1
|
Keppeler K, Pesi A, Lange S, Helmstädter J, Strohm L, Ubbens H, Kuntić M, Kuntić I, Mihaliková D, Vujačić-Mirski K, Rosenberger A, Küster L, Frank C, Oelze M, Finger S, Zakrzewska A, Verdu E, Wild J, Karbach S, Wenzel P, Wild P, Leistner D, Münzel T, Daiber A, Schuppan D, Steven S. Vascular dysfunction and arterial hypertension in experimental celiac disease are mediated by gut-derived inflammation and oxidative stress. Redox Biol 2024; 70:103071. [PMID: 38354629 PMCID: PMC10876911 DOI: 10.1016/j.redox.2024.103071] [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: 01/06/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
AIMS We examined the cardiovascular effects of celiac disease (CeD) in a humanized mouse model, with a focus on vascular inflammation, endothelial dysfunction, and oxidative stress. METHODS AND RESULTS NOD.DQ8 mice genetically predisposed to CeD were subjected to a diet regime and oral gavage to induce the disease (gluten group vs. control). We tested vascular function, confirmed disease indicators, and evaluated inflammation and oxidative stress in various tissues. Plasma proteome profiling was also performed. CeD markers were confirmed in the gluten group, indicating increased blood pressure and impaired vascular relaxation. Pro-inflammatory genes were upregulated in this group, with increased CD11b+ myeloid cell infiltration and oxidative stress parameters observed in aortic and heart tissue. However, heart function remained unaffected. Plasma proteomics suggested the cytokine interleukin-17A (IL-17A) as a link between gut and vascular inflammation. Cardiovascular complications were reversed by adopting a gluten-free diet. CONCLUSION Our study sheds light in the heightened cardiovascular risk associated with active CeD, revealing a gut-to-cardiovascular inflammatory axis potentially mediated by immune cell infiltration and IL-17A. These findings augment our understanding of the link between CeD and cardiovascular disease providing clinically relevant insight into the underlying mechanism. Furthermore, our discovery that cardiovascular complications can be reversed by a gluten-free diet underscores a critical role for dietary interventions in mitigating cardiovascular risks associated with CeD.
Collapse
Affiliation(s)
- Karin Keppeler
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Aline Pesi
- Institute of Translational Immunology (TIM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Simon Lange
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Johanna Helmstädter
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Lea Strohm
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Henning Ubbens
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Marin Kuntić
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ivana Kuntić
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dominika Mihaliková
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ksenija Vujačić-Mirski
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Alexandra Rosenberger
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Leonie Küster
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Charlotte Frank
- Institute of Translational Immunology (TIM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Agnieszka Zakrzewska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Elena Verdu
- Farncombe Digestive Disease Center, McMaster University, Hamilton, Canada
| | - Johannes Wild
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Susanne Karbach
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Philip Wenzel
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Philipp Wild
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - David Leistner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany; Division of Cardiology, Goethe University Frankfurt, University Hospital, Department of Medicine III, Frankfurt a. M., Germany
| | - Thomas Münzel
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Andreas Daiber
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz/Frankfurt a. M., Germany
| | - Detlef Schuppan
- Institute of Translational Immunology (TIM), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sebastian Steven
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Division of Cardiology, Goethe University Frankfurt, University Hospital, Department of Medicine III, Frankfurt a. M., Germany.
| |
Collapse
|
2
|
II T, CHAMBERS JK, NAKASHIMA K, GOTO-KOSHINO Y, UCHIDA K. Intraepithelial lymphocytes are associated with epithelial injury in feline intestinal T-cell lymphoma. J Vet Med Sci 2024; 86:101-110. [PMID: 38072403 PMCID: PMC10849855 DOI: 10.1292/jvms.23-0339] [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: 08/07/2023] [Accepted: 11/25/2023] [Indexed: 01/30/2024] Open
Abstract
Our previous study indicated that cytotoxicity of intraepithelial lymphocytes is a poor prognostic factor in feline intestinal T-cell lymphoma (FITL), but the effect of cytotoxic lymphocytes on mucosal epithelium is still unknown. Thus, we investigated the association between cytotoxic lymphocytes and mucosal epithelium in 71 cases of feline intestinal T-cell lymphoma (FITL): epithelial injury, basement membrane injury, cleaved-caspase-3 positivity of epithelial cells, and the number and Ki67 positivity of intraepithelial lymphocytes in granzyme B (GRB)+ and GRB- FITLs were evaluated. Epithelial injury score and the number of intraepithelial lymphocytes in granzyme B (GRB)+ FITL were significantly higher than those of GRB- FITL (P<0.05, P<0.05), but no significant differences were found in the basement membrane injury score, the percentage of cleaved-caspase-3+ epithelial cells, and the percentage of Ki67+ intraepithelial lymphocytes. There was a significant correlation between the epithelial injury score and the number of intraepithelial lymphocytes (P<0.05), but no significant correlation was observed between the epithelial injury score and Ki67+ percentage of intraepithelial lymphocytes. Because epithelial cell cleaved-caspase-3 positivity was observed in FITL, regardless of GRB expression in lymphocytes, GRB-mediated apoptosis may not contribute to epithelial injury in FITL. The association between increased number of intraepithelial lymphocytes and epithelial injury suggests that intraepithelial lymphocytes infiltration may contribute to epithelial injury in FITL.
Collapse
Affiliation(s)
- Tatsuhito II
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - James K CHAMBERS
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ko NAKASHIMA
- Japan Small Animal Medical Center (JSAMC), Tokorozawa, Saitama, Japan
| | - Yuko GOTO-KOSHINO
- Veterinary Medical Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki UCHIDA
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Ruera CN, Perez F, Iribarren ML, Guzman L, Menendez L, Garbi L, Chirdo FG. Coexistence of apoptosis, pyroptosis, and necroptosis pathways in celiac disease. Clin Exp Immunol 2023; 214:328-340. [PMID: 37455655 PMCID: PMC10719221 DOI: 10.1093/cei/uxad082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/12/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023] Open
Abstract
Usually, the massive elimination of cells under steady-state conditions occurs by apoptosis, which is also acknowledged to explain the loss of enterocytes in the small intestine of celiac disease (CD) patients. However, little is known about the role of proinflammatory cell death pathways in CD. Here, we have used confocal microscopy, western blot, and RT-qPCR analysis to assess the presence of regulated cell death pathways in the duodenum of CD patients. We found an increased number of dead (TUNEL+) cells in the lamina propria of small intestine of CD patients, most of them are plasma cells (CD138+). Many dying cells expressed FAS and were in close contact with CD3+ T cells. Caspase-8 and caspase-3 expression was increased in CD, confirming the activation of apoptosis. In parallel, caspase-1, IL-1β, and GSDMD were increased in CD samples indicating the presence of inflammasome-dependent pyroptosis. Necroptosis was also present, as shown by the increase of RIPK3 and phosphorylate MLKL. Analysis of published databases confirmed that CD has an increased expression of regulated cell death -related genes. Together, these results reveal that CD is characterized by cell death of different kinds. In particular, the presence of proinflammatory cell death pathways may contribute to mucosal damage.
Collapse
Affiliation(s)
- Carolina N Ruera
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Federico Perez
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María Luz Iribarren
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Luciana Guzman
- Servicio de Gastroenterología Hospital de Niños “Sor María Ludovica”, La Plata, Argentina
| | - Lorena Menendez
- Servicio de Gastroenterología Hospital de Niños “Sor María Ludovica”, La Plata, Argentina
| | - Laura Garbi
- Servicio de Gastroenterología, HospitalSan Martin, La Plata, Argentina
| | - Fernando G Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
4
|
Amundsen SF, Stamnaes J, Lundin KEA, Sollid LM. Expression of transglutaminase 2 in human gut epithelial cells: Implications for coeliac disease. PLoS One 2023; 18:e0287662. [PMID: 37368893 PMCID: PMC10298751 DOI: 10.1371/journal.pone.0287662] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Formation of complexes between transglutaminase 2 (TG2) and gluten can mechanistically explain why TG2 serves both as B-cell autoantigen and as an enzyme that creates deamidated gluten epitopes in coeliac disease (CeD). A model has been proposed where TG2 released from shed epithelial cells encounters high concentrations of dietary gluten peptides to form these TG2:gluten complexes. In this work we have characterised TG2 protein expression in gut epithelial cells in humans. METHODS Western blot analysis, immunofluorescence staining and mass spectrometry in combination with laser capture microdissection to gain spatial resolution were used to characterise TG2 expression in the epithelial cell layer of healthy and coeliac disease affected duodenum. FINDINGS TG2 is expressed in human duodenal epithelial cells, including cells in the apical region that are shed into the gut lumen. In untreated CeD the apical expression of TG2 is doubled. Enzymatically active TG2 is readily released from isolated human intestinal epithelial cells. CONCLUSION Shed epithelial cells are a plausible source of pathogenic TG2 enzyme in CeD. Increased epithelial TG2 expression and increased epithelial shedding in active CeD may reinforce action of luminal TG2 in this condition.
Collapse
Affiliation(s)
- Sunniva F. Amundsen
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital—Rikshospitalet, Oslo, Norway
| | - Jorunn Stamnaes
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital—Rikshospitalet, Oslo, Norway
| | - Knut E. A. Lundin
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Oslo University Hospital—Rikshospitalet, Oslo, Norway
| | - Ludvig M. Sollid
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital—Rikshospitalet, Oslo, Norway
| |
Collapse
|
5
|
Błaszczyk F, Sosinka A, Wilczek G, Student S, Rost-Roszkowska M. Effect of gluten on the digestive tract and fat body of Telodeinopus aoutii (Diplopoda). J Morphol 2023; 284:e21546. [PMID: 36533734 DOI: 10.1002/jmor.21546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Adult specimens or larvae of invertebrates used as food for vertebrates are often maintained close to gluten so they might become vectors for cereal proteins. However, the tissues and internal organs can respond differently in animals with different feeding habits. The midgut epithelium might be a first and sufficient barrier preventing uptake and effects of gluten on the whole body, while the fat body is the main organ that accumulates different xenobiotics. Good models for such research are animals that do not feed on gluten-rich products in their natural environment. The project's goal was to investigate alterations in the midgut epithelium and fat body of the herbivorous millipede Telodeinopus aoutii (Diplopoda) and analyze cell death processes activated by gluten. It enabled us to determine whether changes were intensified or reversed by adaptive mechanisms. Adult specimens were divided into control and experimental animals fed with mushrooms supplemented with gluten and analyzed using transmission electron microscopy, flow cytometry, and confocal microscopy. Two organs were isolated for the qualitative and quantitative analysis: the midgut and the fat body. Our study of the herbivorous T. aoutii which does not naturally feed on gluten containing diet showed that continuous and prolonged gluten feeding activates repair processes that inhibit the processes of cell death (apoptosis and necrosis) and induce an increase in cell viability.
