51
|
Stolzer I, Ruder B, Neurath MF, Günther C. Interferons at the crossroad of cell death pathways during gastrointestinal inflammation and infection. Int J Med Microbiol 2021; 311:151491. [PMID: 33662871 DOI: 10.1016/j.ijmm.2021.151491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Interferons (IFNs) are pleiotropic immune-modulatory cytokines that are well known for their essential role in host defense against viruses, bacteria, and other pathogenic microorganisms. They can exert both, protective or destructive functions depending on the microorganism, the targeted tissue and the cellular context. Interferon signaling results in the induction of IFN-stimulated genes (ISGs) influencing different cellular pathways including direct anti-viral/anti-bacterial response, immune-modulation or cell death. Multiple pathways leading to host cell death have been described, and it is becoming clear that depending on the cellular context, IFN-induced cell death can be beneficial for both: host and pathogen. Accordingly, activation or repression of corresponding signaling mechanisms occurs during various types of infection but is also an important pathway for gastrointestinal inflammation and tissue damage. In this review, we summarize the role of interferons at the crossroad of various cell death pathways in the gut during inflammation and infection.
Collapse
Affiliation(s)
- Iris Stolzer
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Barbara Ruder
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander-Universität (FAU), Erlangen, Nürnberg, Germany
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany.
| |
Collapse
|
52
|
A comprehensive review of the strategies to improve oral drug absorption with special emphasis on the cellular and molecular mechanisms. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
53
|
Ewing LE, Skinner CM, Pathak R, Yee EU, Krager K, Gurley PC, Melnyk S, Boerma M, Hauer-Jensen M, Koturbash I. Dietary Methionine Supplementation Exacerbates Gastrointestinal Toxicity in a Mouse Model of Abdominal Irradiation. Int J Radiat Oncol Biol Phys 2021; 109:581-593. [PMID: 33002540 PMCID: PMC7855316 DOI: 10.1016/j.ijrobp.2020.09.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Identification of appropriate dietary strategies for prevention of weight and muscle loss in cancer patients is crucial for successful treatment and prolonged patient survival. High-protein oral nutritional supplements decrease mortality and improve indices of nutritional status in cancer patients; however, high-protein diets are often rich in methionine, and experimental evidence indicates that a methionine-supplemented diet (MSD) exacerbates gastrointestinal toxicity after total body irradiation. Here, we sought to investigate whether MSD can exacerbate gastrointestinal toxicity after local abdominal irradiation, an exposure regimen more relevant to clinical settings. MATERIALS AND METHODS Male CBA/CaJ mice fed either a methionine-adequate diet or MSD (6.5 mg methionine/kg diet vs 19.5 mg/kg) received localized abdominal X-irradiation (220 kV, 13 mA) using the Small Animal Radiation Research Platform, and tissues were harvested 4, 7, and 10 days after irradiation. RESULTS MSD exacerbated gastrointestinal toxicity after local abdominal irradiation with 12.5 Gy. This was evident as impaired nutrient absorption was paralleled by reduced body weight recovery. Mechanistically, significant shifts in the gut ecology, evident as decreased microbiome diversity, and substantially increased bacterial species that belong to the genus Bacteroides triggered proinflammatory responses. The latter were evident as increases in circulating neutrophils with corresponding decreases in lymphocytes and associated molecular alterations, exhibited as increases in mRNA levels of proinflammatory genes Icam1, Casp1, Cd14, and Myd88. Altered expression of the tight junction-related proteins Cldn2, Cldn5, and Cldn6 indicated a possible increase in intestinal permeability and bacterial translocation to the liver. CONCLUSIONS We report that dietary supplementation with methionine exacerbates gastrointestinal syndrome in locally irradiated mice. This study demonstrates the important roles registered dieticians should play in clinical oncology and further underlines the necessity of preclinical and clinical investigations in the role of diet in the success of cancer therapy.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Charles M Skinner
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Rupak Pathak
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Eric U Yee
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kim Krager
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Patrick C Gurley
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Stepan Melnyk
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Marjan Boerma
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Martin Hauer-Jensen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
54
|
Gonneaud A, Turgeon N, Boisvert FM, Boudreau F, Asselin C. JAK-STAT Pathway Inhibition Partially Restores Intestinal Homeostasis in Hdac1- and Hdac2-Intestinal Epithelial Cell-Deficient Mice. Cells 2021; 10:224. [PMID: 33498747 PMCID: PMC7911100 DOI: 10.3390/cells10020224] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Abstract
We have previously reported that histone deacetylase epigenetic regulator Hdac1 and Hdac2 deletion in intestinal epithelial cells (IEC) disrupts mucosal tissue architecture and barrier, causing chronic inflammation. In this study, proteome and transcriptome analysis revealed the importance of signaling pathways induced upon genetic IEC-Hdac1 and Hdac2 deletion. Indeed, Gene Ontology biological process analysis of enriched deficient IEC RNA and proteins identified common pathways, including lipid metabolic and oxidation-reduction process, cell adhesion, and antigen processing and presentation, related to immune responses, correlating with dysregulation of major histocompatibility complex (MHC) class II genes. Top upstream regulators included regulators associated with environmental sensing pathways to xenobiotics, microbial and diet-derived ligands, and endogenous metabolites. Proteome analysis revealed mTOR signaling IEC-specific defects. In addition to mTOR, the STAT and Notch pathways were dysregulated specifically in jejunal IEC. To determine the impact of pathway dysregulation on mutant jejunum alterations, we treated mutant mice with Tofacitinib, a JAK inhibitor. Treatment with the inhibitor partially corrected proliferation and tight junction defects, as well as niche stabilization by increasing Paneth cell numbers. Thus, IEC-specific histone deacetylases 1 (HDAC1) and 2 (HDAC2) support intestinal homeostasis by regulating survival and translation processes, as well as differentiation and metabolic pathways. HDAC1 and HDAC2 may play an important role in the regulation of IEC-specific inflammatory responses by controlling, directly or indirectly, the JAK/STAT pathway. IEC-specific JAK/STAT pathway deregulation may be, at least in part, responsible for intestinal homeostasis disruption in mutant mice.
Collapse
Affiliation(s)
- Alexis Gonneaud
- Département D’immunologie et Biologie Cellulaire, Pavillon de Recherche Appliquée Sur le Cancer, Faculté de Médecine et Des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada; (N.T.); (F.-M.B.); (F.B.); (C.A.)
| | | | | | | | | |
Collapse
|
55
|
Mohammad S, Thiemermann C. Role of Metabolic Endotoxemia in Systemic Inflammation and Potential Interventions. Front Immunol 2021; 11:594150. [PMID: 33505393 PMCID: PMC7829348 DOI: 10.3389/fimmu.2020.594150] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Diet-induced metabolic endotoxemia is an important factor in the development of many chronic diseases in animals and man. The gut epithelium is an efficient barrier that prevents the absorption of liposaccharide (LPS). Structural changes to the intestinal epithelium in response to dietary alterations allow LPS to enter the bloodstream, resulting in an increase in the plasma levels of LPS (termed metabolic endotoxemia). LPS activates Toll-like receptor-4 (TLR4) leading to the production of numerous pro-inflammatory cytokines and, hence, low-grade systemic inflammation. Thus, metabolic endotoxemia can lead to several chronic inflammatory conditions. Obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD) can also cause an increase in gut permeability and potential pharmacological and dietary interventions could be used to reduce the chronic low-grade inflammation associated with endotoxemia.
Collapse
Affiliation(s)
- Shireen Mohammad
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
56
|
Urbauer E, Rath E, Haller D. Mitochondrial Metabolism in the Intestinal Stem Cell Niche-Sensing and Signaling in Health and Disease. Front Cell Dev Biol 2021; 8:602814. [PMID: 33469536 PMCID: PMC7813778 DOI: 10.3389/fcell.2020.602814] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial metabolism, dynamics, and stress responses in the intestinal stem cell niche play a pivotal role in regulating intestinal epithelial cell homeostasis, including self-renewal and differentiation. In addition, mitochondria are increasingly recognized for their involvement in sensing the metabolic environment and their capability of integrating host and microbial-derived signals. Gastrointestinal diseases such as inflammatory bowel diseases and colorectal cancer are characterized by alterations of intestinal stemness, the microbial milieu, and mitochondrial metabolism. Thus, mitochondrial function emerges at the interface of determining health and disease, and failure to adapt mitochondrial function to environmental cues potentially results in aberrant tissue responses. A mechanistic understanding of the underlying role of mitochondrial fitness in intestinal pathologies is still in its infancy, and therapies targeting mitochondrial (dys)function are currently lacking. This review discusses mitochondrial signaling and metabolism in intestinal stem cells and Paneth cells as critical junction translating host- and microbe-derived signals into epithelial responses. Consequently, we propose mitochondrial fitness as a hallmark for intestinal epithelial cell plasticity, determining the regenerative capacity of the epithelium.
Collapse
Affiliation(s)
- Elisabeth Urbauer
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany.,ZIEL Institute for Food & Health, Technische Universität München, Munich, Germany
| |
Collapse
|
57
|
Bruellman R, Llorente C. A Perspective Of Intestinal Immune-Microbiome Interactions In Alcohol-Associated Liver Disease. Int J Biol Sci 2021; 17:307-327. [PMID: 33390852 PMCID: PMC7757023 DOI: 10.7150/ijbs.53589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Uncovering the intricacies of the gut microbiome and how it interacts with the host immune system has opened up pathways in the search for the treatment of disease conditions. Alcohol-associated liver disease is a major cause of death worldwide. Research has shed light on the breakdown of the protective gut barriers, translocation of gut microbes to the liver and inflammatory immune response to microbes all contributing to alcohol-associated liver disease. This knowledge has opened up avenues for alternative therapies to alleviate alcohol-associated liver disease based on the interaction of the commensal gut microbiome as a key player in the regulation of the immune response. This review describes the relevance of the intestinal immune system, the gut microbiota, and specialized and non-specialized intestinal cells in the regulation of intestinal homeostasis. It also reflects how these components are altered during alcohol-associated liver disease and discusses new approaches for potential future therapies in alcohol-associated liver disease.
