumen, and they are responsible for the majority of secreted bactericidal peptide activity. Paneth cell α-defensins confer immunity to oral infection by Salmonella enterica serovar Typhimurium, and they are major determinants of the composition of the small intestinal microbiome. In addition to host defense molecules such as α-defensins, lysozyme, and Pla2g2a, Paneth cells also produce and release proinflammatory mediators as components of secretory granules. Disruption of Paneth cell homeostasis, with subsequent induction of endoplasmic reticulum stress, autophagy, or apoptosis, contributes to inflammation in diverse genetic and experimental mouse models.
Collapse
Affiliation(s)
- André Joseph Ouellette
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC/Norris Cancer Center, Los Angeles, CA 90089-9601, USA.
| |
Collapse
|
35
|
McGuckin MA, Lindén SK, Sutton P, Florin TH. Mucin dynamics and enteric pathogens. Nat Rev Microbiol 2011. [PMID: 21407243 DOI: 10.1038/nrm] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The extracellular secreted mucus and the cell surface glycocalyx prevent infection by the vast numbers of microorganisms that live in the healthy gut. Mucin glycoproteins are the major component of these barriers. In this Review, we describe the components of the secreted and cell surface mucosal barriers and the evidence that they form an effective barricade against potential pathogens. However, successful enteric pathogens have evolved strategies to circumvent these barriers. We discuss the interactions between enteric pathogens and mucins, and the mechanisms that these pathogens use to disrupt and avoid mucosal barriers. In addition, we describe dynamic alterations in the mucin barrier that are driven by host innate and adaptive immune responses to infection.
Collapse
Affiliation(s)
- Michael A McGuckin
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute and The University of Queensland School of Medicine, South Brisbane, Queensland 4101, Australia.
| | | | | | | |
Collapse
|
36
|
|
37
|
Abstract
PURPOSE OF REVIEW Recent evidence shows that disruption of Paneth cell homeostasis by induction of endoplasmic reticulum stress or autophagy, with consequent apoptosis, contributes to inflammation and morbidity in a variety of experimental mouse models. RECENT FINDINGS Recent advances show that proinflammatory mediators in Paneth cell dense core secretory granules mediate tumor necrosis factor-α-induced shock, that Paneth cell α-defensins modulate the composition of the small intestinal microflora, that development of crypt organoid culture systems provides a novel means for investigating the crypt microenvironment, and that varied genetic defects that disrupt Paneth cell homeostasis are emergent as risk factors in inflammatory bowel disease. SUMMARY This recent literature identifies Paneth cells as particularly sensitive targets of endoplasmic reticulum stress responses and implicates this unique small intestinal lineage in inflammatory bowel disease pathogenesis resulting from diverse heritable and environmental causes.
Collapse
Affiliation(s)
- André J Ouellette
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of The University of Southern California, Los Angeles, California 90089-9601, USA.
| |
Collapse
|
38
|
Shanahan MT, Vidrich A, Shirafuji Y, Dubois CL, Henschen-Edman A, Hagen SJ, Cohn SM, Ouellette AJ. Elevated expression of Paneth cell CRS4C in ileitis-prone SAMP1/YitFc mice: regional distribution, subcellular localization, and mechanism of action. J Biol Chem 2010; 285:7493-504. [PMID: 20056603 DOI: 10.1074/jbc.m109.083220] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Paneth cells at the base of small intestinal crypts of Lieberkühn secrete host defense peptides and proteins, including alpha-defensins, as mediators of innate immunity. Mouse Paneth cells also express alpha-defensin-related Defcr-rs genes that code for cysteine-rich sequence 4C (CRS4C) peptides that have a unique CPX triplet repeat motif. In ileitis-prone SAMP1/YitFc mice, Paneth cell levels of CRS4C mRNAs and peptides are induced more than a 1000-fold relative to non-prone strains as early as 4 weeks of age, with the mRNA and peptide levels highest in distal ileum and below detection in duodenum. CRS4C-1 peptides are found exclusively in Paneth cells where they occur only in dense core granules and thus are secreted to function in the intestinal lumen. CRS4C bactericidal peptide activity is membrane-disruptive in that it permeabilizes Escherichia coli and induces rapid microbial cell K(+) efflux, but in a manner different from mouse alpha-defensin cryptdin-4. In in vitro studies, inactive pro-CRS4C-1 is converted to bactericidal CRS4C-1 peptide by matrix metalloproteinase-7 (MMP-7) proteolysis of the precursor proregion at the same residue positions that MMP-7 activates mouse pro-alpha-defensins. The absence of processed CRS4C in protein extracts of MMP-7-null mouse ileum demonstrates the in vivo requirement for intracellular MMP-7 in pro-CRS4C processing.
Collapse
Affiliation(s)
- Michael T Shanahan
- Department of Microbiology and Molecular Genetics, School of Medicine, College of Health Sciences, University of California, Irvine, California 92697-4800, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Stappenbeck TS. The role of autophagy in Paneth cell differentiation and secretion. Mucosal Immunol 2010; 3:8-10. [PMID: 19890269 DOI: 10.1038/mi.2009.121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Paneth cells are a small intestinal epithelial cell lineage that is considered to have a role in innate immune function. Recent studies on mice with diminished and/or loss of autophagy have suggested that Paneth cells are a primary target in vivo. Interestingly, loss of autophagy affects the secretion of antimicrobial proteins from Paneth cells. Understanding the intersection of the autophagy pathway with the secretory apparatus, which is a key feature of differentiation of Paneth cells, is a key unanswered question.
Collapse
Affiliation(s)
- T S Stappenbeck
- Department of Pathology and Immunology Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|