Collapse
Affiliation(s)
- Florentyna Błaszczyk
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Agnieszka Sosinka
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Grażyna Wilczek
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Sebastian Student
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Gliwice, Poland.,Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Magdalena Rost-Roszkowska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
6
|
Li Y, Chen Y, Sun-Waterhouse D. The potential of dandelion in the fight against gastrointestinal diseases: A review. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115272. [PMID: 35405251 DOI: 10.1016/j.jep.2022.115272] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dandelion (Taraxacum officinale Weber ex F. H. Wigg.), as a garden weed grown globally, has long been consumed as a therapeutic herb. Its folkloric uses include treatments of digestive disorders (dyspepsia, anorexia, stomach disorders, gastritis and enteritis) and associate complex ailments involving uterine, liver and lung disorders. AIM OF THE STUDY The present study aims to critically assess the current state of research and summarize the potential roles of dandelion and its constituents in gastrointestinal (GI) -protective actions. A focus is placed on the reported bioactive components, pharmacological activities and modes of action (including molecular mechanisms and interactions among bioactive substances) of dandelion products/preparations and derived active constituents related to GI protection. MATERIALS AND METHODS The available information published prior to August 2021 was reviewed via SciFinder, Web of Science, Google Scholar, PubMed, Elsevier, Wiley On-line Library, and The Plant List. The search was based on the ethnomedical remedies, pharmacological activities, bioactive compounds of dandelion for GI protection, as well as the interactions of the components in dandelion with the gut microbiota or biological regulators, and with other ingested bioactive compounds. The key search words were "Taraxacum" and "dandelion". RESULTS T. coreanum Nakai, T. mongolicum and T. officinale are the most commonly used species for folkloric uses, with the whole plant, leaves and root of dandelion being used more frequently. GI-protective substances of dandelion include taraxasterol, taraxerol, caffeic acid, chicoric acid, chlorogenic acid, luteolin and its glucosides, polysaccharides, inulin, and β-sitosterol. Dandelion products and derived constituents exhibit pharmacological effects against GI disorders, mainly including dyspepsia, gastroesophageal reflux disease, gastritis, small intestinal ulcer, ulcerative colitis, liver diseases, gallstones, acute pancreatitis, and GI malignancy. The underlying molecular mechanisms may include immuno-inflammatory mechanisms, apoptosis mechanism, autophagy mechanism, and cholinergic mechanism, although interactions of dandelion's constituents with GI health-related biological entities (e.g., GI microbiota and associated biological modulators) or other ingested bioactive compounds shouldn't be ignored. CONCLUSION The review reveals some in vivo and in vitro studies on the potential of dandelion derived products as complementary and alternative medicines/therapeutics against GI disorders. The whole herb may alleviate some symptoms related GI immuno-inflammatory basing on the abundant anti-inflammatory and anti-oxide active substances. Dandelion root could be a nontoxic and effective anticancer alternative, owing to its abundant terpenoids and polysaccharides. However, research related to GI protective dandelion-derived products remains limited. Besides the need of identifying bioactive compounds/complexes in various dandelion species, more clinical studies are also required on the metabolism, bioavailability and safety of these substances to support their applications in food, medicine and pharmaceuticals.
Collapse
Affiliation(s)
- Yanni Li
- College of Food Science and Engineering, Shandong Agricultural University, Taian, 271018, Shandong Province, China
| | - Yilun Chen
- College of Food Science and Engineering, Shandong Agricultural University, Taian, 271018, Shandong Province, China.
| | - Dongxiao Sun-Waterhouse
- School of Chemical Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand.
| |
Collapse
|
7
|
Richter JF, Hildner M, Mrowka R, Schmauder R. Probing the leak pathway: Live-cell imaging of macromolecule passage through epithelia. Ann N Y Acad Sci 2022; 1516:151-161. [PMID: 35766317 DOI: 10.1111/nyas.14847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Epithelia compartmentalize multicellular organisms and provide interfacing between the inside and outside. Apart from regulating the exchange of solutes, uptake of nutrients, and excretion of waste products, their major function is to prevent uncontrolled access of foreign material to immune-competent compartments. Progress in understanding this barrier function toward larger solutes and its possible defects, as can be seen in a variety of diseases, is largely hampered by a lack of methods to spatiotemporally resolve transepithelial passage of macromolecules. Using different cell culture epithelia, we applied biotinylated dextran tracers carrying an acceptor fluorophore. These bind to cell-adherent avidin carrying donor fluorophore at the basolateral membranes of single-layered epithelial sheets. Confocal fluorescence microscopy was applied to living epithelia in order to image apical-to-basolateral tracer passage as a Förster resonance energy transfer signal of the fluorescent dextran-avidin pair over time. Stimulated macromolecule passage using barrier-perturbing agents proved its effectiveness for the leak imaging method presented herein. Over hours of imaging, spontaneous leaks were rare, occurring transiently on the scale of minutes and for the most part associated with rearranging cell junctions. The discussed approach to leak imaging is expected to promote the understanding of epithelial barriers, particularly, the nature and dynamics of the epithelial cell leak pathway.
Collapse
Affiliation(s)
- Jan F Richter
- Jena University Hospital, Institute of Anatomy II, Friedrich Schiller University Jena, Jena, Germany
| | - Markus Hildner
- Jena University Hospital, Institute of Anatomy II, Friedrich Schiller University Jena, Jena, Germany
| | - Ralf Mrowka
- Jena University Hospital, KIM III Department of Experimental Nephrology, Friedrich Schiller University Jena, Jena, Germany
| | - Ralf Schmauder
- Jena University Hospital, Institute of Physiology II, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
8
|
Ohara TE, Colonna M, Stappenbeck TS. Adaptive differentiation promotes intestinal villus recovery. Dev Cell 2022; 57:166-179.e6. [PMID: 35016013 PMCID: PMC9092613 DOI: 10.1016/j.devcel.2021.12.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/28/2021] [Accepted: 12/10/2021] [Indexed: 01/26/2023]
Abstract
Loss of differentiated cells to tissue damage is a hallmark of many diseases. In slow-turnover tissues, long-lived differentiated cells can re-enter the cell cycle or transdifferentiate to another cell type to promote repair. Here, we show that in a high-turnover tissue, severe damage to the differentiated compartment induces progenitors to transiently acquire a unique transcriptional and morphological postmitotic state. We highlight this in an acute villus injury model in the mouse intestine, where we identified a population of progenitor-derived cells that covered injured villi. These atrophy-induced villus epithelial cells (aVECs) were enriched for fetal markers but were differentiated and lineage committed. We further established a role for aVECs in maintaining barrier integrity through the activation of yes-associated protein (YAP). Notably, loss of YAP activity led to impaired villus regeneration. Thus, we define a key repair mechanism involving the activation of a fetal-like program during injury-induced differentiation, a process we term "adaptive differentiation."
Collapse
Affiliation(s)
- Takahiro E Ohara
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
9
|
Perez F, Ruera CN, Miculan E, Carasi P, Chirdo FG. Programmed Cell Death in the Small Intestine: Implications for the Pathogenesis of Celiac Disease. Int J Mol Sci 2021; 22:7426. [PMID: 34299046 PMCID: PMC8306608 DOI: 10.3390/ijms22147426] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022] Open
Abstract
The small intestine has a high rate of cell turnover under homeostatic conditions, and this increases further in response to infection or damage. Epithelial cells mostly die by apoptosis, but recent studies indicate that this may also involve pro-inflammatory pathways of programmed cell death, such as pyroptosis and necroptosis. Celiac disease (CD), the most prevalent immune-based enteropathy, is caused by loss of oral tolerance to peptides derived from wheat, rye, and barley in genetically predisposed individuals. Although cytotoxic cells and gluten-specific CD4+ Th1 cells are the central players in the pathology, inflammatory pathways induced by cell death may participate in driving and sustaining the disease through the release of alarmins. In this review, we summarize the recent literature addressing the role of programmed cell death pathways in the small intestine, describing how these mechanisms may contribute to CD and discussing their potential implications.
Collapse
Affiliation(s)
- Federico Perez
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina; (C.N.R.); (E.M.); (P.C.)
| | | | | | | | - Fernando Gabriel Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina; (C.N.R.); (E.M.); (P.C.)
| |
Collapse
|
10
|
Hakim SA, Abd El-Kareem D. Evaluation of crypt apoptotic bodies and apoptotic indices in pediatric celiac disease by routine staining and H2AX immunostaining. Int J Immunopathol Pharmacol 2021; 35:20587384211026791. [PMID: 34137295 PMCID: PMC8216351 DOI: 10.1177/20587384211026791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated disorder with premature apoptosis occurring along the entire crypt-villous axis. H2AX is the end product of the intrinsic apoptotic pathway. This is the first study to assess apoptotic body counts (ABC) by H&E and apoptotic indices (AI) by immunohistochemistry (IHC) in pediatric CD. The aim of the current study was to evaluate ABC in pediatric patients with CD prior to and following institution of a gluten free diet (GFD). Sixty-three pediatric endoscopic duodenal samples were assessed and divided into three groups. A total of 21 samples from treatment naïve CD patients, 21 from the same patients after instituting a GFD, and 21 from non-celiac patients as a control group. Histopathological evaluation of ABC by H&E, and immunohistochemistry assessment of apoptotic indices (AI) by H2AX antibody were performed. The mean maximum ABC and AI were significantly higher in treatment naïve CD than in GFD and control samples. These values were also significantly higher in treatment naïve Marsh 3C (flat) than in Marsh 1, 2, 3A, and 3B (non-flat) CD cases. GFD samples with persistent flat lesions had significantly higher ABC and AI than GFD non-flat cases. ROC analysis of the mean maximum ABC and AI of treatment naïve CD cases had a statistically significant predictive potential for persistent villous atrophy at a cut-off level ⩾6.61 (P = 0.008) and ⩾105.4 (P = 0.003), respectively. Histopathological evaluation of crypt apoptotic bodies could provide predictive potential for continued villous atrophy following GFD.
Collapse
Affiliation(s)
- Sarah Adel Hakim
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Sarah Adel Hakim, Faculty of Medicine, Ain Shams University, Abbasseya Square, 11561 Cairo, Egypt.
| | | |
Collapse
|
11
|
Cell death in the gut epithelium and implications for chronic inflammation. Nat Rev Gastroenterol Hepatol 2020; 17:543-556. [PMID: 32651553 DOI: 10.1038/s41575-020-0326-4] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
The intestinal epithelium has one of the highest rates of cellular turnover in a process that is tightly regulated. As the transit-amplifying progenitors of the intestinal epithelium generate ~300 cells per crypt every day, regulated cell death and sloughing at the apical surface keeps the overall cell number in check. An aberrant increase in the rate of intestinal epithelial cell (IEC) death underlies instances of extensive epithelial erosion, which is characteristic of several intestinal diseases such as inflammatory bowel disease and infectious colitis. Emerging evidence points to a crucial role of necroptosis, autophagy and pyroptosis as important modes of programmed cell death in the intestine in addition to apoptosis. The mode of cell death affects tissue restitution responses and ultimately the long-term risks of intestinal fibrosis and colorectal cancer. A vicious cycle of intestinal barrier breach, misregulated cell death and subsequent inflammation is at the heart of chronic inflammatory and infectious gastrointestinal diseases. This Review discusses the underlying molecular and cellular underpinnings that control programmed cell death in IECs, which emerge during intestinal diseases. Translational aspects of cell death modulation for the development of novel therapeutic alternatives for inflammatory bowel diseases and colorectal cancer are also discussed.
Collapse
|
12
|
Dunne MR, Byrne G, Chirdo FG, Feighery C. Coeliac Disease Pathogenesis: The Uncertainties of a Well-Known Immune Mediated Disorder. Front Immunol 2020; 11:1374. [PMID: 32733456 PMCID: PMC7360848 DOI: 10.3389/fimmu.2020.01374] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Coeliac disease is a common small bowel enteropathy arising in genetically predisposed individuals and caused by ingestion of gluten in the diet. Great advances have been made in understanding the role of the adaptive immune system in response to gluten peptides. Despite detailed knowledge of these adaptive immune mechanisms, the complete series of pathogenic events responsible for development of the tissue lesion remains less certain. This review contributes to the field by discussing additional mechanisms which may also contribute to pathogenesis. These include the production of cytokines such as interleukin-15 by intestinal epithelial cells and local antigen presenting cells as a pivotal event in the disease process. A subset of unconventional T cells called gamma/delta T cells are also persistently expanded in the coeliac disease (CD) small intestinal epithelium and recent analysis has shown that these cells contribute to pathogenic inflammation. Other unconventional T cell subsets may play a local immunoregulatory role and require further study. It has also been suggested that, in addition to activation of pathogenic T helper cells by gluten peptides, other peptides may directly interact with the intestinal mucosa, further contributing to the disease process. We also discuss how myofibroblasts, a major source of tissue transglutaminase and metalloproteases, may play a key role in intestinal tissue remodeling. Contribution of each of these factors to pathogenesis is discussed to enhance our view of this complex disorder and to contribute to a wider understanding of chronic immune-mediated disease.