Collapse
Affiliation(s)
- Ryan Bruellman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
58
|
Olshan KL, Leonard MM, Serena G, Zomorrodi AR, Fasano A. Gut microbiota in Celiac Disease: microbes, metabolites, pathways and therapeutics. Expert Rev Clin Immunol 2020; 16:1075-1092. [PMID: 33103934 DOI: 10.1080/1744666x.2021.1840354] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Current evidence supports a vital role of the microbiota on health outcomes, with alterations in an otherwise healthy balance linked to chronic medical conditions like celiac disease (CD). Recent advances in microbiome analysis allow for unparalleled profiling of the microbes and metabolites. With the growing volume of data available, trends are emerging that support a role for the gut microbiota in CD pathogenesis. AREAS COVERED In this article, the authors review the relationship between factors such as genes and antibiotic exposure on CD onset and the intestinal microbiota. The authors also review other microbiota within the human body (like the oropharynx) that may play a role in CD pathogenesis. Finally, the authors discuss implications for disease modification and the ultimate goal of prevention. The authors reviewed literature from PubMed, EMBASE, and Web of Science. EXPERT OPINION CD serves as a unique opportunity to explore the role of the intestinal microbiota on the development of chronic autoimmune disease. While research to date provides a solid foundation, most studies have been case-control and thus do not have capacity to explore the mechanistic role of the microbiota in CD onset. Further longitudinal studies and integrated multi-omics are necessary for investigating CD pathogenesis.
Collapse
Affiliation(s)
- Katherine L Olshan
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Ali R Zomorrodi
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,European Biomedical Research Institute of Salerno (EBRIS) , Salerno, Italy
| |
Collapse
|
59
|
Nikolenko VN, Oganesyan MV, Sankova MV, Bulygin KV, Vovkogon AD, Rizaeva NA, Sinelnikov MY. Paneth cells: Maintaining dynamic microbiome-host homeostasis, protecting against inflammation and cancer. Bioessays 2020; 43:e2000180. [PMID: 33244814 DOI: 10.1002/bies.202000180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022]
Abstract
The human intestines are constantly under the influence of numerous pathological factors: enteropathogenic microorganisms, food antigens, physico-chemical stress associated with digestion and bacterial metabolism, therefore it must be provided with a system of protection against adverse impact. Recent studies have shown that Paneth cells play a crucial role in maintaining homeostasis of the small intestines. Paneth cells perform many vital functions aimed at maintaining a homeostatic balance between normal microbiota, infectious pathogens and the human body, regulate the qualitative composition and number of intestinal microorganisms, prevent the introduction of potentially pathogenic species, and protect stem cells from damage. Paneth cells take part in adaptive and protective-inflammatory reactions. Paneth cells maintain dynamic balance between microbial populations, and the macroorganism, preventing the development of intestinal infections and cancer. They play a crucial role in gastrointestinal homeostasis and may be key factors in the etiopathological progression of intestinal diseases.
Collapse
Affiliation(s)
- Vladimir N Nikolenko
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia.,Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Marine V Oganesyan
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Maria V Sankova
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia.,Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Andzhela D Vovkogon
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Negoriya A Rizaeva
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | | |
Collapse
|
60
|
Markus J, Landry T, Stevens Z, Scott H, Llanos P, Debatis M, Armento A, Klausner M, Ayehunie S. Human small intestinal organotypic culture model for drug permeation, inflammation, and toxicity assays. In Vitro Cell Dev Biol Anim 2020; 57:160-173. [PMID: 33237403 PMCID: PMC7687576 DOI: 10.1007/s11626-020-00526-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
The gastrointestinal tract (GIT), in particular, the small intestine, plays a significant role in food digestion, fluid and electrolyte transport, drug absorption and metabolism, and nutrient uptake. As the longest portion of the GIT, the small intestine also plays a vital role in protecting the host against pathogenic or opportunistic microbial invasion. However, establishing polarized intestinal tissue models in vitro that reflect the architecture and physiology of the gut has been a challenge for decades and the lack of translational models that predict human responses has impeded research in the drug absorption, metabolism, and drug-induced gastrointestinal toxicity space. Often, animals fail to recapitulate human physiology and do not predict human outcomes. Also, certain human pathogens are species specific and do not infect other hosts. Concerns such as variability of results, a low throughput format, and ethical considerations further complicate the use of animals for predicting the safety and efficacy xenobiotics in humans. These limitations necessitate the development of in vitro 3D human intestinal tissue models that recapitulate in vivo–like microenvironment and provide more physiologically relevant cellular responses so that they can better predict the safety and efficacy of pharmaceuticals and toxicants. Over the past decade, much progress has been made in the development of in vitro intestinal models (organoids and 3D-organotypic tissues) using either inducible pluripotent or adult stem cells. Among the models, the MatTek’s intestinal tissue model (EpiIntestinal™ Ashland, MA) has been used extensively by the pharmaceutical industry to study drug permeation, metabolism, drug-induced GI toxicity, pathogen infections, inflammation, wound healing, and as a predictive model for a clinical adverse outcome (diarrhea) to pharmaceutical drugs. In this paper, our review will focus on the potential of in vitro small intestinal tissues as preclinical research tool and as alternative to the use of animals.
Collapse
Affiliation(s)
- Jan Markus
- In Vitro Life Science Laboratories, Bratislava, Slovak Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Dabir PD, van der Post RS, Nagtegaal ID. Incidental morphological findings in colorectal adenomas. Histopathology 2020; 78:348-357. [PMID: 32981102 PMCID: PMC7894322 DOI: 10.1111/his.14263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/01/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022]
Abstract
Owing to a sharp increase in the frequency of diagnosis of colorectal adenomas in the current era of population screening, distinctive morphological features are increasingly being observed. These may present diagnostic challenges and cause clinical management issues. Paneth cell metaplasia is a more common occurrence, but the incidence rates of squamous metaplasia, clear cell metaplasia, osseous metaplasia, neuroendocrine differentiation and signet‐ring cell‐like lesion are low, and they can be seen in <1% of colorectal adenomas. Their histomorphological characteristics are quite unique; ancillary studies are not very helpful and often not needed. In this review, we give an overview and describe the potential clinical consequences of such incidental and special morphological findings in colorectal adenomas.
Collapse
Affiliation(s)
- Parag D Dabir
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institute of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
62
|
Khaloian S, Rath E, Hammoudi N, Gleisinger E, Blutke A, Giesbertz P, Berger E, Metwaly A, Waldschmitt N, Allez M, Haller D. Mitochondrial impairment drives intestinal stem cell transition into dysfunctional Paneth cells predicting Crohn's disease recurrence. Gut 2020; 69:1939-1951. [PMID: 32111634 PMCID: PMC7569388 DOI: 10.1136/gutjnl-2019-319514] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Reduced Paneth cell (PC) numbers are observed in inflammatory bowel diseases and impaired PC function contributes to the ileal pathogenesis of Crohn's disease (CD). PCs reside in proximity to Lgr5+ intestinal stem cells (ISC) and mitochondria are critical for ISC-renewal and differentiation. Here, we characterise ISC and PC appearance under inflammatory conditions and describe the role of mitochondrial function for ISC niche-maintenance. DESIGN Ileal tissue samples from patients with CD, mouse models for mitochondrial dysfunction (Hsp60Δ/ΔISC) and CD-like ileitis (TNFΔARE), and intestinal organoids were used to characterise PCs and ISCs in relation to mitochondrial function. RESULTS In patients with CD and TNFΔARE mice, inflammation correlated with reduced numbers of Lysozyme-positive granules in PCs and decreased Lgr5 expression in crypt regions. Disease-associated changes in PC and ISC appearance persisted in non-inflamed tissue regions of patients with CD and predicted the risk of disease recurrence after surgical resection. ISC-specific deletion of Hsp60 and inhibition of mitochondrial respiration linked mitochondrial function to the aberrant PC phenotype. Consistent with reduced stemness in vivo, crypts from inflamed TNFΔARE mice fail to grow into organoids ex vivo. Dichloroacetate-mediated inhibition of glycolysis, forcing cells to shift to mitochondrial respiration, improved ISC niche function and rescued the ability of TNFΔARE mice-derived crypts to form organoids. CONCLUSION We provide evidence that inflammation-associated mitochondrial dysfunction in the intestinal epithelium triggers a metabolic imbalance, causing reduced stemness and acquisition of a dysfunctional PC phenotype. Blocking glycolysis might be a novel drug target to antagonise PC dysfunction in the pathogenesis of CD.
Collapse
Affiliation(s)
- Sevana Khaloian
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Nassim Hammoudi
- Department of Gastroenterology, Hôpital Saint-Louis, APHP, INSERM U1160, Université de Paris 1, Paris, Île-de-France, France
| | - Elisabeth Gleisinger
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Pieter Giesbertz
- Chair of Nutrition Physiology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Emanuel Berger
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Nadine Waldschmitt
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, APHP, INSERM U1160, Université de Paris 1, Paris, Île-de-France, France
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany .,ZIEL Institute for Food & Health, Technische Universität München, München, Germany
| |
Collapse
|
63
|
Escudero-Hernández C. Epithelial cell dysfunction in coeliac disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 358:133-164. [PMID: 33707053 DOI: 10.1016/bs.ircmb.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The intestinal epithelium limits host-luminal interactions and maintains gut homeostasis. Breakdown of the epithelial barrier and villous atrophy are hallmarks of coeliac disease. Besides the well characterized immune-mediated epithelial damage induced in coeliac mucosa, constitutional changes and early gluten direct effects disturb intestinal epithelial cells. The subsequent modifications in key epithelial signaling pathways leads to outnumbered immature epithelial cells that, in turn, facilitate epithelial dysfunction, promote crypt hyperplasia, and increase intestinal permeability. Consequently, underlying immune cells have a greater access to gluten, which boosts the proinflammatory immune response against gluten and positively feedback the epithelial damage loop. Gluten-free diet is an indispensable treatment for coeliac disease patients, but additional therapies are under development, including those that reinforce intestinal epithelial healing. In this chapter, we provide an overview of intestinal epithelial cell disturbances that develop during gluten intake in coeliac disease mucosa.