Collapse
Affiliation(s)
- Margaret R. Dunne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Greg Byrne
- School of Biological & Health Sciences, Technological University, Dublin, Ireland
| | - Fernando G. Chirdo
- Instituto de Estudios Inmunologicos y Fisiopatologicos - IIFP (UNLP-CONICET), National University of La Plata, La Plata, Argentina
| | - Conleth Feighery
- Department of Immunology, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
13
|
Verdile N, Pasquariello R, Scolari M, Scirè G, Brevini TAL, Gandolfi F. A Detailed Study of Rainbow Trout ( Onchorhynchus mykiss) Intestine Revealed That Digestive and Absorptive Functions Are Not Linearly Distributed along Its Length. Animals (Basel) 2020; 10:ani10040745. [PMID: 32344584 PMCID: PMC7223369 DOI: 10.3390/ani10040745] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Aquaculture is the fastest growing food-producing sector due to the increase of fish intended for human consumption. However, aquaculture growth generates concerns, since carnivorous fish are extensively fed using fish-meal and fish-oil. This constitutes a severe limit to the aquaculture industry, questioning its sustainability. Consequently, alternative feeds are continuously searched through extensive in vivo feeding trials. Undoubtedly, to evaluate their impact on the gastrointestinal tract health, detailed knowledge of the intestine morphology and physiology is required. To date, extensive studies have been performed in several livestock species; however, available information on fish is limited nowadays, most importantly because their alimentary canal is able to easily adapt to external stimuli, and their intestinal morphology is affected by external factors. Therefore, it is essential to establish accurate reference values, especially along the productive cycle of animals raised in standardized conditions. Here, we performed a detailed characterization of the epithelial cells lining the intestinal mucosa in rainbow trout along the first year of development. We studied the absorptive and secretory activity as well as its ability to self-renewal. Our results indicate that, in this species, both digestive and absorptive functions are not linearly distributed along the intestinal length. Abstract To increase the sustainability of trout farming, the industry requires alternatives to fish-based meals that do not compromise animal health and growth performances. To develop new feeds, detailed knowledge of intestinal morphology and physiology is required. We performed histological, histochemical, immunohistochemical and morphometric analysis at typical time points of in vivo feeding trials (50, 150 and 500 g). Only minor changes occurred during growth whereas differences characterized two compartments, not linearly distributed along the intestine. The first included the pyloric caeca, the basal part of the complex folds and the villi of the distal intestine. This was characterized by a significantly smaller number of goblet cells with smaller mucus vacuoles, higher proliferation and higher apoptotic rate but a smaller extension of fully differentiated epithelial cells and by the presence of numerous pinocytotic vacuolization. The second compartment was formed by the proximal intestine and the apical part of the posterior intestine complex folds. Here we observed more abundant goblet cells with bigger vacuoles, low proliferation rate, few round apoptotic cells, a more extended area of fully differentiated cells and no pinocytotic vacuoles. Our results suggest that rainbow trout intestine is physiologically arranged to mingle digestive and absorptive functions along its length.
Collapse
Affiliation(s)
- Nicole Verdile
- Department of Agricultural and Environmental Sciences, University of Milan, 20133 Milano, Italy;
- Correspondence: (N.V.); (F.G.); Tel.: +39-02-5031-6449 (N.V.); +39-02-5031-7990 (F.G.)
| | - Rolando Pasquariello
- Department of Agricultural and Environmental Sciences, University of Milan, 20133 Milano, Italy;
| | - Marco Scolari
- Skretting Aquaculture Research Centre, 37100 Verona, Italy; (M.S.); (G.S.)
| | - Giulia Scirè
- Skretting Aquaculture Research Centre, 37100 Verona, Italy; (M.S.); (G.S.)
| | - Tiziana A. L. Brevini
- Department of Health, Animal Science and Food Safety, University of Milan, 20133 Milano, Italy;
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences, University of Milan, 20133 Milano, Italy;
- Correspondence: (N.V.); (F.G.); Tel.: +39-02-5031-6449 (N.V.); +39-02-5031-7990 (F.G.)
| |
Collapse
|
14
|
Lee M, Betman S, Iuga A, Yang HM, Fleming J, Green PHR, Lebwohl B, Lagana SM. An association between crypt apoptotic bodies and mucosal flattening in celiac disease patients exposed to dietary gluten. Diagn Pathol 2019; 14:98. [PMID: 31472694 PMCID: PMC6717634 DOI: 10.1186/s13000-019-0878-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/26/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Celiac disease (CD) is characterized histologically by inflammation and villous atrophy. Villous atrophy is thought to result from a disruption of epithelial cellular proliferation and death. Epithelial cells in intestinal mucosa normally proliferate in the crypts and migrate towards the lumen, eventually dying. Apoptotic bodies in crypts are usually abnormal and are associated with certain disease states. The presence of crypt apoptosis in celiac disease has not been thoroughly examined by routine histologic assessment of crypt apoptotic body count (ABC). METHODS We quantified the ABC in duodenal biopsies from celiac patients before and after initiation of a gluten-free diet (GFD). We examined twenty-three duodenal biopsies from adult patients with celiac disease at diagnosis and following GFD and determined the maximum ABC in 10 consecutive crypts. Fourteen biopsies from heartburn patients served as controls. RESULTS Mean duration between paired biopsies was 2.9 (0.5-8.5) years. Mean maximum ABC in active celiac disease was 5.44 per crypt and decreased to 2.60 with GFD (p = <.0001). The mean maximum ABC in controls was 1.79, lower than both active celiac disease and GFD (p = <.0001 and p = .019 respectively). Flat lesions with total villous atrophy (mean: 6.44) showed a higher ABC compared to non-flat lesions (mean: 4.87); p = .04. CONCLUSIONS Crypt ABC is markedly elevated in active celiac disease and decreases significantly with GFD, however it does not achieve normalcy. Total villous atrophy is associated with a higher ABC than all other lesions. Crypt apoptosis is likely a significant contributor to villous atrophy in celiac disease and can be appreciated by routine histologic examination.
Collapse
Affiliation(s)
- Michael Lee
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, VC14-240A, New York, NY, 10032, USA.
| | - Shane Betman
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, VC14-240A, New York, NY, 10032, USA
| | - Alina Iuga
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, VC14-240A, New York, NY, 10032, USA
| | - Hui-Min Yang
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, VC14-240A, New York, NY, 10032, USA
| | - Jude Fleming
- Internal Medicine, Columbia University Medical Center, New York, NY, USA
| | - Peter H R Green
- Celiac Disease Center, Columbia University Medical Center, New York, NY, USA
| | - Benjamin Lebwohl
- Celiac Disease Center, Columbia University Medical Center, New York, NY, USA
| | - Stephen M Lagana
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, VC14-240A, New York, NY, 10032, USA
| |
Collapse
|
15
|
Oxidative stress, DNA stability and evoked inflammatory signaling in young celiac patients consuming a gluten-free diet. Eur J Nutr 2019; 59:1577-1584. [PMID: 31144026 DOI: 10.1007/s00394-019-02013-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 05/24/2019] [Indexed: 01/01/2023]
Abstract
PURPOSE Celiac disease (CD) is a multifactorial, autoimmune, gluten-sensitive inflammatory disorder of the small intestine. Taking into account the pathogenesis of CD, a strict gluten-free diet (GFD) is the only treatment able to restore epithelium integrity and eliminate complications. The current study was designed to assess whether the use of a GFD is sufficient for maintaining a correct oxidative/antioxidant balance and ameliorating the evoked inflammatory signaling in young patients with CD. METHODS The study covered 80 children, aged between 7 and 18 years, attending the Gastroenterology Service of the Gastroenterology, Hepatology and Child Nutrition Service from the Virgen de las Nieves Hospital in Granada. Children with CD diagnosed were included in the celiac group who followed a strict GFD for 2 years (n = 40) and the control group (n = 40) included healthy children, with negative serological screening. Soluble superoxide dismutase 1 and 2, total antioxidant status, 8-hydroxy-2'-deoxyguanosine, cortisol, melatonin and inflammatory parameters in plasma, 15-F2t-isoprostanes in urine, and DNA breaks in peripheral blood lymphocytes were analysed. RESULTS No differences were found in oxidative stress between CD patients and controls; however, IFN-γ, IL-1α, IP-10 and TNF-β were higher in the CD patients. VEGF was also higher than in the control group. CONCLUSION The GFD in the CD patients is enough to reduce the oxidative stress; however, in the case of the inflammatory signaling, the initial exposure to gluten prior to stablish the GFD is strong enough to induce an inflammatory state which is maintained (even when consuming the GFD); meanwhile the increase in VEGF recorded in the CD group could be a compensatory mechanism to restore the damaged mucosa and duodenal villous atrophy, due to its role in endothelial activation and generation of new functional and stable vascular networks.
Collapse
|
16
|
Martucciello S, Paolella G, Esposito C, Lepretti M, Caputo I. Anti-type 2 transglutaminase antibodies as modulators of type 2 transglutaminase functions: a possible pathological role in celiac disease. Cell Mol Life Sci 2018; 75:4107-4124. [PMID: 30136165 PMCID: PMC11105699 DOI: 10.1007/s00018-018-2902-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/30/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
Abstract
Auto-antibodies to the ubiquitous enzyme type-2 transglutaminase (TG2) are a specific hallmark of celiac disease (CD), a widely diffused, multi-factorial disease, affecting genetically predisposed subjects. In CD an inflammatory response, at the intestinal level, is triggered by diet consumption of gluten-containing cereals. Intestinal mucosa displays various degrees of atrophy and hyperplasia, with consequent global intestinal dysfunction and other relevant extra-intestinal symptoms. Through deamidation of specific glutamines of gluten-derived gliadin peptides, TG2 strongly enhances gliadin immunogenicity. In addition, TG2 cross-linking activity may generate complexes between TG2 itself and gliadin peptides, and these complexes seem to cause the auto-immune response by means of an apten-carrier-like mechanism of antigen presentation. Anti-TG2 antibodies can be early detected in the intestinal mucosa of celiac patients and are also abundantly present into the serum, thus potentially reaching other organs and tissues by blood circulation. Recently, the possible pathogenetic role of auto-antibodies to TG2 in CD has been investigated. Here, we report an overview about the genesis of these antibodies, their specificity, their modulating ability toward TG2 enzymatic or non-enzymatic activities and their biological effects exerted by interacting with extracellular TG2 or with cell-surface TG2. We also discuss the auto-immune response occurring in CD against other TG members (i.e. type 3 and type 6) and analyze the occurrence of anti-TG2 antibodies in other auto-immune CD-related diseases. Data now available let us to suppose that, even if antibodies to TG2 do not represent the triggering molecules in CD, they could be important players in disease progression and manifestations.
Collapse
Affiliation(s)
- Stefania Martucciello
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano (SA), Italy
| | - Gaetana Paolella
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano (SA), Italy
| | - Carla Esposito
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano (SA), Italy
- Interuniversity Centre "European Laboratory for the Investigation of Food-Induced Diseases" (ELFID), University of Salerno, Fisciano (SA), Italy
| | - Marilena Lepretti
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano (SA), Italy
| | - Ivana Caputo
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano (SA), Italy.
- Interuniversity Centre "European Laboratory for the Investigation of Food-Induced Diseases" (ELFID), University of Salerno, Fisciano (SA), Italy.
| |
Collapse
|
17
|
Li Z, Kabir I, Tietelman G, Huan C, Fan J, Worgall T, Jiang XC. Sphingolipid de novo biosynthesis is essential for intestine cell survival and barrier function. Cell Death Dis 2018; 9:173. [PMID: 29415989 PMCID: PMC5833386 DOI: 10.1038/s41419-017-0214-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/13/2017] [Accepted: 12/07/2017] [Indexed: 12/15/2022]
Abstract
Serine palmitoyltransferase (SPT) is the rate-limiting enzyme for sphingolipid biosynthesis. SPT has two major subunits, SPTLC1 and SPTLC2. We previously found that liver Sptlc2 deficiency in early life impairs the development of adherens junctions. Here, we investigated the role of Sptlc2 deficiency in intestine. We treated Sptlc2-Flox/villin-Cre-ERT2 mice with tamoxifen (days 1, 2, and 3) to ablate Sptlc2 specifically in the intestine. At day 6 after tamoxifen treatment, Sptlc2-deficient mice had significantly decreased body weight with concurrent diarrhea and rectal bleeding. The number of goblet cells was reduced in both large and small intestine of Sptlc2-deficient mice compared with controls. Sptlc2 deficiency suppressed the level of mucin2 in the colon and increased circulating lipopolysaccharides, suggesting that SPT activity has a housekeeping function in the intestine. All Sptlc2-deficient mice died 7-10 days after tamoxifen treatment. Notably, supplementation with antibiotics and dexamethasone reduced lethality by 70%. We also found that colon specimens from patients with inflammatory bowel diseases had significantly reduced Sptlc2 expression, SPTLC2 staining, and goblet cell numbers. SPT activity is crucial for intestinal cell survival and barrier function.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA
- Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, NY, 11209, USA
| | - Inamul Kabir
- Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA
| | - Gladys Tietelman
- Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA
| | - Chongmin Huan
- Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA
| | - Jianglin Fan
- Department of Molecular Pathology, University of Yamanashi, Yamanashi, Japan
| | - Tilla Worgall
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA.
- Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, NY, 11209, USA.
| |
Collapse
|
18
|
Piatek-Guziewicz A, Ptak-Belowska A, Przybylska-Felus M, Pasko P, Zagrodzki P, Brzozowski T, Mach T, Zwolinska-Wcislo M. Intestinal parameters of oxidative imbalance in celiac adults with extraintestinal manifestations. World J Gastroenterol 2017; 23:7849-7862. [PMID: 29209126 PMCID: PMC5703914 DOI: 10.3748/wjg.v23.i44.7849] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/03/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate selected intestinal parameters of oxidative stress, and antioxidant capacity in adult celiac disease patients with extraintestinal manifestations. METHODS The study involved 85 adult patients divided into the following subgroups: (1) patients with newly diagnosed celiac disease (CD) (n = 7); (2) celiac patients not adhering to a gluten-free diet (GFD) (n = 22); (3) patients with CD on the GFD (n = 31); and (4) patients with functional disorders of the gastrointestinal tract, serving as controls (n = 25). Celiac patients presented with non-classic symptoms or extraintestinal manifestations. Standard blood tests including serum antioxidant levels (uric acid, bilirubin, and vitamin D), celiac antibody levels, and histopathological status of duodenal biopsy specimens have been determined. The expression of mRNA for tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), interleukin 10 (IL-10), superoxide dismutase (SOD), heat-shock protein 70 (HSP-70), hypoxia-inducible factor 1 (HIF-1α), and BAX in the duodenal mucosa of patients was analyzed by reverse transcriptase-polymerase chain reaction. RESULTS The mean plasma uric acid level in patients with active CD (newly diagnosed and nonadherent patients) and treated celiac patients was significantly higher than in controls (260.17 ± 53.65 vs 190.8 ± 22.98, P < 0.001, and 261.7 ± 51.79 vs 190.8 ± 22.98, P < 0.001, respectively). The mean bilirubin concentration in active and treated celiac patients was significantly lower than in controls (8.23 ± 5.04 vs 10.48 ± 4.08, P < 0.05 and 8.06 ± 3.31 vs 10.48 ± 4.08, P < 0.05, respectively). The mean plasma vitamin D level was significantly lower in active celiac patients than in treated celiac patients and controls (19.37 ± 9.03 vs 25.15 ± 11.2, P < 0.05 and 19.37 ± 9.03 vs 29.67 ± 5.12, P < 0.001, respectively). The expression of TNF-α, IL-10, and HSP-70 mRNAs was significantly elevated in the celiac groups regardless of the diet when compared with controls. Patients on the GFD presented a significantly lower mRNA expression of TNF-α and IL-10 than in newly diagnosed and nonadherent patients (P < 0.05). The expression of SOD mRNA was significantly elevated in celiac patients compared with controls (P < 0.05), with a significant difference between treated and untreated patients (P < 0.05). The expression of HIF-1α mRNA and BAX mRNA was significantly higher in patients with active CD compared with controls and patients on GFD, while no difference was observed between the latter two groups. CONCLUSION Increased intestinal expression of HSP-70 despite GFD indicates that GFD only partially reduced oxidative stress. CD patients exhibited an oxidative imbalance and inflammatory response despite GFD. Uric acid may act as an important antioxidant in CD.
Collapse
Affiliation(s)
| | - Agata Ptak-Belowska
- Department of Physiology, Jagiellonian University Medical College, Cracow 31531, Poland
| | - Magdalena Przybylska-Felus
- Department of Gastroenterology, Hepatology and Infectious Diseases, Jagiellonian University Medical College, Cracow 31531, Poland
| | - Pawel Pasko
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Cracow 30688, Poland
| | - Pawel Zagrodzki
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Cracow 30688, Poland
- Henryk Niewodniczanski Institute of Nuclear Physics, Cracow 31342, Poland
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, Cracow 31531, Poland
| | - Tomasz Mach
- Department of Gastroenterology, Hepatology and Infectious Diseases, Jagiellonian University Medical College, Cracow 31531, Poland
| | - Malgorzata Zwolinska-Wcislo
- Department of Gastroenterology, Hepatology and Infectious Diseases, Jagiellonian University Medical College, Cracow 31531, Poland
| |
Collapse
|
19
|
Epple HJ, Friebel J, Moos V, Troeger H, Krug SM, Allers K, Schinnerling K, Fromm A, Siegmund B, Fromm M, Schulzke JD, Schneider T. Architectural and functional alterations of the small intestinal mucosa in classical Whipple's disease. Mucosal Immunol 2017; 10:1542-1552. [PMID: 28176790 DOI: 10.1038/mi.2017.6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/10/2017] [Indexed: 02/04/2023]
Abstract
Classical Whipple's disease (CWD) affects the gastrointestinal tract and rather elicits regulatory than inflammatory immune reactions. Mechanisms of malabsorption, diarrhea, and systemic immune activation are unknown. We here analyzed mucosal architecture, barrier function, and immune activation as potential diarrheal trigger in specimens from 52 CWD patients. Our data demonstrate villus atrophy and crypt hyperplasia associated with epithelial apoptosis and reduced alkaline phosphatase expression in the duodenum of CWD patients. Electrophysiological and flux experiments revealed increased duodenal permeability to small solutes and macromolecules. Duodenal architecture and permeability ameliorated upon antibiotic treatment. Structural correlates for these alterations were concordant changes of membranous claudin-1, claudin-2, claudin-3, and tricellulin expression. Tumor necrosis factor-α and interleukin-13 were identified as probable mediators of epithelial apoptosis, and altered tight junction expression. Increased serum markers of microbial translocation and their decline following treatment corroborated the biological significance of the mucosal barrier defect. Hence, mucosal immune responses in CWD elicit barrier dysfunction. Diarrhea is caused by loss of absorptive capacity and leak flux of ions and water. Downregulation of tricellulin causes increased permeability to macromolecules and subsequent microbial translocation contributes to systemic inflammation. Thus, therapeutic strategies to reconstitute the mucosal barrier and control inflammation could assist symptomatic control of CWD.
Collapse
Affiliation(s)
- H-J Epple
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - J Friebel
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - V Moos
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - H Troeger
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - S M Krug
- Institute of Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - K Allers
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - K Schinnerling
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - A Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - B Siegmund
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - M Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - J D Schulzke
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany.,Institute of Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - T Schneider
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
20
|
Felli C, Baldassarre A, Masotti A. Intestinal and Circulating MicroRNAs in Coeliac Disease. Int J Mol Sci 2017; 18:ijms18091907. [PMID: 28878141 PMCID: PMC5618556 DOI: 10.3390/ijms18091907] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression at the post-transcriptional level and play a key role in the pathogenesis of autoimmune and gastrointestinal diseases. Previous studies have revealed that miRNAs are dysregulated in intestinal biopsies of patients affected by coeliac disease (CD). Combined bioinformatics analyses of miRNA expression profiles and mRNA target genes as classified by Gene Ontology, are powerful tools to investigate the functional role of miRNAs in coeliac disease. However, little is still known about the function of circulating miRNAs, their expression level compared to tissue miRNAs, and whether the mechanisms of post-transcriptional regulation are the same of tissue miRNAs. In any case, if we assume that a cell-cell communication process has to occur, and that circulating miRNAs are delivered to recipient cells, we can derive useful information by performing target predictions. Interestingly, all of the mRNA targets of dysregulated miRNAs reported in the literature (i.e., miR-31-5p, miR-192, miR-194, miR-449a and miR-638) belong to several important biological processes, such as Wnt signaling, cell proliferation and differentiation, and adherens junction pathways. Although we think that these predictions have to be necessarily confirmed by “wet-lab” data, the miRNAs dysregulated during the development of CD could be potentially involved in the pathogenesis of coeliac disease and their correlation with circulating miRNAs offers new possibilities to use them as disease biomarkers.
Collapse
Affiliation(s)
- Cristina Felli
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, V.le di San Paolo 15, 00146 Rome, Italy.
| | - Antonella Baldassarre
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, V.le di San Paolo 15, 00146 Rome, Italy.
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, V.le di San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
21
|
Parker A, Maclaren OJ, Fletcher AG, Muraro D, Kreuzaler PA, Byrne HM, Maini PK, Watson AJM, Pin C. Cell proliferation within small intestinal crypts is the principal driving force for cell migration on villi. FASEB J 2016; 31:636-649. [PMID: 27811059 PMCID: PMC5241155 DOI: 10.1096/fj.201601002] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/11/2016] [Indexed: 12/05/2022]
Abstract
The functional integrity of the intestinal epithelial barrier relies on tight coordination of cell proliferation and migration, with failure to regulate these processes resulting in disease. It is not known whether cell proliferation is sufficient to drive epithelial cell migration during homoeostatic turnover of the epithelium. Nor is it known precisely how villus cell migration is affected when proliferation is perturbed. Some reports suggest that proliferation and migration may not be related while other studies support a direct relationship. We used established cell-tracking methods based on thymine analog cell labeling and developed tailored mathematical models to quantify cell proliferation and migration under normal conditions and when proliferation is reduced and when it is temporarily halted. We found that epithelial cell migration velocities along the villi are coupled to cell proliferation rates within the crypts in all conditions. Furthermore, halting and resuming proliferation results in the synchronized response of cell migration on the villi. We conclude that cell proliferation within the crypt is the primary force that drives cell migration along the villus. This methodology can be applied to interrogate intestinal epithelial dynamics and characterize situations in which processes involved in cell turnover become uncoupled, including pharmacological treatments and disease models.—Parker, A., Maclaren, O. J., Fletcher, A. G., Muraro, D., Kreuzaler, P. A., Byrne, H. M., Maini, P. K., Watson, A. J. M., Pin, C. Cell proliferation within small intestinal crypts is the principal driving force for cell migration on villi.