Collapse
|
64
|
Hartl K, Sigal M. Microbe-Driven Genotoxicity in Gastrointestinal Carcinogenesis. Int J Mol Sci 2020; 21:E7439. [PMID: 33050171 PMCID: PMC7587957 DOI: 10.3390/ijms21207439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium serves as a barrier to discriminate the outside from the inside and is in constant exchange with the luminal contents, including nutrients and the microbiota. Pathogens have evolved mechanisms to overcome the multiple ways of defense in the mucosa, while several members of the microbiota can exhibit pathogenic features once the healthy barrier integrity of the epithelium is disrupted. This not only leads to symptoms accompanying the acute infection but may also contribute to long-term injuries such as genomic instability, which is linked to mutations and cancer. While for Helicobacter pylori a link between infection and cancer is well established, many other bacteria and their virulence factors have only recently been linked to gastrointestinal malignancies through epidemiological as well as mechanistic studies. This review will focus on those pathogens and members of the microbiota that have been linked to genotoxicity in the context of gastric or colorectal cancer. We will address the mechanisms by which such bacteria establish contact with the gastrointestinal epithelium-either via an existing breach in the barrier or via their own virulence factors as well as the mechanisms by which they interfere with host genomic integrity.
Collapse
Affiliation(s)
- Kimberly Hartl
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, 10117 Berlin, Germany;
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Michael Sigal
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, 10117 Berlin, Germany;
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| |
Collapse
|
65
|
Marcon CF, Ferreira PTM, Franco PS, Ribeiro M, Silva RJ, Sousa RAP, Oliveira CJF, Rodrigues Junior V, Gomes MLM, Lazo Chica JE, Mineo TWP, Mineo JR, Barbosa BF, Ferro EAV, Gomes AO. Macrophage migration inhibitory factor (MIF) and pregnancy may impact the balance of intestinal cytokines and the development of intestinal pathology caused by Toxoplasma gondii infection. Cytokine 2020; 136:155283. [PMID: 32947151 DOI: 10.1016/j.cyto.2020.155283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Toxoplasma gondii (T. gondii) is an intracellular parasite responsible for causing toxoplasmosis. When infection occurs during pregnancy, it can produce severe congenital infection with ocular and neurologic damage to the infant. From the oral infection parasite reaches the intestine, causing inflammatory response, damage in tissue architecture and systemic dissemination. Macrophage migration inhibition factor (MIF) is a cytokine secreted from both immune and non-immune cells, including gut epithelial cells. MIF is described to promote inflammatory responses, to be associated in colitis pathogenesis and also to play role in maintaining the intestinal barrier. The aim of the present study was to evaluate the influence of the pregnancy and MIF deficiency on T. gondii infection in the intestinal microenvironment and to address how these factors can impact on the intestinal architecture and local cytokine profile. For this purpose, small intestine of pregnant and non-pregnant C57BL/6 MIF deficient mice (MIF-/-) and Wild-type (WT) orally infected with 5 cysts of ME-49 strain of T. gondii were collected on day 8th of infection. Intestines were processed for morphological and morphometric analyses, parasite quantification and for cytokines mensuration. Our results showed that the absence of MIF and pregnancy caused an increase in T. gondii infection index. T. gondii immunolocalization demonstrated that segments preferentially infected with T. gondii were duodenum and ileum. The infection caused a reduction in the size of the intestinal villi, whereas, infection associated with pregnancy caused an increase in villi size due to edema caused by the infection. Also, the goblet cell number was increased in the ileum of MIF-/- mice, when compared to the corresponding WT group. Analyses of cytokine production in the small intestine showed that MIF was up regulated in the gut of pregnant WT mice due to infection. Also, infection provoked an intense Th1 response that was more exacerbated in pregnant MIF-/- mice. We also detected that the Th2/Treg response was more pronounced in MIF-/- mice. Altogether, our results demonstrated that pregnancy and MIF deficiency interferes in the balance of the intestinal cytokines and favors a Th1-immflamatory profile, which in turn, impact in the development of pathology caused by T. gondii infection in the intestinal microenvironment.
Collapse
Affiliation(s)
- Camila Ferreira Marcon
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Paula Tatiana Mutão Ferreira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Priscila Silva Franco
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Roberto Augusto Pereira Sousa
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Carlo José Freire Oliveira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Virmondes Rodrigues Junior
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marcos Lucca Moreira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Javier Emílio Lazo Chica
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Angelica Oliveira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
66
|
Di W, Lv Y, Xia F, Sheng Y, Liu J, Ding G. Improvement of intestinal stem cells and barrier function via energy restriction in middle-aged C57BL/6 mice. Nutr Res 2020; 81:47-57. [PMID: 32877836 DOI: 10.1016/j.nutres.2020.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 05/14/2020] [Accepted: 06/22/2020] [Indexed: 01/21/2023]
Abstract
This study aimed to reveal the impact of energy restriction on the intestine via structural and molecular changes in terms of intestinal stem cell (ISC) function, ISC niche, intestinal epithelial barrier function, and intestinal immune function. Female C57BL/6J mice, aged 12 months, fed a commercial chow were used in this study. The ISC function, ISC niche, intestinal epithelial barrier function, and intestinal immune function were assessed. Energy restriction reversed aging-induced intestinal shortening and made the crypts shallower. The intestinal epithelial cells isolated from the intestine showed a significant increase in the expression levels of stem cell-associated genes in small intestinal epithelial cells as detected by flow cytometry. Despite the increase in the number of stem cells and the expression levels of markers, no increase or decrease was found in the enteroid complexity of the small intestine and colonic enteroid formation in vitro. The colonic mucous layer was measured in mice of the energy restricted (ER)-treated group to investigate the epithelial barrier function in the colon. The results revealed that the barrier was more complete. The fluorescence intensity of tight junction markers claudin-2 and zonula occludens-1 increased and the mRNA expression profiles of monocyte chemotactic protein 1 and interleukin-6 decreased in the colon of mice in the ER-treated group. The beneficial effects of ER on the colon in terms of the integrity of the mucosal barrier and alleviation of inflammation were confirmed, thus highlighting the importance of modulating the intestinal function in developing effective antiaging dietary interventions.
Collapse
Affiliation(s)
- Wenjuan Di
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Lv
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan Xia
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunlu Sheng
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxian Ding
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
67
|
Rosa JM, Pazini FL, Camargo A, Wolin IAV, Olescowicz G, Eslabão LB, Romero OB, Winkelmann-Duarte EC, S Rodrigues AL. Prophylactic effect of physical exercise on Aβ 1-40-induced depressive-like behavior and gut dysfunction in mice. Behav Brain Res 2020; 393:112791. [PMID: 32599000 DOI: 10.1016/j.bbr.2020.112791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disease that is highly comorbid with depression. Gut dysfunction has been proposed as a possible risk factor for both clinical conditions. In the present study, we investigated the ability of treadmill exercise for 4 weeks (5 days/week, 40 min/day) to counteract amyloid β1-40 peptide (Aβ1-40)-induced depressive-like behavior, alterations in morphological parameters of the duodenum, and the abundance of Firmicutes and Bacteroidetes phyla. Aβ1-40 administration (400 pmol/mouse, i.c.v.) increased immobility time in the tail suspension test (TST) and reduced time spent sniffing in the female urine sniffing test (FUST), indicating behavioral despair and impairment in reward-seeking behavior. These behavioral alterations, indicative of depressive-like behavior, were accompanied by reduced villus width in the duodenum. Moreover, photomicrographs obtained by transmission electron microscopy revealed abnormal epithelial microvilli in the duodenum from sedentary Aβ1-40-exposed mice, characterized by shorter microvilli and heterogeneity in the length of these structures that exhibit a disordered packing. Regarding the ultrastructure of Paneth cells, Aβ1-40 administration caused a reduction in the secretory granule diameter, as well as an enlarged peripheral halo. These animals also presented reduced Firmicutes and increased Bacteroidetes abundance, and increased Bacteroidetes/Firmicutes ratio. Most of the alterations observed in Aβ1-40-exposed mice were prevented by the practice of physical exercise. Altogether the results provide evidence of the prophylactic effect of physical exercise on Aβ1-40-induced depressive-like behavior and gut dysfunction in mice, suggesting that physical exercise could be useful for preventing depression associated with AD.
Collapse
Affiliation(s)
- Julia M Rosa
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Francis L Pazini
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Ingrid A V Wolin
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Gislaine Olescowicz
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Livia B Eslabão
- Department of Microbiology, Immunology and Parasitology, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Oscar Bruna Romero
- Department of Microbiology, Immunology and Parasitology, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Elisa C Winkelmann-Duarte
- Department of Morphological Sciences, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil.
| |
Collapse
|
68
|
Marcon CF, Ferreira PTM, Franco PS, Ribeiro M, Silva RJ, Sousa RAP, Oliveira CJF, Junior VR, Gomes MLM, Chica JEL, Mineo TWP, Mineo JR, Barbosa BF, Ferro EAV, Gomes AO. WITHDRAWN: Macrophage migration inhibitory factor (MIF) and pregnancy may impact the balance of intestinal cytokines and the development of intestinal pathology caused by Toxoplasma gondii infection. Cytokine X 2020; 2:100034. [PMID: 33604559 PMCID: PMC7885889 DOI: 10.1016/j.cytox.2020.100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Camila Ferreira Marcon
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Paula Tatiana Mutão Ferreira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Priscila Silva Franco
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Roberto Augusto Pereira Sousa
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Carlo José Freire Oliveira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Virmondes Rodrigues Junior
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marcos Lucca Moreira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Javier Emílio Lazo Chica
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Angelica Oliveira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| |
Collapse
|
69
|
Atypical Salmonellosis in a Horse: Implications for Hospital Safety. Case Rep Vet Med 2020; 2020:7062408. [PMID: 32566354 PMCID: PMC7292965 DOI: 10.1155/2020/7062408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 11/17/2022] Open
Abstract
A 17-year-old Quarter Horse mare was evaluated for colic of 24-hour duration. Clinical signs and diagnostic evaluation were consistent with duodenitis-proximal jejunitis. The horse's clinical condition deteriorated despite medical treatment and was euthanized. Aerobic culture collected from small intestinal ingesta was positive for Salmonella enterica subsp. enterica serovar Hadar. Salmonella sp. is commonly implicated in nosocomial infections in equine veterinary hospitals usually through feces containing the organism. Considering Salmonella sp. was cultured from the jejunal luminal contents and the large volume of nasogastric reflux that was evacuated in this case, a perceived risk of Salmonella sp. transmission from infected gastric reflux to other hospitalized cases was realized. Infectious agent biosecurity precautions should be undertaken in horses with nasogastric reflux to prevent hospital-acquired transmission.
Collapse
|
70
|
Fakhoury HMA, Kvietys PR, AlKattan W, Anouti FA, Elahi MA, Karras SN, Grant WB. Vitamin D and intestinal homeostasis: Barrier, microbiota, and immune modulation. J Steroid Biochem Mol Biol 2020; 200:105663. [PMID: 32194242 DOI: 10.1016/j.jsbmb.2020.105663] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/28/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023]
Abstract
Vitamin D plays a pivotal role in intestinal homeostasis. Vitamin D can impact the function of virtually every cell in the gut by binding to its intracellular receptor (VDR) and subsequently transcribing relevant genes. In the lumen, the mucus layer and the underlying epithelium serve to keep resident microbiota at bay. Vitamin D ensures an appropriate level of antimicrobial peptides in the mucus and maintains epithelial integrity by reinforcing intercellular junctions. Should bacteria penetrate the epithelial layer and enter the interstitium, immune sentinel cells (e.g. macrophages, dendritic cells, and innate lymphoid cells) elicit inflammation and trigger the adaptive immune response by activating Th1/Th17 cells. Vitamin D/VDR signaling in these cells ensures clearance of the bacteria. Subsequently, vitamin D also quiets the adaptive immune system by suppressing the Th1/Th17 cells and favoring Treg cells. The importance of vitamin D/VDR signaling in intestinal homeostasis is evidenced by the development of a chronic inflammatory state (e.g. IBD) when this signaling system is disrupted.
Collapse
Affiliation(s)
- Hana M A Fakhoury
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Peter R Kvietys
- Department of Physiology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Wael AlKattan
- Department of Surgery, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Fatme Al Anouti
- College of Natural and Health Sciences, Department of Public Health and Nutrition, Zayed University, Abu Dhabi, United Arab Emirates
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Spyridon N Karras
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - William B Grant
- Director, Sunlight, Nutrition, and Health Research Center, San Francisco, CA, USA
| |
Collapse
|
71
|
Overcoming the intestinal barrier: A look into targeting approaches for improved oral drug delivery systems. J Control Release 2020; 322:486-508. [DOI: 10.1016/j.jconrel.2020.04.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/17/2022]
|
72
|
Colquhoun C, Duncan M, Grant G. Inflammatory Bowel Diseases: Host-Microbial-Environmental Interactions in Dysbiosis. Diseases 2020; 8:E13. [PMID: 32397606 PMCID: PMC7348996 DOI: 10.3390/diseases8020013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Crohn's Disease (CD) and Ulcerative Colitis (UC) are world-wide health problems in which intestinal dysbiosis or adverse functional changes in the microbiome are causative or exacerbating factors. The reduced abundance and diversity of the microbiome may be a result of a lack of exposure to vital commensal microbes or overexposure to competitive pathobionts during early life. Alternatively, many commensal bacteria may not find a suitable intestinal niche or fail to proliferate or function in a protective/competitive manner if they do colonize. Bacteria express a range of factors, such as fimbriae, flagella, and secretory compounds that enable them to attach to the gut, modulate metabolism, and outcompete other species. However, the host also releases factors, such as secretory IgA, antimicrobial factors, hormones, and mucins, which can prevent or regulate bacterial interactions with the gut or disable the bacterium. The delicate balance between these competing host and bacteria factors dictates whether a bacterium can colonize, proliferate or function in the intestine. Impaired functioning of NOD2 in Paneth cells and disrupted colonic mucus production are exacerbating features of CD and UC, respectively, that contribute to dysbiosis. This review evaluates the roles of these and other the host, bacterial and environmental factors in inflammatory bowel diseases.
Collapse
Affiliation(s)
| | | | - George Grant
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (C.C.); (M.D.)
| |
Collapse
|
73
|
Pearce SC, Weber GJ, van Sambeek DM, Soares JW, Racicot K, Breault DT. Intestinal enteroids recapitulate the effects of short-chain fatty acids on the intestinal epithelium. PLoS One 2020; 15:e0230231. [PMID: 32240190 PMCID: PMC7117711 DOI: 10.1371/journal.pone.0230231] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
Enteroids are cultured primary intestinal epithelial cells that recapitulate epithelial lineage development allowing for a more complex and physiologically relevant model for scientific study. The large presence of intestinal stem cells (ISC) in these enteroids allows for the study of metabolite effects on cellular processes and resulting progeny cells. Short-chain fatty acids (SCFA) such as butyrate (BUT) are bacterial metabolites produced in the gastrointestinal tract that are considered to be beneficial to host cells. Therefore, the objective was to study the effects of SCFAs on biomarkers of ISC activity, differentiation, barrier function and epithelial defense in the intestine using mouse and human enteroid models. Enteroids were treated with two concentrations of acetate (ACET), propionate (PROP), or BUT for 24 h. Enteroids treated with BUT or PROP showed a decrease in proliferation via EdU uptake relative to the controls in both mouse and human models. Gene expression of Lgr5 was shown to decrease with BUT and PROP treatments, but increased with ACET. As a result of BUT and PROP treatments, there was an increase in differentiation markers for enterocyte, Paneth, goblet, and enteroendocrine cells. Gene expression of antimicrobial proteins Reg3β, Reg3γ, and Defb1 were stimulated by BUT and PROP, but not by ACET which had a greater effect on expression of tight junction genes Cldn3 and Ocln in 3D enteroids. Similar results were obtained with human enteroids treated with 10 mM SCFAs and grown in either 3D or Transwell™ model cultures, although tight junctions were influenced by BUT and PROP, but not ACET in monolayer format. Furthermore, BUT and PROP treatments increased transepithelial electrical resistance after 24 h compared to ACET or control. Overall, individual SCFAs are potent stimulators of cellular gene expression, however, PROP and especially BUT show great efficacy for driving cell differentiation and gene expression.
Collapse
Affiliation(s)
- Sarah C. Pearce
- Performance Nutrition Team, Combat Feeding Directorate, Combat Capabilities Development Command Soldier Center, Natick, Massachusetts, United States of America
- * E-mail:
| | - Gregory J. Weber
- Performance Nutrition Team, Combat Feeding Directorate, Combat Capabilities Development Command Soldier Center, Natick, Massachusetts, United States of America
| | - Dana M. van Sambeek
- Performance Nutrition Team, Combat Feeding Directorate, Combat Capabilities Development Command Soldier Center, Natick, Massachusetts, United States of America
| | - Jason W. Soares
- Biological Sciences & Technology Team, Soldier Performance Optimization Directorate, Combat Capabilities Development Command Soldier Center, Natick, Massachusetts, United States of America
| | - Kenneth Racicot
- Biological Sciences & Technology Team, Soldier Performance Optimization Directorate, Combat Capabilities Development Command Soldier Center, Natick, Massachusetts, United States of America
| | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Cambridge, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
74
|
Singh R, Balasubramanian I, Zhang L, Gao N. Metaplastic Paneth Cells in Extra-Intestinal Mucosal Niche Indicate a Link to Microbiome and Inflammation. Front Physiol 2020; 11:280. [PMID: 32296343 PMCID: PMC7138011 DOI: 10.3389/fphys.2020.00280] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Paneth cells are residents of the intestinal epithelium. Abnormal appearance of Paneth cells has been widely documented in non-intestinal tissues within the digestive tract and even observed in non-gastrointestinal organs. Although metaplastic Paneth cells are part of the overarching pathology of intestinal metaplasia (IM), only a fraction of intestinal metaplastic lesions contain Paneth cells. We survey literature documenting metaplastic Paneth cells to gain insights into mechanism underlying their etiologic development as well as their potential relevance to human health. A synthesized view from this study suggests that the emergence of metaplastic Paneth cells at extra-intestinal mucosal sites likely represents a protective, anti-bacterial, and inflammatory response evoked by an altered microbial activity.