Collapse
Affiliation(s)
- Aimee Parker
- Gut Health and Food Safety Research Programme, Institute of Food Research, Norwich, United Kingdom
| | - Oliver J Maclaren
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Alexander G Fletcher
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom.,School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom.,Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Daniele Muraro
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Peter A Kreuzaler
- Department of Biochemistry, University of Cambridge, United Kingdom; and
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | | - Carmen Pin
- Gut Health and Food Safety Research Programme, Institute of Food Research, Norwich, United Kingdom;
| |
Collapse
|
22
|
Cheat S, Pinton P, Cossalter AM, Cognie J, Vilariño M, Callu P, Raymond-Letron I, Oswald IP, Kolf-Clauw M. The mycotoxins deoxynivalenol and nivalenol show in vivo synergism on jejunum enterocytes apoptosis. Food Chem Toxicol 2016; 87:45-54. [DOI: 10.1016/j.fct.2015.11.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/09/2023]
|
23
|
Hematopoietic Stem Cell Transplantation. PATHOLOGY OF TRANSPLANTATION 2016. [PMCID: PMC7124099 DOI: 10.1007/978-3-319-29683-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
24
|
Montén C, Gudjonsdottir AH, Browaldh L, Arnell H, Nilsson S, Agardh D, Naluai ÅT. Genes involved in muscle contractility and nutrient signaling pathways within celiac disease risk loci show differential mRNA expression. BMC MEDICAL GENETICS 2015; 16:44. [PMID: 26123480 PMCID: PMC4630939 DOI: 10.1186/s12881-015-0190-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 06/16/2015] [Indexed: 01/17/2023]
Abstract
Background Risk gene variants for celiac disease, identified in genome-wide linkage and association studies, might influence molecular pathways important for disease development. The aim was to examine expression levels of potential risk genes close to these variants in the small intestine and peripheral blood and also to test if the non-coding variants affect nearby gene expression levels in children with celiac disease. Methods Intestinal biopsy and peripheral blood RNA was isolated from 167 children with celiac disease, 61 with potential celiac disease and 174 disease controls. Transcript levels for 88 target genes, selected from celiac disease risk loci, were analyzed in biopsies of a smaller sample subset by qPCR. Differentially expressed genes (3 from the pilot and 8 previously identified) were further validated in the larger sample collection (n = 402) of both tissues and correlated to nearby celiac disease risk variants. Results All genes were significantly down- or up-regulated in the intestinal mucosa of celiac disease children, NTS being most down-regulated (Fold change 3.6, p < 0.001). In contrast, PPP1R12B isoform C was up-regulated in the celiac disease mucosa (Fold change 1.9, p < 0.001). Allele specific expression of GLS (rs6741418, p = 0.009), INSR (rs7254060, p = 0.003) and NCALD (rs652008, p = 0.005) was also detected in the biopsies. Two genes (APPL2 and NCALD) were differentially expressed in peripheral blood but no allele specific expression was observed in this tissue. Conclusion The differential expression of NTS and PPP1R12B indicate a potential role for smooth muscle contractility and cell proliferation in celiac disease, whereas other genes like GLS, NCALD and INSR suggests involvement of nutrient signaling and energy homeostasis in celiac disease pathogenesis. A disturbance in any of these pathways might contribute to development of childhood celiac disease. Electronic supplementary material The online version of this article (doi:10.1186/s12881-015-0190-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caroline Montén
- Diabetes & Celiac Disease Unit, Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, CRC, 91:10, 202 05, Malmö, Sweden.
| | - Audur H Gudjonsdottir
- Department of Pediatrics, Queen Silvia Children's Hospital, Sahlgrenska Academy, Gothenburg, Sweden.
| | - Lars Browaldh
- Department of Clinical Science & Education, Karolinska Institute Södersjukhuset, Stockholm, Sweden.
| | - Henrik Arnell
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Karolinska University Hospital, Stockholm, Sweden.
| | - Staffan Nilsson
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden.
| | - Daniel Agardh
- Diabetes & Celiac Disease Unit, Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, CRC, 91:10, 202 05, Malmö, Sweden.
| | - Åsa Torinsson Naluai
- Department of Medical & Clinical Genetics, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg, Sweden.
| |
Collapse
|
25
|
Pagliari D, Urgesi R, Frosali S, Riccioni ME, Newton EE, Landolfi R, Pandolfi F, Cianci R. The Interaction among Microbiota, Immunity, and Genetic and Dietary Factors Is the Condicio Sine Qua Non Celiac Disease Can Develop. J Immunol Res 2015; 2015:123653. [PMID: 26090475 PMCID: PMC4451297 DOI: 10.1155/2015/123653] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 10/14/2014] [Indexed: 12/24/2022] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy, triggered by dietary wheat gluten and similar proteins of barley and rye in genetically susceptible individuals. This is a complex disorder involving both environmental and immune-genetic factors. The major genetic risk factor for CD is determined by HLA-DQ genes. Dysfunction of the innate and adaptive immune systems can conceivably cause impairment of mucosal barrier function and development of localized or systemic inflammatory and autoimmune processes. Exposure to gluten is the main environmental trigger responsible for the signs and symptoms of the disease, but exposure to gluten does not fully explain the manifestation of CD. Thus, both genetic determination and environmental exposure to gluten are necessary for the full manifestation of CD; neither of them is sufficient alone. Epidemiological and clinical data suggest that other environmental factors, including infections, alterations in the intestinal microbiota composition, and early feeding practices, might also play a role in disease development. Thus, this interaction is the condicio sine qua non celiac disease can develop. The breakdown of the interaction among microbiota, innate immunity, and genetic and dietary factors leads to disruption of homeostasis and inflammation; and tissue damage occurs. Focusing attention on this interaction and its breakdown may allow a better understanding of the CD pathogenesis and lead to novel translational avenues for preventing and treating this widespread disease.
Collapse
Affiliation(s)
- D. Pagliari
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - R. Urgesi
- Gastroenterology and Digestive Endoscopy Unit, Belcolle Hospital, 01100 Viterbo, Italy
| | - S. Frosali
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - M. E. Riccioni
- Digestive Endoscopy Unit, Catholic University, 00168 Rome, Italy
| | | | - R. Landolfi
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - F. Pandolfi
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| | - R. Cianci
- Institute of Internal Medicine, Catholic University, 00168 Rome, Italy
| |
Collapse
|
26
|
Peng H, Ma J, Bai Y, Lu J, Yu T. MeDiA: Mean Distance Association and Its Applications in Nonlinear Gene Set Analysis. PLoS One 2015; 10:e0124620. [PMID: 25915206 PMCID: PMC4411044 DOI: 10.1371/journal.pone.0124620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 03/17/2015] [Indexed: 11/23/2022] Open
Abstract
Probabilistic association discovery aims at identifying the association between random vectors, regardless of number of variables involved or linear/nonlinear functional forms. Recently, applications in high-dimensional data have generated rising interest in probabilistic association discovery. We developed a framework based on functions on the observation graph, named MeDiA (Mean Distance Association). We generalize its property to a group of functions on the observation graph. The group of functions encapsulates major existing methods in association discovery, e.g. mutual information and Brownian Covariance, and can be expanded to more complicated forms. We conducted numerical comparison of the statistical power of related methods under multiple scenarios. We further demonstrated the application of MeDiA as a method of gene set analysis that captures a broader range of responses than traditional gene set analysis methods.
Collapse
Affiliation(s)
- Hesen Peng
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, United States of America
| | - Junjie Ma
- Department of Hematology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Yun Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine, Suwanee, Georgia, United States of America
| | - Jianwei Lu
- School of Software Engineering, Tongji University, Shanghai, China
- Advanced Institute of Translational Medicine, Tongji University, Shanghai, China
| | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
27
|
Stuckey BGA, Sallie R. Alendronate-induced unmasking or deterioration of coeliac disease: a case series. Osteoporos Int 2015; 26:411-4. [PMID: 25349054 DOI: 10.1007/s00198-014-2942-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/16/2014] [Indexed: 10/24/2022]
Affiliation(s)
- B G A Stuckey
- Keogh Institute for Medical Research, 3rd Floor, A Block, QEII Medical Centre, Nedlands, Western Australia, Australia,
| | | |
Collapse
|
28
|
Hematopoietic stem cell transplantation: graft versus host disease and pathology of gastrointestinal tract, liver, and lung. Adv Anat Pathol 2014; 21:301-20. [PMID: 25105933 DOI: 10.1097/pap.0000000000000032] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hematopoietic stem cell transplantation (HCT), formerly known as bone marrow transplantation, is an integral part of treatment for many hematological malignancies. HCT is associated with several complications and comorbidities with differential effects on a wide spectrum of organs and tissues. We present an update on HCT-associated complications such as graft versus host disease (GVHD) and infection, with focus on the surgical pathology of the gastrointestinal (GI) tract, liver, and lung. Although the grading system for GI tract acute GVHD was proposed 40 years ago, recent studies have shed light on minimal histologic criteria for diagnosis of GVHD, as well as its differential diagnosis, including histologic effects of various medications. GI dysfunction in autologous transplant recipients is increasingly appreciated and patients are often biopsied. Acute liver injury in HCT is often due to sinusoidal obstruction syndrome (previously known as venoocclusive disease), or acute GVHD. Liver dysfunction at later time posttransplantation may be associated with acute or chronic GVHD, iron overload, or other causes of hepatitis. Lung injury in HCT is multifactorial, and it remains crucially important to diagnose and treat pulmonary infections. The pulmonary biopsy yields clinically unsuspected diagnoses in the majority of cases and its utilization is likely to increase. The pathology of the skin and kidney in HCT patients are detailed in accompanying articles.
Collapse
|
29
|
Mazzarella G, Bergamo P, Maurano F, Luongo D, Rotondi Aufiero V, Bozzella G, Palmieri G, Troncone R, Auricchio S, David C, Rossi M. Gliadin intake alters the small intestinal mucosa in indomethacin-treated HLA-DQ8 transgenic mice. Am J Physiol Gastrointest Liver Physiol 2014; 307:G302-G312. [PMID: 24924747 DOI: 10.1152/ajpgi.00002.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Celiac disease (CD) is an enteropathy caused by the ingestion of wheat gluten in genetically susceptible individuals. A complete understanding of the pathogenic mechanisms in CD has been hindered because of the lack of adequate in vivo models. In the present study, we explored the events after the intragastric administration of gliadin and of the albumin/globulin fraction from wheat in human leukocyte antigen-DQ8 transgenic mice (DQ8 mice) treated with indomethacin, an inhibitor of cyclooxygenases (COXs). After 10 days of treatment, mice showed a significant reduction of villus height, increased crypt depth, increased number of lamina propria-activated macrophages, and high basal interferon-γ secretion in mesenteric lymph nodes, all of which were specifically related to gliadin intake, whereas the albumin/globulin fraction of wheat was unable to induce similar changes. Cotreatment with NS-398, a specific inhibitor of COX-2, also induced the intestinal lesion. Enteropathy onset was further characterized by high levels of oxidative stress markers, similar to CD. Biochemical assessment of the small intestine revealed the specific activation of matrix metalloproteinases 2 and 9, high caspase-3 activity, and a significant increase of tissue transglutaminase protein levels associated with the intestinal lesion. Notably, after 30 days of treatment, enteropathic mice developed serum antibodies toward gliadin (IgA) and tissue transglutaminase (IgG). We concluded that gliadin intake in combination with COX inhibition caused a basal inflammatory status and an oxidative stress condition in the small intestine of DQ8 mice, thus triggering the mucosal lesion and, subsequently, an antigen-specific immunity.