Collapse
Affiliation(s)
- Rajbir Singh
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States
| | | | - Lanjing Zhang
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.,Department of Pathology, Princeton Medical Center, Plainsboro, NJ, United States
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
75
|
Rees WD, Sly LM, Steiner TS. How do immune and mesenchymal cells influence the intestinal epithelial cell compartment in inflammatory bowel disease? Let's crosstalk about it! J Leukoc Biol 2020; 108:309-321. [PMID: 32057139 DOI: 10.1002/jlb.3mir0120-567r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/12/2020] [Accepted: 01/17/2020] [Indexed: 12/18/2022] Open
Abstract
Intestinal epithelial cells provide a front line of defense by establishing a barrier against food Ags, pathogens, and commensal microorganisms. This defense includes the establishment of a tolerogenic environment in the gastrointestinal (GI) tract. The intestinal epithelium replenishes itself by cell turnover every 4-5 days, and this process is facilitated by various pathways of communication between the intestinal epithelial cells (IECs), the underlying stromal cell network, and professional immune cells, which together help establish a proper intestinal stem cell (ISC) niche in the crypt. However, during a state of inflammation, such as in inflammatory bowel diseases (IBD), these communication pathways can be altered, and this can lead to the development of inflammatory IECs within the crypt that further drive inflammation. Here, we review the current literature looking at crosstalk between immune cells, stromal cells, and IECs: how does the immune system potentially alter the ISC niche, and how do IECs influence intestinal immunity? We discuss the latest research using single cell RNA sequencing and intestinal organoid cultures to help answer these questions. A better understanding of this complex crosstalk can help lead to a better understanding of intestinal biology in general, and more efficient therapeutic approaches to treat IBD.
Collapse
Affiliation(s)
- William D Rees
- Department of Medicine, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura M Sly
- Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Theodore S Steiner
- Department of Medicine, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
76
|
Potential of glucocorticoids to treat intestinal inflammation during sepsis. Curr Opin Pharmacol 2020; 53:1-7. [PMID: 31991314 DOI: 10.1016/j.coph.2019.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/16/2019] [Accepted: 12/22/2019] [Indexed: 01/07/2023]
Abstract
Glucocorticoids (GCs) are steroid hormones characterized by their anti-inflammatory and immunosuppressive nature. Although GCs are very commonly prescribed, in several diseases, including sepsis, their clinical treatment is hampered by side effects and by the occurrence of glucocorticoid resistance (GCR). Sepsis is defined as a life-threatening organ dysfunction, initiated by a dysregulated systemic host response to infections. With at least 19 million cases per year and a lethality rate of about 25%, sepsis is one of the most urgent unmet medical needs. The gut is critically affected during sepsis and is considered as a driving force in this disease. Despite there is no effective treatment for sepsis, pre-clinical studies show promising results by preserving or restoring gut integrity. Since GC treatment reveals therapeutic effects in Crohn's disease (CD) and in pre-clinical sepsis models, we hypothesize that targeting GCs to the gut or stimulating local GC production in the gut forms an interesting strategy for sepsis treatment. According to recent findings that show that dimerization of the glucocorticoid receptor (GR) is essential in inducing anti-inflammatory effects in pre-clinical sepsis models, we predict that new generation GCs that selectively dimerize the GR, can therefore positively affect the outcome of sepsis treatment.
Collapse
|
77
|
Zhang X, Fisher R, Hou W, Shields D, Epperly MW, Wang H, Wei L, Leibowitz BJ, Yu J, Alexander LM, VAN Pijkeren JP, Watkins S, Wipf P, Greenberger JS. Second-generation Probiotics Producing IL-22 Increase Survival of Mice After Total Body Irradiation. In Vivo 2020; 34:39-50. [PMID: 31882461 PMCID: PMC6984118 DOI: 10.21873/invivo.11743] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/21/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM Intestinal damage induced by total body irradiation (TBI) reduces leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5)-expressing stem cells, goblet, and Paneth cells, breaching the epithelial lining, and facilitating bacterial translocation, sepsis, and death. MATERIALS AND METHODS Survival was measured after TBI in animals that received wild-type or recombinant bacteria producing interleukin-22 (IL-22). Changes in survival due to microbially delivered IL-22 were measured. Lactobacillus reuteri producing IL-22, or Escherichia coli-IL-22 were compared to determine which delivery system is better. RESULTS C57BL/6 mice receiving IL-22 probiotics at 24 h after 9.25 Gy TBI, demonstrated green fluorescent protein-positive bacteria in the intestine, doubled the number of Lgr5+ intestinal stem cells, and increased 30-day survival. Bacteria were localized to the jejunum, ileum, and colon. CONCLUSION Second-generation probiotics appear to be valuable for mitigation of TBI, and radiation protection during therapeutic total abdominal irradiation.
Collapse
Affiliation(s)
- Xichen Zhang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Liang Wei
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Brian J Leibowitz
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, U.S.A
| | | | - Simon Watkins
- Center for Imaging, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A.
| |
Collapse
|
78
|
Generating an Artificial Intestine for the Treatment of Short Bowel Syndrome. Gastroenterol Clin North Am 2019; 48:585-605. [PMID: 31668185 DOI: 10.1016/j.gtc.2019.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Intestinal failure is defined as the inability to maintain fluid, nutrition, energy, and micronutrient balance that leads to the inability to gain or maintain weight, resulting in malnutrition and dehydration. Causes of intestinal failure include short bowel syndrome (ie, the physical loss of intestinal surface area and severe intestinal dysmotility). For patients with intestinal failure who fail to achieve enteral autonomy through intestinal rehabilitation programs, the current treatment options are expensive and associated with severe complications. Therefore, the need persists for next-generation therapies, including cell-based therapy, to increase intestinal regeneration, and development of the tissue-engineered small intestine.
Collapse
|
79
|
Das B, Okamoto K, Rabalais J, Kozan PA, Marchelletta RR, McGeough MD, Durali N, Go M, Barrett KE, Das S, Sivagnanam M. Enteroids expressing a disease-associated mutant of EpCAM are a model for congenital tufting enteropathy. Am J Physiol Gastrointest Liver Physiol 2019; 317:G580-G591. [PMID: 31433211 PMCID: PMC6879886 DOI: 10.1152/ajpgi.00098.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Congenital tufting enteropathy (CTE) is an autosomal recessive disease characterized by severe intestinal failure in infancy and mutations in the epithelial cell adhesion molecule (EPCAM) gene. Previous studies of CTE in mice expressing mutant EpCAM show neonatal lethality. Hence, to study the cellular, molecular, and physiological alterations that result from EpCAM mutation, a tamoxifen-inducible mutant EpCAM enteroid model has been generated. The presence of mutant EpCAM in the model was confirmed at both mRNA and protein levels. Immunofluorescence microscopy demonstrated the reduced expression of mutant EpCAM. Mutant enteroids had reduced budding potential as well as significantly decreased mRNA expression for epithelial lineage markers (Mucin 2, lysozyme, sucrase-isomaltase), proliferation marker Ki67, and secretory pathway transcription factors (Atoh1, Hnf1b). Significantly decreased numbers of Paneth and goblet cells were confirmed by staining. These findings were correlated with intestinal tissue from CTE patients and the mutant mice model that had significantly fewer Paneth and goblet cells than in healthy counterparts. FITC-dextran studies demonstrated significantly impaired barrier function in monolayers derived from mutant enteroids compared with control monolayers. In conclusion, we have established an ex vivo CTE model. The role of EpCAM in the budding potential, differentiation, and barrier function of enteroids is noted. Our study establishes new facets of EpCAM biology that will aid in understanding the pathophysiology of CTE and role of EpCAM in health and disease.NEW & NOTEWORTHY Here, we develop a novel ex vivo enteroid model for congenital tufting enteropathy (CTE) based on epithelial cell adhesion molecule (EPCAM) gene mutations found in patients. With this model we demonstrate the role of EpCAM in maintaining the functional homeostasis of the intestinal epithelium, including differentiation, proliferation, and barrier integrity. This study further establishes a new direction in EpCAM biology that will help in understanding the detailed pathophysiology of CTE and role of EpCAM.
Collapse
Affiliation(s)
- Barun Das
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kevin Okamoto
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Rabalais
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Philip A. Kozan
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | | | - Matthew D. McGeough
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Nassim Durali
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Maria Go
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kim E. Barrett
- 2Department of Medicine, University of California, San Diego, La Jolla, California
| | - Soumita Das
- 3Department of Pathology, University of California, San Diego, La Jolla, California
| | - Mamata Sivagnanam
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California,4Rady Children’s Hospital, San Diego, California
| |
Collapse
|
80
|
Simbrunner B, Mandorfer M, Trauner M, Reiberger T. Gut-liver axis signaling in portal hypertension. World J Gastroenterol 2019; 25:5897-5917. [PMID: 31660028 PMCID: PMC6815800 DOI: 10.3748/wjg.v25.i39.5897] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/15/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023] Open
Abstract
Portal hypertension (PHT) in advanced chronic liver disease (ACLD) results from increased intrahepatic resistance caused by pathologic changes of liver tissue composition (structural component) and intrahepatic vasoconstriction (functional component). PHT is an important driver of hepatic decompensation such as development of ascites or variceal bleeding. Dysbiosis and an impaired intestinal barrier in ACLD facilitate translocation of bacteria and pathogen-associated molecular patterns (PAMPs) that promote disease progression via immune system activation with subsequent induction of proinflammatory and profibrogenic pathways. Congestive portal venous blood flow represents a critical pathophysiological mechanism linking PHT to increased intestinal permeability: The intestinal barrier function is affected by impaired microcirculation, neoangiogenesis, and abnormal vascular and mucosal permeability. The close bidirectional relationship between the gut and the liver has been termed “gut-liver axis”. Treatment strategies targeting the gut-liver axis by modulation of microbiota composition and function, intestinal barrier integrity, as well as amelioration of liver fibrosis and PHT are supposed to exert beneficial effects. The activation of the farnesoid X receptor in the liver and the gut was associated with beneficial effects in animal experiments, however, further studies regarding efficacy and safety of pharmacological FXR modulation in patients with ACLD are needed. In this review, we summarize the clinical impact of PHT on the course of liver disease, discuss the underlying pathophysiological link of PHT to gut-liver axis signaling, and provide insight into molecular mechanisms that may represent novel therapeutic targets.
Collapse
Affiliation(s)
- Benedikt Simbrunner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
- Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna 1180, Austria
| | - Mattias Mandorfer
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
- Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna 1180, Austria
| | - Michael Trauner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
| | - Thomas Reiberger
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
- Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna 1180, Austria
| |
Collapse
|
81
|
Celiac Disease and the Microbiome. Nutrients 2019; 11:nu11102403. [PMID: 31597349 PMCID: PMC6835875 DOI: 10.3390/nu11102403] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the hypothesis that changes in both the composition and function of the intestinal microbiome are associated with a number of chronic inflammatory diseases including celiac disease (CD). One of the major advances in the field of microbiome studies over the last few decades has been the development of culture-independent approaches to identify and quantify the components of the human microbiota. The study of nucleic acids DNA and RNA found in feces or other biological samples bypasses the need for tissue cultures and also allows the characterization of non-cultivable microbes. Current evidence on the composition of the intestinal microbiome and its role as a causative trigger for CD is highly heterogeneous and sometimes contradictory. This review is aimed at summarizing both pre-clinical (basic science data) and clinical (cross-sectional and prospective studies) evidence addressing the relationship between the intestinal microbiome and CD.
Collapse
|
82
|
The inflammatory event of birth: How oxytocin signaling may guide the development of the brain and gastrointestinal system. Front Neuroendocrinol 2019; 55:100794. [PMID: 31560883 DOI: 10.1016/j.yfrne.2019.100794] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/02/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023]
Abstract
The role of oxytocin (OT) as a neuropeptide that modulates social behavior has been extensively studied and reviewed, but beyond these functions, OT's adaptive functions at birth are quite numerous, as OT coordinates many physiological processes in the mother and fetus to ensure a successful delivery. In this review we explore in detail the potential adaptive roles of oxytocin as an anti-inflammatory, protective molecule at birth for the developing fetal brain and gastrointestinal system based on evidence that birth is a potent inflammatory/immune event. We discuss data with relevance for a number of neurodevelopmental disorders, as well as the emerging role of the gut-brain axis for health and disease. Finally, we discuss the potential relevance of sex differences in OT signaling present at birth in the increased male vulnerability to neurodevelopmental disabilities.
Collapse
|
83
|
Metabolically and immunologically beneficial impact of extra virgin olive and flaxseed oils on composition of gut microbiota in mice. Eur J Nutr 2019; 59:2411-2425. [PMID: 31506767 PMCID: PMC7413911 DOI: 10.1007/s00394-019-02088-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/30/2019] [Indexed: 12/30/2022]
Abstract
Purpose Extra virgin olive oil (EVOO) and flaxseed oil (FO) contain a variety of constituents beneficial for chronic inflammation and cardio-metabolic derangement. However, little is known about the impact of EVOO and FO on dysbiosis of gut microbiota, intestinal immunity, and barrier. We, therefore, aimed to assess the impact of EVOO and FO on gut microbiota, mucosal immunity, barrier integrity, and metabolic health in mice. Methods C57BL/6 J mice were exposed to a low-fat (LF), lard (HF), high fat-extra virgin olive oil (HF-EVOO), or high fat-flaxseed oil (HF-FO) diet for 10 weeks. Gut microbiota assessment was undertaken using 16S rRNA sequencing. Levels of mRNA for genes involved in intestinal inflammation and barrier maintenance in the intestine and bacterial infiltration in the liver were measured by qPCR. Results HF-EVOO or HF-FO mice showed greater diversity in gut microbiota as well as a lower abundance of the Firmicutes phylum in comparison with HF mice (P < 0.05). The qPCR analyses revealed that mRNA level of FoxP3, a transcription factor, and IL-10, an inducer of regulatory T cells, was significantly elevated in the intestines of mice-fed HF-EVOO in comparison with mice-fed HF (P < 0.05). The mRNA level of the antimicrobial peptide, RegӀӀӀγ, was markedly elevated in the intestines of HF-EVOO and HF-FO compared with HF group (P < 0.05). Conclusions Our data suggest that the consumption of EVOO or FO can beneficially impact gut microbiota, enhance gut immunity, and assist in the preservation of metabolic health in mice. Electronic supplementary material The online version of this article (10.1007/s00394-019-02088-0) contains supplementary material, which is available to authorized users.
Collapse
|
84
|
Abstract
An organ-specific chronic inflammation–remodeling–carcinoma sequence has been proposed, mainly for the alimentary tract. As representative diseases, gastroesophageal reflux disease, chronic gastritis and inflammatory bowel disease (ulcerative colitis and Crohn’s disease of the colitis type) were adopted for this discussion. Tissue remodeling is such an important part of tumorigenesis in this sequence that an organ-specific chronic inflammation–remodeling–carcinoma sequence has been proposed in detail. Chronic inflammation accelerates the cycle of tissue injury and regeneration; in other words, cell necrosis (or apoptosis) and proliferation result in tissue remodeling in long-standing cases of inflammation. Remodeling encompasses epithelial cell metaplasia and stromal fibrosis, and modifies epithelial–stromal cell interactions. Further, the accumulation of genetic, epigenetic and molecular changes—as well as morphologic disorganization—also occurs during tissue remodeling. The expression of mucosal tissue adapted to chronic inflammatory injury is thought to occur at an early stage. Subsequently, dysplasia and carcinoma develop on a background of remodeling due to continuous, active inflammation. Accordingly, organ-specific chronic inflammation should be ameliorated or well controlled with appropriate monitoring if complete healing is unachievable.
Collapse
|
85
|
Liu L, Guo Y, Zheng J, Lu Y, Shen Y, Huang C, Zeng Y, Wang X. Paneth cell ablation increases the small intestinal injury during acute necrotizing pancreatitis in rats. Mol Med Rep 2019; 20:473-484. [PMID: 31180547 PMCID: PMC6579996 DOI: 10.3892/mmr.2019.10274] [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: 09/03/2018] [Accepted: 03/29/2019] [Indexed: 01/11/2023] Open
Abstract
The present work aimed to investigate the role of Paneth cells in small intestinal injury during acute necrotizing pancreatitis (ANP) using rat models established by injection of dithizone, a metal chelator of zinc with the ability to selectively ablate Paneth cells. Sprague‑Dawley rats were randomly divided into four groups: Sham‑operated group, ANP group (3.5% sodium taurocholate solution, 1 ml/kg body weight), dithizone group (100 mg/kg of body weight) and ANP + dithizone group (sodium taurocholate solution was administered 6 h after dithizone injection). Each group was further divided into five subgroups (6, 12, 24, 36 and 48 h) based on the time period between induction of the model and sample collection. The present results suggested the number of Paneth cells was gradually decreased in the ANP group in a time‑dependent manner. Most of the Paneth cells were ablated in the ANP + dithizone group at 6 h, but a subset of Paneth cells recovered after 24‑48 h. Compared with the ANP group, combination of dithizone and ANP significantly induced more severe histopathological injuries in the pancreas and distal ileum, with higher Schmidt and Chiu's scores, respectively. Additionally, increased expression levels of tumor necrosis factor‑α (TNF‑α), interleukin (IL)‑1β and IL‑17A were detected in the ileum, causing an increase in intestinal permeability, as assessed by a decrease in the expression level of the intestinal tight junction protein occludin and high plasma levels of diamine oxidase and D‑lactate. The increase in intestinal permeability led to the translocation of bacteria to the bloodstream, triggering systemic inflammation, as assessed by the increased plasma levels of TNF‑α, IL‑1β and IL‑17A, reducing the survival rates of rats, which was 66.7% and 83.3% in the ANP + dithizone and the ANP group, respectively. The increase in intestinal endoplasmic reticulum stress, as assessed by high expression levels of binding‑immunoglobulin protein and activating transcription factor 6, may be one mechanism associated with Paneth cells loss and intestinal barrier impairment during ANP. Collectively, the present study suggested that the absence of Paneth cells may be an important factor involved in intestinal injury, promoting the progression of ANP.
Collapse
Affiliation(s)
- Liyan Liu
- International Medical Care Center, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Yuecheng Guo
- Department of Gastroenterology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Junyuan Zheng
- Department of Gastroenterology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Yingying Lu
- Department of Gastroenterology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Yucui Shen
- Department of Gastroenterology, Shanghai Fourth People's Hospital, Shanghai 200080, P.R. China
| | - Chunlan Huang
- Department of Gastroenterology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Yue Zeng
- Department of Gastroenterology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| |
Collapse
|
86
|
Vomhof-DeKrey EE, Lee J, Lansing J, Brown C, Darland D, Basson MD. Schlafen 3 knockout mice display gender-specific differences in weight gain, food efficiency, and expression of markers of intestinal epithelial differentiation, metabolism, and immune cell function. PLoS One 2019; 14:e0219267. [PMID: 31260507 PMCID: PMC6602453 DOI: 10.1371/journal.pone.0219267] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
Self-renewal and differentiation are essential for intestinal epithelium absorptive functioning and adaptation to pathological states such as short gut syndrome, ulcers, and inflammatory bowel disease. The rodent Slfn3 and its human analog Slfn12 are critical in regulating intestinal epithelial differentiation. We sought to characterize intestinal function in Slfn3 knockout (KO) mice. Male and female pair-fed Slfn3KO mice gained less weight with decreased food efficiency than wild type (WT) mice, with more pronounced effects in females. RNA sequencing performed on intestinal mucosa of Slfn3KO and WT mice showed gene ontology decreases in cell adhesion molecule signaling, tumor necrosis factor receptor binding, and adaptive immune cell proliferation/functioning genes in Slfn3KO mice, with greater effects in females. qPCR analysis of fatty acid metabolism genes, Pla2g4c, Pla2g2f, and Cyp3c55 revealed an increase in Pla2g4c, and a decrease in Pla2g2f in Slfn3KO females. Additionally, adipogenesis genes, Fabp4 and Lpl were decreased and ketogenesis gene Hmgcs2 was increased in female Slfn3KO mice. Sequencing did not reveal significant changes in differentiation markers, so qPCR was utilized. Slfn3KO tended to have decreased expression of intestinal differentiation markers sucrase isomaltase, dipeptidyl peptidase 4, villin 1, and glucose transporter 1 (Glut1) vs. WT males, although these trends did not achieve statistical significance unless data from several markers was pooled. Differentiation markers, Glut2 and sodium-glucose transporter 1 (SGLT1), did show statistically significant sex-dependent differences. Glut2 mRNA was reduced in Slfn3KO females, while SGLT1 increased in Slfn3KO males. Notch2 and Cdx2 were only increased in female Slfn3KO mice. Although Slfn3KO mice gain less weight and decreased food efficiency, their biochemical phenotype is more subtle and suggests a complex interplay between gender effects, Slfn3, and another regulatory pathway yet to be identified that compensates for the chronic loss of Slfn3.