Collapse
Affiliation(s)
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | | | - Gianna Palmieri
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Riccardo Troncone
- European Laboratory for Investigation of Food Induced Diseases and Department of Pediatrics, University "Federico II" of Naples, Naples, Italy; and
| | - Salvatore Auricchio
- European Laboratory for Investigation of Food Induced Diseases and Department of Pediatrics, University "Federico II" of Naples, Naples, Italy; and
| | - Chella David
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy;
| |
Collapse
|
30
|
Ashour DS, Othman AA, Radi DA. Insights into regulatory molecules of intestinal epithelial cell turnover during experimental infection by Heterophyes heterophyes. Exp Parasitol 2014; 143:48-54. [PMID: 24852217 DOI: 10.1016/j.exppara.2014.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 04/27/2014] [Accepted: 05/11/2014] [Indexed: 12/24/2022]
Abstract
Heterophyiasis is an intestinal disease that remains endemic in many parts of the world, particularly the Nile Delta of Egypt and Southeast Asia, yet the populations at risk of infection expand throughout the world. The main histopathological feature of infection is villous atrophy, but the underlying factors are not well understood. Apoptosis of the villous epithelial cells was previously reported to be enhanced during intestinal parasitic infections; however, the role of Heterophyes heterophyes on enterocyte apoptosis was to be explored. Therefore, intestinal sections from mice experimentally infected with H. heterophyes were studied histopathologically and immunohistochemically for caspase-3 and NF-κB and compared to non-infected control mice. Atrophic villi covered by poorly differentiated epithelial cells were observed in the 2nd week post-infection. Also, we noted marked hyperplasia of the intestinal crypts with abundant inflammatory cellular infiltrate in the lamina propria, as well as apoptosis of cells lining the intestinal villi. Both caspase-3 and NF-κB showed positive staining in the intestinal epithelial cells with varying grades of intensity over the length of infection. Caspase-3 expression rose at the 2nd week p.i. then decreased over time, whereas NF-κB expression showed progressive increase throughout the weeks of infection. In conclusion, caspase-3 activation may be an important factor in the apoptotic pathway in early heterophyiasis, and, on the other hand, NF-κB seems to play a role in protecting the intestinal cells from excessive apoptosis. These observations may help open new avenues for tissue protective therapies that avoid or control the deleterious processes of apoptosis in various inflammatory conditions.
Collapse
Affiliation(s)
- Dalia S Ashour
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ahmad A Othman
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Dina A Radi
- Department of Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
31
|
Leite CAC, Fagundes-Neto U, Haapalainen EF. Evaluation of the ultrastructure of the small intestine of hiv infected children by transmission and scanning electronic microscopy. ARQUIVOS DE GASTROENTEROLOGIA 2013; 50:70-7. [PMID: 23657310 DOI: 10.1590/s0004-28032013000100013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 10/30/2012] [Indexed: 02/14/2023]
Abstract
OBJECTIVES To describe HIV children's small intestinal ultrastructural findings. METHODS Descriptive, observational study of small intestine biopsies performed between August 1994 and May 1995 at São Paulo, SP, Brazil. This material pertained to 11 HIV infected children and was stored in a laboratory in paraffin blocks. Scanning and transmission electronic microscopy were used to view those intestine samples and ultrastructural findings were described by analyzing digitalized photos of this material. Ethical Committee approval was obtained. RESULTS In most samples scanning microscopy showed various degrees of shortening and decreasing number of microvilli and also completes effacements in some areas. Derangement of the enterocytes was seen frequently and sometimes cells well defined borders limits seemed to be loosened. In some areas a mucous-fibrin like membrane with variable thickness and extension appeared to partially or totally coat the epithelial surface. Fat drops were present in the intestinal lumen in various samples and a bacterium morphologically resembling bacilli was seen in two occasions. Scanning microscopy confirmed transmission microscopy microvilli findings and also showed little "tufts" of those structures. In addition, it showed an increased number of vacuoles and multivesicular bodies inside various enterocytes, an increased presence of intraepithelial lymphocytes, mitochondrial vacuolization and basement membrane enlargement in the majority of samples analyzed. However, some samples exhibited normal aspect. CONCLUSIONS Our study showed the common occurrence of various important intestinal ultrastructural alterations with variable degrees among HIV infected children, some of them in our knowledge not described before.
Collapse
|
32
|
Allegretti YL, Bondar C, Guzman L, Cueto Rua E, Chopita N, Fuertes M, Zwirner NW, Chirdo FG. Broad MICA/B expression in the small bowel mucosa: a link between cellular stress and celiac disease. PLoS One 2013; 8:e73658. [PMID: 24058482 PMCID: PMC3772809 DOI: 10.1371/journal.pone.0073658] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 07/19/2013] [Indexed: 01/15/2023] Open
Abstract
The MICA/B genes (MHC class I chain related genes A and B) encode for non conventional class I HLA molecules which have no role in antigen presentation. MICA/B are up-regulated by different stress conditions such as heat-shock, oxidative stress, neoplasic transformation and viral infection. Particularly, MICA/B are expressed in enterocytes where they can mediate enterocyte apoptosis when recognised by the activating NKG2D receptor present on intraepithelial lymphocytes. This mechanism was suggested to play a major pathogenic role in active celiac disease (CD). Due to the importance of MICA/B in CD pathogenesis we studied their expression in duodenal tissue from CD patients. By immunofluorescence confocal microscopy and flow cytometry we established that MICA/B was mainly intracellularly located in enterocytes. In addition, we identified MICA/B+ T cells in both the intraepithelial and lamina propria compartments. We also found MICA/B+ B cells, plasma cells and some macrophages in the lamina propria. The pattern of MICA/B staining in mucosal tissue in severe enteropathy was similar to that found in in vitro models of cellular stress. In such models, MICA/B were located in stress granules that are associated to the oxidative and ER stress response observed in active CD enteropathy. Our results suggest that expression of MICA/B in the intestinal mucosa of CD patients is linked to disregulation of mucosa homeostasis in which the stress response plays an active role.
Collapse
Affiliation(s)
- Yessica L. Allegretti
- Laboratorio de Investigación en el Sistema Inmune – LISIN, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Constanza Bondar
- Laboratorio de Investigación en el Sistema Inmune – LISIN, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Luciana Guzman
- Servicio de Gastroenterología, Hospital de Niños “Sor María Ludovica,” La Plata, Argentina
| | - Eduardo Cueto Rua
- Servicio de Gastroenterología, Hospital de Niños “Sor María Ludovica,” La Plata, Argentina
| | - Nestor Chopita
- Servicio de Gastroenterología, Hospital San Martin La Plata, La Plata, Argentina
| | - Mercedes Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Norberto W. Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernando G. Chirdo
- Laboratorio de Investigación en el Sistema Inmune – LISIN, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
- * E-mail:
| |
Collapse
|
33
|
Shalimar DM, Das P, Sreenivas V, Gupta SD, Panda SK, Makharia GK. Mechanism of villous atrophy in celiac disease: role of apoptosis and epithelial regeneration. Arch Pathol Lab Med 2013; 137:1262-1269. [PMID: 23991739 DOI: 10.5858/arpa.2012-0354-oa] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
CONTEXT The data on status of apoptosis in patients with celiac disease are conflicting. Furthermore, complex interaction between intrinsic and common apoptotic pathways, apoptotic inhibitors, and epithelial cell proliferation is largely unclear for patients with celiac disease. OBJECTIVES To determine the role of apoptosis and epithelial cell regeneration in celiac disease. DESIGN Twenty-five treatment-naïve patients with celiac disease and 6 patients with functional dyspepsia, as controls, were included and duodenal biopsy specimens from all were subjected to immunohistochemistry with markers of intrinsic apoptotic pathway (AIF, H2AX, p53), common pathway (CC3, M30), apoptotic inhibitors (XIAP, Bcl2), and epithelial proliferation (Ki-67). Apoptotic and proliferation indices were calculated. RESULTS Expression of end-apoptotic products, that is, H2AX in the cell nuclei (P = .01) and M30 in the cell cytoplasm (P < .01), was significantly upregulated in celiac disease in comparison to controls. Cleaved caspase-3 was also upregulated in villous cytoplasm in celiac disease. Apoptotic inhibitor Bcl2 was significantly down-regulated in celiac disease in comparison to controls. In addition, Ki-67 proliferation index was upregulated both in the crypts and villous mucosal epithelium in comparison to the crypts of the controls. CONCLUSIONS Treatment-naïve patients with celiac disease have significantly higher level of apoptosis that involves both the common and intrinsic apoptotic pathways. Increased apoptosis and unequaled cell regeneration in crypts probably results in villous atrophy. Down-regulation of apoptotic inhibitors in treatment-naïve celiac disease imparts an additional pro-apoptotic effect.
Collapse
Affiliation(s)
- D M Shalimar
- Department of Gastroenterology and Human Nutrition, the All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | | | |
Collapse
|
34
|
Adriaanse MPM, Tack GJ, Passos VL, Damoiseaux JGMC, Schreurs MWJ, van Wijck K, Riedl RG, Masclee AAM, Buurman WA, Mulder CJJ, Vreugdenhil ACE. Serum I-FABP as marker for enterocyte damage in coeliac disease and its relation to villous atrophy and circulating autoantibodies. Aliment Pharmacol Ther 2013; 37:482-90. [PMID: 23289539 DOI: 10.1111/apt.12194] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 07/19/2012] [Accepted: 12/07/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Enterocyte damage is the hallmark of coeliac disease (CD) resulting in malabsorption. Little is known about the recovery of enterocyte damage and its clinical consequences. Serum intestinal fatty acid binding protein (I-FABP) is a sensitive marker to study enterocyte damage. AIMS To evaluate the severity of enterocyte damage in adult-onset CD and its course upon a gluten-free diet (GFD). Furthermore, the correlation among enterocyte damage, CD autoantibodies and histological abnormalities during the course of disease is studied. METHODS Serum I-FABP levels were determined in 96 biopsy-proven adult CD patients and in 69 patients repeatedly upon a GFD. A total of 141 individuals with normal antitissue transglutaminase antibody (IgA-tTG) levels served as controls. I-FABP levels were related to the degree of villous atrophy (Marsh grade) and IgA-tTG. RESULTS I-FABP levels were elevated in untreated CD (median 691 pg/mL) compared with controls (median 178 pg/mL, P < 0.001) and correlated with Marsh grade (r = 0.265, P < 0.05) and IgA-tTG (r = 0.403, P < 0.01). Upon a GFD serum levels decreased significantly, however, not within the range observed in controls, despite the common observed normalisation of IgA-tTG levels and Marsh grade. CD patients with elevated I-FABP levels nonresponding to GFD showed persistent histological abnormalities. CONCLUSIONS Enterocyte damage assessed by serum I-FABP correlates with the severity of villous atrophy in coeliac disease at the time of diagnosis. Although enterocyte damage improves upon treatment, substantial enterocyte damage persists despite absence of villous atrophy and low IgA-tTG levels in the majority of cases. Elevated I-FABP levels nonresponding to gluten-free diet are indicative of histological abnormalities and warrant further evaluation.