Collapse
Affiliation(s)
- Emilie E. Vomhof-DeKrey
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, United States of America
| | - Jun Lee
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, United States of America
| | - Jack Lansing
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, United States of America
| | - Chris Brown
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, United States of America
| | - Diane Darland
- Department of Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Marc D. Basson
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, Grand Forks, ND, United States of America
| |
Collapse
|
87
|
Alcohol-induced IL-17A production in Paneth cells amplifies endoplasmic reticulum stress, apoptosis, and inflammasome-IL-18 activation in the proximal small intestine in mice. Mucosal Immunol 2019; 12:930-944. [PMID: 31105269 PMCID: PMC6599481 DOI: 10.1038/s41385-019-0170-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/05/2019] [Accepted: 04/13/2019] [Indexed: 02/06/2023]
Abstract
Gut microbial translocation contributes to alcoholic hepatitis. Using a mouse model of alcoholic hepatitis, we investigated the effects of chronic alcohol plus binge and found increased abundance of Paneth cells and IL-17A in the proximal small intestine (PSI). Alcohol increased IL-17A production and pro-apoptotic signaling evidenced by Bax, Bim, caspase-3, and caspase-8 increases via endoplasmic reticulum (ER) stress indicated by C/EBP homologous protein (CHOP) upregulation; this was prevented by the ER stress inhibitor, 4-PBA, in isolated crypts in vitro and in vivo. Mechanistically, IL-17 augmented alcohol-induced ER stress in isolated crypts. In vivo IL-17A blocking antibody administration in alcohol-treated mice attenuated ER stress-mediated apoptosis and IL-18 induction and prevented alcohol-induced impairment of tight junctions in the PSI and LPS translocation to the liver. Acute-on-chronic alcohol resulted in inflammasome activation, caspase-1 cleavage, and IL-18 production in the PSI. In vivo treatment with antibiotics or 4-PBA prevented CHOP upregulation and inflammasome activation. Our data suggest that alcohol upregulates innate immune mechanisms by increasing Paneth cell numbers and IL-17A release contributing to apoptosis amplification, inflammasome activation, and gut leakiness in the PSI. Binge alcohol-induced Paneth cell expansion, ER stress, and inflammasome activation in the PSI are modulated by the gut microbiome.
Collapse
|
88
|
Curciarello R, Canziani KE, Docena GH, Muglia CI. Contribution of Non-immune Cells to Activation and Modulation of the Intestinal Inflammation. Front Immunol 2019; 10:647. [PMID: 31024529 PMCID: PMC6467945 DOI: 10.3389/fimmu.2019.00647] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/11/2019] [Indexed: 12/27/2022] Open
Abstract
The mucosal immune system constitutes a physical and dynamic barrier against foreign antigens and pathogens and exerts control mechanisms to maintain intestinal tolerance to the microbiota and food antigens. Chronic alterations of the intestinal homeostasis predispose to inflammatory diseases of the gastrointestinal tract, such as Inflammatory Bowel Diseases (IBD). There is growing evidence that the frequency and severity of these diseases are increasing worldwide, which may be probably due to changes in environmental factors. Several stromal and immune cells are involved in this delicate equilibrium that dictates homeostasis. In this review we aimed to summarize the role of epithelial cells and fibroblasts in the induction of mucosal inflammation in the context of IBD. It has been extensively described that environmental factors are key players in this process, and the microbiome of the gastrointestinal tract is currently being intensively investigated due to its profound impact the immune response. Recent findings have demonstrated the interplay between dietary and environmental components, the gut microbiome, and immune cells. "Western" dietary patterns, such as high caloric diets, and pollution can induce alterations in the gut microbiome that in turn affect the intestinal and systemic homeostasis. Here we summarize current knowledge on the influence of dietary components and air particulate matters on gut microbiome composition, and the impact on stromal and immune cells, with a particular focus on promoting local inflammation.
Collapse
Affiliation(s)
- Renata Curciarello
- Instituto de Estudios Inmunológicos y Fisiopatológicos, CONICET, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.,Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Karina Eva Canziani
- Instituto de Estudios Inmunológicos y Fisiopatológicos, CONICET, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.,Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Guillermo Horacio Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos, CONICET, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.,Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Cecilia Isabel Muglia
- Instituto de Estudios Inmunológicos y Fisiopatológicos, CONICET, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.,Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
89
|
HDAC1 and HDAC2 independently regulate common and specific intrinsic responses in murine enteroids. Sci Rep 2019; 9:5363. [PMID: 30926862 PMCID: PMC6441098 DOI: 10.1038/s41598-019-41842-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023] Open
Abstract
Both HDAC1 and HDAC2 are class I deacetylases acting as erasers of lysine-acetyl marks on histones and non-histone proteins. Several histone deacetylase inhibitors, either endogenous to the cell, such as the ketogenic β-hydroxybutyrate metabolite, or exogenous, such as butyrate, a microbial-derived metabolite, regulate HDAC activity. Different combinations of intestinal epithelial cell (IEC)-specific Hdac1 and/or Hdac2 deletion differentially alter mucosal homeostasis in mice. Thus, HDAC1 and HDAC2 could act as sensors and transmitters of environmental signals to the mucosa. In this study, enteroid culture models deleted for Hdac1 or Hdac2 were established to determine IEC-specific function as assessed by global transcriptomic and proteomic approaches. Results show that Hdac1 or Hdac2 deficiency altered differentiation of Paneth and goblet secretory cells, which sustain physical and chemical protection barriers, and increased intermediate secretory cell precursor numbers. Furthermore, IEC Hdac1- and Hdac2-dependent common and specific biological processes were identified, including oxidation-reduction, inflammatory responses, and lipid-related metabolic processes, as well as canonical pathways and upstream regulators related to environment-dependent signaling through steroid receptor pathways, among others. These findings uncover unrecognized regulatory similarities and differences between Hdac1 and Hdac2 in IEC, and demonstrate how HDAC1 and HDAC2 may complement each other to regulate the intrinsic IEC phenotype.
Collapse
|
90
|
Abstract
Background The gut is hypothesized to be the “motor” of critical illness. Under basal conditions, the gut plays a crucial role in the maintenance of health. However, in critical illness, all elements of the gut are injured, potentially worsening multiple organ dysfunction syndrome. Main body Under basal conditions, the intestinal epithelium absorbs nutrients and plays a critical role as the first-line protection against pathogenic microbes and as the central coordinator of mucosal immunity. In contrast, each element of the gut is impacted in critical illness. In the epithelium, apoptosis increases, proliferation decreases, and migration slows. In addition, gut barrier function is worsened via alterations to the tight junction, resulting in intestinal hyperpermeability. This is associated with damage to the mucus that separates the contents of the intestinal lumen from the epithelium. Finally, the microbiome of the intestine is converted into a pathobiome, with an increase in disease-promoting bacteria and induction of virulence factors in commensal bacteria. Toxic factors can then leave the intestine via both portal blood flow and mesenteric lymph to cause distant organ damage. Conclusion The gut plays a complex role in both health and critical illness. Here, we review gut integrity in both health and illness and highlight potential strategies for targeting the intestine for therapeutic gain in the intensive care unit.