Collapse
Affiliation(s)
- M P M Adriaanse
- Department of Paediatrics & Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Byrne G, Freeley M, Feighery C, Whelan A, Long A. Protein kinase C delta is a substrate of tissue transglutaminase and a novel autoantigen in coeliac disease. Clin Immunol 2013; 147:1-8. [PMID: 23454274 DOI: 10.1016/j.clim.2013.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/13/2012] [Accepted: 01/17/2013] [Indexed: 12/30/2022]
Abstract
Post-translational modification of proteins by deamidation or transamidation by tissue transglutaminase (tTG) has been suggested as a possible mechanism for the development of autoimmunity. Sequence analysis of protein kinase C delta (PKCδ) identified an amino acid motif that suggested the possibility that PKCδ was a glutamine substrate of tTG and MALDI-TOF analysis of synthesised peptides from PKCδ proved that this was the case. Polymerisation experiments using recombinant tTG and biotinylated hexapeptide substrate incorporation assays demonstrated that PKCδ is a substrate for tTG-mediated transamidation. Elevated levels of anti-PKCδ antibodies were detected in sera from patients with coeliac disease (p<0.0001) but not from patients with other autoimmune disorders. These data suggest that a subset of patients with coeliac disease produce autoantibodies against PKCδ and that this response may stem from a tTG-PKCδ substrate interaction.
Collapse
Affiliation(s)
- Greg Byrne
- School of Biological Sciences, Dublin Institute of Technology, Dublin 8, Ireland.
| | - Michael Freeley
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Con Feighery
- Department of Immunology, St. James's Hospital & Trinity College Dublin, Dublin, Ireland
| | - Alex Whelan
- Department of Immunology, St. James's Hospital & Trinity College Dublin, Dublin, Ireland
| | - Aideen Long
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| |
Collapse
|
36
|
|
37
|
Shalimar, Das P, Sreenivas V, Datta Gupta S, Panda SK, Makharia GK. Effect of addition of short course of prednisolone to gluten-free diet on mucosal epithelial cell regeneration and apoptosis in celiac disease: a pilot randomized controlled trial. Dig Dis Sci 2012; 57:3116-3125. [PMID: 22752636 DOI: 10.1007/s10620-012-2294-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 06/11/2012] [Indexed: 01/08/2023]
Abstract
BACKGROUND Identification of adjuvant treatment is necessary for rapid and effective treatment in patients with celiac disease. In a pilot randomized controlled trial, the effect of prednisolone on enterocyte apoptosis and regeneration in celiac disease was investigated. PATIENTS AND METHODS Thirty-three treatment-naïve patients with celiac disease were randomized to either gluten-free diet (GFD, n = 17) or GFD + prednisolone (1 mg/kg for 4 weeks, n = 16). Duodenal biopsies were taken at baseline and at 4 and 8 weeks posttreatment. Six patients with functional dyspepsia were recruited as controls. All these biopsies were stained for markers of intrinsic apoptotic pathway (AIF, H2AX, p53), common apoptotic pathway (CC3, M30), apoptotic inhibitors (XIAP, Bcl2), and epithelial proliferation (Ki-67). Apoptotic (AI) and proliferation indices (PI) were compared. RESULTS At baseline duodenal biopsies, the end apoptotic products H2AX and M30 were significantly increased. In comparison with those treated with GFD alone, after 4 weeks of GFD + prednisolone treatment, some markers of both intrinsic and common apoptotic pathways showed rapid decline. After prednisolone withdrawal, there was overexpression of H2AX, CC3, and p53 in the latter group. In comparison with those treated with only GFD, patients treated with prednisolone showed suppression of mucosal PI, which started rising again after withdrawal of prednisolone. CONCLUSIONS Apoptosis takes place in mucosal epithelium in celiac disease. Addition of short course of prednisolone suppresses apoptosis rapidly. However, it also suppresses epithelial regeneration; hence, if used, it should be withdrawn after an initial short course.
Collapse
Affiliation(s)
- Shalimar
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | | | | | | | | | | |
Collapse
|
38
|
Schumann M, Kamel S, Pahlitzsch ML, Lebenheim L, May C, Krauss M, Hummel M, Daum S, Fromm M, Schulzke JD. Defective tight junctions in refractory celiac disease. Ann N Y Acad Sci 2012; 1258:43-51. [PMID: 22731714 DOI: 10.1111/j.1749-6632.2012.06565.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In celiac disease, the gut-associated immune system is activated in response to the ingestion of gluten, causing an atrophy of the small intestinal mucosa. Although this condition is, in most cases, responsive to a gluten-free diet, celiac disease refractory to treatment occurs in a small percentage of celiacs. An epithelial barrier defect is known to be an integral part of celiac pathophysiology. However, the mucosa in refractory celiac disease underlies a constant inflammatory process. The epithelial barrier has not been addressed in this condition so far. Herein, the tight junction-associated barrier in refractory celiac disease is investigated functionally and structurally. Although normally expressed in celiac disease, claudin-4 is shown to be downregulated in refractory cases, presumably by two mechanisms, reduced protein expression and increased claudin endocytosis. Furthermore, the tightening claudin-5 is downregulated and the pore-forming claudin-2 is upregulated.
Collapse
Affiliation(s)
- Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Actin reorganization as the molecular basis for the regulation of apoptosis in gastrointestinal epithelial cells. Cell Death Differ 2012; 19:1514-24. [PMID: 22421965 DOI: 10.1038/cdd.2012.28] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The gastrointestinal (GI) epithelium is a rapidly renewing tissue in which apoptosis represents part of the overall homeostatic process. Regulation of apoptosis in the GI epithelium is complex with a precise relationship between cell position and apoptosis. Apoptosis occurs spontaneously and in response to radiation and cytotoxic drugs at the base of the crypts. By contrast, the villus epithelial cells are extremely resistant to apoptosis. The molecular mechanism underlying this loss of function of villus epithelial cells to undergo apoptosis shortly after their exit from the crypt is unknown. In this study we demonstrate for the first time, that deletion of two homologous actin-binding proteins, villin and gelsolin renders villus epithelial cells extremely sensitive to apoptosis. Ultrastructural analysis of the villin-gelsolin(-/-) double-knockout mice shows an abnormal accumulation of damaged mitochondria demonstrating that villin and gelsolin function on an early step in the apoptotic signaling at the level of the mitochondria. A characterization of functional and ligand-binding mutants demonstrate that regulated changes in actin dynamics determined by the actin severing activities of villin and gelsolin are required to maintain cellular homeostasis. Our study provides a molecular basis for the regulation of apoptosis in the GI epithelium and identifies cell biological mechanisms that couple changes in actin dynamics to apoptotic cell death.
Collapse
|
40
|
Olivares M, Laparra M, Sanz Y. Influence of Bifidobacterium longum CECT 7347 and gliadin peptides on intestinal epithelial cell proteome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:7666-7671. [PMID: 21651295 DOI: 10.1021/jf201212m] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Celiac disease is an enteropathy caused by an abnormal immune response to cereal gluten proteins (gliadin). To unravel the possible role of the interactions between gliadin peptides and specific intestinal bacteria, the response of intestinal epithelial (Caco-2) cells to gliadin subjected to gastrointestinal digestion in the presence or absence of Bifidobacterium longum CECT 7347 has been studied. Changes in the proteome of Caco-2 cells were determined by 2DE and MALDI-TOF. Gliadins digested without B. longum altered the expression of a higher number of proteins than in the presence of the bacterium (21 versus 9), and these proteins were involved in disorganization of cell cytoskeleton, inflammation, and apoptosis. Gliadins digested in the presence of the bacterium influenced the production of proteins involved in calcium homeostasis and cell survival and function. Therefore, B. longum CECT 7347 might ameliorate gliadin toxicity and modify the responses of intestinal epithelial cells to the gliadin challenge.
Collapse
Affiliation(s)
- Marta Olivares
- Instituto de Agroquímica y Tecnología de los Alimentos, Spanish National Research Council, Valencia, Spain
| | | | | |
Collapse
|
41
|
Laparra JM, Sanz Y. Bifidobacteria inhibit the inflammatory response induced by gliadins in intestinal epithelial cells via modifications of toxic peptide generation during digestion. J Cell Biochem 2010; 109:801-7. [PMID: 20052669 DOI: 10.1002/jcb.22459] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Celiac disease (CD) is a chronic enteropathy triggered by intake of gliadin, the toxic component of gluten. This study aims at evaluating the capacity of different Bifidobacterium strains to counteract the inflammatory effects of gliadin-derived peptides in intestinal epithelial (Caco-2) cells. A commercial extract of several gliadin (Gld) types (alpha, beta, gamma, [symbol: see text] ) was subjected to in vitro gastrointestinal digestion (pepsin at pH 3, pancreatin-bile at pH 6), inoculated or not with cell suspensions (10(8) colony forming units/ml) of either B. animalis IATA-A2, B. longum IATA-ES1, or B. bifidum IATA-ES2, in a bicameral system. The generated gliadin-derived peptides were identified by reverse phase-HPLC-ESI-MS/MS. Caco-2 cell cultures were exposed to the different gliadin peptide digestions (0.25 mg protein/ml), and the mRNA expression of nuclear factor kappa-B (NF-kappaB), tumor necrosis factor alpha (TNF-alpha), and chemokine CXCR3 receptor were analyzed by semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) in stimulated cells. The production of the pro-inflammatory markers NF-kappaB p50, TNF-alpha, and IL-1beta (interleukine 1beta) by Caco-2 cells was also determined by ELISA. The peptides from gliadin digestions inoculated with bifidobacteria did not exhibit the toxic amino acid sequences identified in those noninoculated (alpha/beta-Gld [158-164] and alpha/beta-Gld [122-141]). The RT-PCR analysis evidenced a down-regulation in mRNA expression of pro-inflammatory biomarkers. Consistent with these results the production of NF-kappaB, TNF-alpha, and IL-1beta was reduced (18.2-22.4%, 28.0-64.8%, and abolished, respectively) in cell cultures exposed to gliadin digestions inoculated with bifidobacteria. Therefore, bifidobacteria change the gliadin-derived peptide pattern and, thereby, attenuate their pro-inflammatory effects on Caco-2 cells.
Collapse
Affiliation(s)
- J M Laparra
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Burjassot (Valencia), Spain
| | | |
Collapse
|
42
|
Szebeni B, Vannay A, Sziksz E, Prókai A, Cseh A, Veres G, Dezsofi A, Gyorffy H, Szabó IRK, Arató A. Increased expression of serum- and glucocorticoid-regulated kinase-1 in the duodenal mucosa of children with coeliac disease. J Pediatr Gastroenterol Nutr 2010; 50:147-153. [PMID: 19966577 DOI: 10.1097/mpg.0b013e3181b47608] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Enterocyte apoptosis induced by activated intraepithelial lymphocytes is increased in coeliac disease (CD). Serum- and glucocorticoid-regulated kinase-1 (SGK1) is a serine/threonine protein kinase that may inhibit apoptosis and compensate for the excessive death of surface epithelial cells. The significance of SGK1 in CD is elusive so far. The aim of this study was to characterise the expression and localisation of SGK1 in duodenal biopsy samples taken from children with untreated CD, children with treated CD, and controls. PATIENTS AND METHODS Duodenal biopsy specimens were collected from 16 children with untreated CD, 9 children with treated CD, and 10 controls. The mRNA expression of SGK1 was determined by real-time reverse transcription-polymerase chain reaction. SGK1 and phosphorylated (P)-SGK1 protein levels and their localisation were determined by Western blot and immunofluorescent staining, respectively. RESULTS We found increased SGK1-mRNA expression as well as higher SGK1 and P-SGK1 protein levels in the duodenal mucosa of children with untreated CD compared with controls. In the duodenal mucosa of children with treated CD, SGK1-mRNA expression was decreased and SGK1 and P-SGK1 protein levels were lower than in untreated CD. SGK1 and P-SGK1 staining intensity was stronger in duodenal villous enterocytes of children with untreated CD compared with treated CD. CONCLUSIONS Our results of increased expression of SGK1 in untreated CD may suggest its contribution to the enterocyte survival in this disease.
Collapse
Affiliation(s)
- Beáta Szebeni
- Research Group for Pediatrics and Nephrology, Semmelweis University and Hungarian Academy of Sciences, Bókay J. u. 53-54, Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hwang JB, Kim SP, Kang YN, Lee SR, Suh SI, Kwon TK. Apoptosis and upregulation of TNF-α and TRAIL receptor 1 (DR4) in the pathogenesis of food protein-induced enterocolitis syndrome. KOREAN JOURNAL OF PEDIATRICS 2010. [DOI: 10.3345/kjp.2010.53.4.525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Jin-Bok Hwang
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| | - Sang Pyo Kim
- Department of Pathology, Keimyung University School of Medicine, Daegu, Korea
| | - Yu Na Kang
- Department of Pathology, Keimyung University School of Medicine, Daegu, Korea
| | - Seong-Ryong Lee
- Institute for Medical Science, Keimyung University School of Medicine, Daegu, Korea
| | - Seong-Il Suh
- Institute for Medical Science, Keimyung University School of Medicine, Daegu, Korea
| | - Taeg Kyu Kwon
- Institute for Medical Science, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
44
|
Gao C, Wang AY. Significance of increased apoptosis and Bax expression in human small intestinal adenocarcinoma. J Histochem Cytochem 2009; 57:1139-48. [PMID: 19729672 DOI: 10.1369/jhc.2009.954446] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human small intestine accounts for 75% of the gastrointestinal (GI) length but for only 1-5% of GI tumors. The reason remains as yet unclearly understood. Our study was designed to examine whether increased apoptosis and expression of related genes/proteins, especially those of the Bcl-2 family, contribute to this difference. For this purpose, 77 samples from patients were examined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and immunohistochemistry, including 40 cases from normal small intestine (jejunum), 7 cases from jejunum and ileum adenocarcinomas, and 30 cases from normal colon. The results showed that a significantly higher level of enterocyte apoptosis was observed in normal small intestine compared with small intestinal adenocarcinomas and normal colon (median of apoptotic index, 15.2% vs 0.1% and 1.6%, p<0.01). A similar pattern was observed for Bax (expression-positive, 77.5% vs 28.6% and 53.3%, p<0.05) but not for Bcl-2 (42.5% vs 42.9% and 46.7%, p>0.05) or Bax/Bcl-2 ratio (percent of samples having a ratio > or =1, 45.0% vs 14.3% and 36.7%, p>0.05). In conclusion, increased apoptosis and expression of Bax, not Bcl-2 or the Bax/Bcl-2 ratio, may play some role in the relatively lower incidence of human small intestinal carcinomas. However, more studies are required for a better understanding of these changes.
Collapse
Affiliation(s)
- Chun Gao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | | |
Collapse
|
45
|
Turan A, Gill R, Dudeja PK, Mohan H, Mahmood A. Effect of fat feeding on pro-oxidant and anti-oxidant enzyme systems in rat intestine: possible role in the turnover of enterocytes. Dig Dis Sci 2009; 54:1229-36. [PMID: 18989782 PMCID: PMC2679095 DOI: 10.1007/s10620-008-0490-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Accepted: 08/22/2008] [Indexed: 12/31/2022]
Abstract
Immature epithelial cells generated in the crypt base undergo differentiation while progressing to the villus tip, where the cells upon apoptosis are detached from the underlying muscular tissue. We previously reported that lipid peroxidation might be involved in the turnover of enterocytes across the crypt-villus axis in rat intestine (Dig Dis Sci 52:1840-1844, 2007). To examine whether long-term feeding of fat with different fatty-acid composition influences this process, in the present study we investigated the effect of feeding fish oil (n - 3) and corn oil (n - 6) polyunsaturated fatty acids on lipid per-oxidation and anti-oxidant systems in different epithelial cell fractions isolated in rat intestine. Feeding fish oil or corn oil markedly enhanced lipid per-oxidation levels of enterocytes throughout villus height compared with control, but there was no difference in the distribution profile of pro- and anti-oxidant enzyme systems and lipid per-oxidation across the crypt-villus axis under these conditions. Analysis of lipid peroxidation levels in different cell fractions revealed that the thiobarbituric acid reactive substance were 9- to 11-fold higher at the villus tip compared with at the crypt base. The activities of glutathione reductase and glutathione-S-transferase were 2- to 5-fold higher in villus tip compared to the crypt region. However, the activities of superoxide dismutase and catalase were 6- to 8-fold high at the crypt base compared with at villus tip cells. Immunocytolocalization of superoxide dismutase showed high staining in crypt base compared with that in villus, tip cells. These findings further suggest that generation of reactive oxygen species in enterocytes across the crypt-villus axis may be involved in turnover of enterocytes across the crypt-villus unit in rat intestine.
Collapse
Affiliation(s)
- Aasma Turan
- Department of Biochemistry, Panjab University, Chandigarh 160014, India, e-mail:
| | - Ravinder Gill
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Pradeep K. Dudeja
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Harsh Mohan
- Department of Pathology, Government Medical College, Chandigarh 160032, India
| | - Akhtar Mahmood
- Department of Biochemistry, Panjab University, Chandigarh 160014, India, e-mail:
| |
Collapse
|
46
|
Vidal C, Borg J, Xuereb-Anastasi A, Scerri CA. Variants within protectin (CD59) and CD44 genes linked to an inherited haplotype in a family with coeliac disease. TISSUE ANTIGENS 2009; 73:225-235. [PMID: 19254252 DOI: 10.1111/j.1399-0039.2008.01193.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Coeliac disease (CD) is an autoimmune disorder characterised by inflammation, villous atrophy and hyperplasia of the small intestinal mucosa that affects genetically susceptible individuals. A genome-wide scan was performed in 17 family members with high incidence of CD. Highest nonparametric linkage (NPL) and logarithm of odds (LOD) scores were of 6.21 (P = 0.0107) and 2.57, respectively, to a region on chromosome 11p13-12. Following fine mapping, NPL and LOD scores did not change, but the linkage interval on chromosome 11 was narrowed to a region that is approximately 50.94 cM from pTer. Two inherited haplotypes on chromosomes 11p13-12 and 9q21 were observed in all affected members but not in the majority of clinically normal individuals. Sequencing of genes at region 11p13-12 showed a number of sequence variants, two of which were linked with the inherited haplotype. One of these variants in the CD59 gene was found at a very low frequency in the population and could possibly affect pre-messenger RNA splicing. This study is of particular importance for the identification of novel genes that might be responsible for CD other than human leukocyte antigen.
Collapse
Affiliation(s)
- C Vidal
- Laboratory of Molecular Genetics, Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | | | | | | |
Collapse
|
47
|
Bracken S, Byrne G, Kelly J, Jackson J, Feighery C. Altered gene expression in highly purified enterocytes from patients with active coeliac disease. BMC Genomics 2008; 9:377. [PMID: 18691394 PMCID: PMC2533024 DOI: 10.1186/1471-2164-9-377] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Accepted: 08/08/2008] [Indexed: 02/07/2023] Open
Abstract
Background Coeliac disease is a multifactorial inflammatory disorder of the intestine caused by ingestion of gluten in genetically susceptible individuals. Genes within the HLA-DQ locus are considered to contribute some 40% of the genetic influence on this disease. However, information on other disease causing genes is sparse. Since enterocytes are considered to play a central role in coeliac pathology, the aim of this study was to examine gene expression in a highly purified isolate of these cells taken from patients with active disease. Epithelial cells were isolated from duodenal biopsies taken from five coeliac patients with active disease and five non-coeliac control subjects. Contaminating T cells were removed by magnetic sorting. The gene expression profile of the cells was examined using microarray analysis. Validation of significantly altered genes was performed by real-time RT-PCR and immunohistochemistry. Results Enterocyte suspensions of high purity (98–99%) were isolated from intestinal biopsies. Of the 3,800 genes investigated, 102 genes were found to have significantly altered expression between coeliac disease patients and controls (p < 0.05). Analysis of these altered genes revealed a number of biological processes that are potentially modified in active coeliac disease. These processes include events likely to contibute to coeliac pathology, such as altered cell proliferation, differentiation, survival, structure and transport. Conclusion This study provides a profile of the molecular changes that occur in the intestinal epithelium of coeliac patients with active disease. Novel candidate genes were revealed which highlight the contribution of the epithelial cell to the pathogenesis of coeliac disease.
Collapse
Affiliation(s)
- Suzanne Bracken
- Department of Immunology, St, James's Hospital, Dublin and Trinity College Dublin, Dublin Molecular Medicine Centre, Dublin, Ireland.
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Tissue transglutaminase in celiac disease: role of autoantibodies. Amino Acids 2008; 36:693-9. [PMID: 18600381 DOI: 10.1007/s00726-008-0120-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Accepted: 05/25/2008] [Indexed: 12/13/2022]
Abstract
In celiac disease (CD), gluten, the disease-inducing toxic component in wheat, induces the secretion of IgA-class autoantibodies which target tissue transglutaminase (tTG). These autoantibodies are produced in the small-intestinal mucosa, and, during gluten consumption, they can also be detected in patients' serum but disappear slowly from the circulation on a gluten-free diet. Interestingly, after adoption of a gluten-free diet the serum autoantibodies disappear from the circulation more rapidly than the small-intestinal mucosal autoantibody deposits. The finding of IgA deposits on extracellular tTG in the liver, kidney, lymph nodes and muscles of patients with CD indicates that tTG is accessible to the gut-derived autoantibodies. Although the specific autoantibody response directed against tTG is very characteristic in celiac patients, their role in the immunopathology of the celiac mucosal lesion is a matter of debate. Here we report a brief summary of anti-tTG antibody effects demonstrating that these antibodies are functional and not mere bystanders in the disease pathogenesis. In fact, they inhibit intestinal epithelial cell differentiation, induce intestinal epithelial cell proliferation, increase epithelial permeability and activate monocytes and disturb angiogenesis.
Collapse
|
50
|
Abstract
Pathogens are exogenous agents capable of causing disease in susceptible organisms. In celiac sprue, a disease triggered by partially hydrolyzed gluten peptides in the small intestine, the offending immunotoxins cannot replicate, but otherwise have many hallmarks of classical pathogens. First, dietary gluten and its peptide metabolites are ubiquitous components of the modern diet, yet only a small, genetically susceptible fraction of the human population contracts celiac sprue. Second, immunotoxic gluten peptides have certain unusual structural features that allow them to survive the harsh proteolytic conditions of the gastrointestinal tract and thereby interact extensively with the mucosal lining of the small intestine. Third, they invade across epithelial barriers intact to access the underlying gut-associated lymphoid tissue. Fourth, they possess recognition sequences for selective modification by an endogenous enzyme, transglutaminase 2, allowing for in situ activation to a more immunotoxic form via host subversion. Fifth, they precipitate a T cell–mediated immune reaction comprising both innate and adaptive responses that causes chronic inflammation of the small intestine. Sixth, complete elimination of immunotoxic gluten peptides from the celiac diet results in remission, whereas reintroduction of gluten in the diet causes relapse. Therefore, in analogy with antibiotics, orally administered proteases that reduce the host's exposure to the immunotoxin by accelerating gluten peptide destruction have considerable therapeutic potential. Last but not least, notwithstanding the power of in vitro methods to reconstitute the essence of the immune response to gluten in a celiac patient, animal models for the disease, while elusive, are likely to yield fundamentally new systems-level insights.
Collapse
|