Collapse
Affiliation(s)
- Shunsuke Otani
- 1Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA 30322 USA.,2Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.,3Department of General Medical Science, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-8670 Japan
| | - Craig M Coopersmith
- 1Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA 30322 USA
| |
Collapse
|
91
|
Jones EJ, Matthews ZJ, Gul L, Sudhakar P, Treveil A, Divekar D, Buck J, Wrzesinski T, Jefferson M, Armstrong SD, Hall LJ, Watson AJM, Carding SR, Haerty W, Di Palma F, Mayer U, Powell PP, Hautefort I, Wileman T, Korcsmaros T. Integrative analysis of Paneth cell proteomic and transcriptomic data from intestinal organoids reveals functional processes dependent on autophagy. Dis Model Mech 2019; 12:dmm037069. [PMID: 30814064 PMCID: PMC6451430 DOI: 10.1242/dmm.037069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
Paneth cells are key epithelial cells that provide an antimicrobial barrier and maintain integrity of the small-intestinal stem cell niche. Paneth cell abnormalities are unfortunately detrimental to gut health and are often associated with digestive pathologies such as Crohn's disease or infections. Similar alterations are observed in individuals with impaired autophagy, a process that recycles cellular components. The direct effect of autophagy impairment on Paneth cells has not been analysed. To investigate this, we generated a mouse model lacking Atg16l1 specifically in intestinal epithelial cells, making these cells impaired in autophagy. Using three-dimensional intestinal organoids enriched for Paneth cells, we compared the proteomic profiles of wild-type and autophagy-impaired organoids. We used an integrated computational approach combining protein-protein interaction networks, autophagy-targeted proteins and functional information to identify the mechanistic link between autophagy impairment and disrupted pathways. Of the 284 altered proteins, 198 (70%) were more abundant in autophagy-impaired organoids, suggesting reduced protein degradation. Interestingly, these differentially abundant proteins comprised 116 proteins (41%) that are predicted targets of the selective autophagy proteins p62, LC3 and ATG16L1. Our integrative analysis revealed autophagy-mediated mechanisms that degrade key proteins in Paneth cell functions, such as exocytosis, apoptosis and DNA damage repair. Transcriptomic profiling of additional organoids confirmed that 90% of the observed changes upon autophagy alteration have effects at the protein level, not on gene expression. We performed further validation experiments showing differential lysozyme secretion, confirming our computationally inferred downregulation of exocytosis. Our observations could explain how protein-level alterations affect Paneth cell homeostatic functions upon autophagy impairment.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Emily J Jones
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Zoe J Matthews
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Lejla Gul
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
| | - Padhmanand Sudhakar
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
| | - Agatha Treveil
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
| | - Devina Divekar
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Jasmine Buck
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | | | - Matthew Jefferson
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Stuart D Armstrong
- National Institute of Health Research, University of Liverpool, Liverpool L3 5RF, UK
| | - Lindsay J Hall
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
| | - Alastair J M Watson
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Simon R Carding
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
| | | | - Ulrike Mayer
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Penny P Powell
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | | | - Tom Wileman
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
- Quadram Institute, Norwich Research Park, Norwich NR4 7UA, UK
| |
Collapse
|
92
|
Abstract
Necrotizing enterocolitis is a devastating intestinal disease affecting preterm infants. In spite of ongoing research and advancement in neonatal care, mortality remains high, especially in infants with advanced disease. The mechanism of disease development, the progression of intestinal injury, and management remain areas of ongoing research and controversy. In this review, we examine our current understanding of the disease, its epidemiology, the risk factors associated with the development of the disease, and its pathophysiology. We also describe current management and new emerging research highlighting potential future directions.
Collapse
Affiliation(s)
- Mashriq Alganabi
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carol Lee
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Edoardo Bindi
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
93
|
Jabari S, Schrödl F, Kaser-Eichberger A, Kofler B, Brehmer A. Alarin in different human intestinal epithelial cell types. Histochem Cell Biol 2019; 151:513-520. [PMID: 30612153 DOI: 10.1007/s00418-018-1763-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2018] [Indexed: 12/18/2022]
Abstract
Alarin (AL), a new member of the galanin family, has been localized in various CNS regions, mainly in rodents. Among other effects, it modulates food intake. Therefore, we analyzed the immunohistochemical distribution pattern of AL in human intestinal epithelia. Cryosections of 12 human bowel samples were immunohistochemically double-stained for AL and α-defensin 5 (αD; first set). Two further sets of sections were quadruple-stained either (second set) for AL, chromogranin (CG), synaptophysin (SY), and somatostatin (SO) or (third set) for AL, CG, Peptide Y (PY), and 5-hydroxytryptamine (5-HT). Slides were digitized and quantitative analysis of co-localization rates was undertaken. Small bowel: most of AL-positive cells (56%) were αD-positive Paneth cells located within the base of the crypts (first set). In the second set, about 27% of AL-labeled cells were co-reactive for SY and CG, likely representing entero-endocrine cells. In the third set, the largest subpopulation of AL-positive cells was not co-reactive for other markers applied (89%); most of them were likely Paneth cells. Large bowel: co-localization of AL with αD was not detected (first set). In the second set, AL was frequently co-localized with the other three markers applied (68%). In the third set, AL was frequently co-localized with 5-HT and CG (31%) as well as with PY and 5-HT (22%). Due to its presence in various enteroendocrine as well as Paneth cells, AL may be involved in different physiological and pathological processes.
Collapse
Affiliation(s)
- Samir Jabari
- Institute of Neuropathology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - Falk Schrödl
- Department of Ophthalmology/Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University, Salzburg, Austria.,Department of Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- Department of Ophthalmology/Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University, Salzburg, Austria.,Department of Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Axel Brehmer
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
94
|
Abstract
The intestinal epithelium withstands continuous mechanical, chemical and biological insults despite its single-layered, simple epithelial structure. The crypt-villus tissue architecture in combination with rapid cell turnover enables the intestine to act both as a barrier and as the primary site of nutrient uptake. Constant tissue replenishment is fuelled by continuously dividing stem cells that reside at the bottom of crypts. These cells are nurtured and protected by specialized epithelial and mesenchymal cells, and together constitute the intestinal stem cell niche. Intestinal stem cells and early progenitor cells compete for limited niche space and, therefore, the ability to retain or regain stemness. Those cells unable to do so differentiate to one of six different mature cell types and move upwards towards the villus, where they are shed into the intestinal lumen after 3-5 days. In this Review, we discuss the signals, cell types and mechanisms that control homeostasis and regeneration in the intestinal epithelium. We investigate how the niche protects and instructs intestinal stem cells, which processes drive differentiation of mature cells and how imbalance in key signalling pathways can cause human disease.
Collapse
|
95
|
Figueroa-Lozano S, de Vos P. Relationship Between Oligosaccharides and Glycoconjugates Content in Human Milk and the Development of the Gut Barrier. Compr Rev Food Sci Food Saf 2018; 18:121-139. [DOI: 10.1111/1541-4337.12400] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Susana Figueroa-Lozano
- Immunoendocrinology, Div. of Medical Biology, Dept. of Pathology and Medical Biology; Univ. of Groningen and University Medical Center Groningen; Groningen The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Div. of Medical Biology, Dept. of Pathology and Medical Biology; Univ. of Groningen and University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
96
|
Ma T, Suzuki Y, Guan LL. Dissect the mode of action of probiotics in affecting host-microbial interactions and immunity in food producing animals. Vet Immunol Immunopathol 2018; 205:35-48. [PMID: 30459000 DOI: 10.1016/j.vetimm.2018.10.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/29/2018] [Accepted: 10/16/2018] [Indexed: 02/07/2023]
Abstract
Prophylactic antimicrobials have been widely used in food animal production with the aim to prevent infectious diseases, enhance feed efficiency, and promote growth. However, the extensive use of antimicrobials in food animal production systems has led to the emergence of antimicrobial resistant pathogens, which are potential threats to human and animal health. Probiotics have been proposed to be a promising alternative of prophylactic antimicrobials, with potential beneficial effects on the host animal by improving the balance of intestinal microbiota and host immunity. Although an increasing body of evidence shows that probiotics could directly or indirectly affect gut microbiota and host immune functions, the lack of the understanding of how probiotics influence host-microbial interaction and immunity is one of the reasons for controversial findings from many animal trials, especially in food production animals. Therefore, in this review we focused on the most recent (last ten years) studies on how gut microbiota and host immune function changes in response to probiotics in food production animals (swine, poultry, and ruminant). In addition, the relationship between microbial changes and host immune function was illustrated, and how such relationship differs among animal species was further compared. Moreover, the future directions concerning the mechanisms of how probiotics modulate host-microbial interactions and host immunity were highlighted, which may assist in the optimal supplementation strategy to maximize the efficacy of probiotics to improve animal gut health and productivity.
Collapse
Affiliation(s)
- Tao Ma
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; Feed Research Institute, Chinese Academy of Agricultural Sciences, Key laboratory of Feed Biotechnology of the Ministry of Agriculture, Beijing, China
| | - Yutaka Suzuki
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; Laboratory of Animal Function and Nutrition, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
97
|
Rappaport JA, Waldman SA. The Guanylate Cyclase C-cGMP Signaling Axis Opposes Intestinal Epithelial Injury and Neoplasia. Front Oncol 2018; 8:299. [PMID: 30131940 PMCID: PMC6091576 DOI: 10.3389/fonc.2018.00299] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022] Open
Abstract
Guanylate cyclase C (GUCY2C) is a transmembrane receptor expressed on the luminal aspect of the intestinal epithelium. Its ligands include bacterial heat-stable enterotoxins responsible for traveler's diarrhea, the endogenous peptide hormones uroguanylin and guanylin, and the synthetic agents, linaclotide, plecanatide, and dolcanatide. Ligand-activated GUCY2C catalyzes the synthesis of intracellular cyclic GMP (cGMP), initiating signaling cascades underlying homeostasis of the intestinal epithelium. Mouse models of GUCY2C ablation, and recently, human populations harboring GUCY2C mutations, have revealed the diverse contributions of this signaling axis to epithelial health, including regulating fluid secretion, microbiome composition, intestinal barrier integrity, epithelial renewal, cell cycle progression, responses to DNA damage, epithelial-mesenchymal cross-talk, cell migration, and cellular metabolic status. Because of these wide-ranging roles, dysregulation of the GUCY2C-cGMP signaling axis has been implicated in the pathogenesis of bowel transit disorders, inflammatory bowel disease, and colorectal cancer. This review explores the current understanding of cGMP signaling in the intestinal epithelium and mechanisms by which it opposes intestinal injury. Particular focus will be applied to its emerging role in tumor suppression. In colorectal tumors, endogenous GUCY2C ligand expression is lost by a yet undefined mechanism conserved in mice and humans. Further, reconstitution of GUCY2C signaling through genetic or oral ligand replacement opposes tumorigenesis in mice. Taken together, these findings suggest an intriguing hypothesis that colorectal cancer arises in a microenvironment of functional GUCY2C inactivation, which can be repaired by oral ligand replacement. Hence, the GUCY2C signaling axis represents a novel therapeutic target for preventing colorectal cancer.
Collapse
Affiliation(s)
- Jeffrey A Rappaport
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, United States
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|