51
|
Ménard S, Lacroix-Lamandé S, Ehrhardt K, Yan J, Grassl GA, Wiedemann A. Cross-Talk Between the Intestinal Epithelium and Salmonella Typhimurium. Front Microbiol 2022; 13:906238. [PMID: 35733975 PMCID: PMC9207452 DOI: 10.3389/fmicb.2022.906238] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Salmonella enterica serovars are invasive gram-negative bacteria, causing a wide range of diseases from gastroenteritis to typhoid fever, representing a public health threat around the world. Salmonella gains access to the intestinal lumen after oral ingestion of contaminated food or water. The crucial initial step to establish infection is the interaction with the intestinal epithelium. Human-adapted serovars such as S. Typhi or S. Paratyphi disseminate to systemic organs and induce life-threatening disease known as typhoid fever, whereas broad-host serovars such as S. Typhimurium usually are limited to the intestine and responsible for gastroenteritis in humans. To overcome intestinal epithelial barrier, Salmonella developed mechanisms to induce cellular invasion, intracellular replication and to face host defence mechanisms. Depending on the serovar and the respective host organism, disease symptoms differ and are linked to the ability of the bacteria to manipulate the epithelial barrier for its own profit and cross the intestinal epithelium.This review will focus on S. Typhimurium (STm). To better understand STm pathogenesis, it is crucial to characterize the crosstalk between STm and the intestinal epithelium and decipher the mechanisms and epithelial cell types involved. Thus, the purpose of this review is to summarize our current knowledge on the molecular dialogue between STm and the various cell types constituting the intestinal epithelium with a focus on the mechanisms developed by STm to cross the intestinal epithelium and access to subepithelial or systemic sites and survive host defense mechanisms.
Collapse
Affiliation(s)
- Sandrine Ménard
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | | | - Katrin Ehrhardt
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Jin Yan
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Agnès Wiedemann
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- *Correspondence: Agnès Wiedemann,
| |
Collapse
|
52
|
Cangiano L, Villot C, Renaud J, Ipharraguerre I, McNeil B, DeVries T, Steele M. Induction of leaky gut by repeated intramuscular injections of indomethacin to preweaning Holstein calves. J Dairy Sci 2022; 105:7125-7139. [DOI: 10.3168/jds.2021-21768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/12/2022] [Indexed: 11/19/2022]
|
53
|
Zhao X, Wang Y, Yang Y, Pan Y, Liu J, Ge S. Association between preoperative nutritional status, inflammation, and intestinal permeability in elderly patients undergoing gastrectomy: a prospective cohort study. J Gastrointest Oncol 2022; 13:997-1006. [PMID: 35837193 DOI: 10.21037/jgo-22-367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/27/2022] [Indexed: 11/06/2022] Open
Abstract
Background Malnutrition is relatively common among elderly patients with gastric cancer. This study sought to analyze whether preoperative nutritional status, inflammatory cytokines, and intestinal permeability were correlated in elderly gastric cancer patients undergoing surgery, and their effects on postoperative recovery. Methods This study was a single-center prospective cohort study. Patients aged 65-90 years who underwent gastrectomy were included. Preoperative nutritional status was assessed by the Mini Nutritional Score (MNA), Nutritional Risk Screening-2002 (NRS2002), body mass index (BMI), free fatty acids (FFAs), albumin, and prealbumin concentrations. Intestinal permeability was assessed by D-lactate and intestinal fatty acid-binding protein (i-FABP). The inflammatory factors included interleukin (IL)-6, IL-10, neutrophil, and lymphocyte counts. The time to first defecation, time to first liquid diet, length of hospital stay (LOS), and postoperative complications were recorded. Results A total of 134 patients were included. According to the MNA, 50.7% and 32.1% of the cohort had mild to moderate malnutrition, and severe malnutrition, respectively. According to the NRS2002, 38.8% of the patients scored >4 points. I-FABP was significantly negatively correlated with albumin (r=-0.409, P<0.001) and prealbumin (r=-0.397, P<0.001), and significantly positively correlated with MNA (r=0.291, P=0.001), the NRS2002 (r=0.284, P=0.001), and LOS (r=0.245, P=0.004). D-lactate was significantly negatively correlated with BMI (r=-0.229, P=0.008), albumin (r=-0.426, P<0.001), and prealbumin (r=-0.358, P<0.001), and significantly positively correlated with the NRS2002 (r=0.187, P=0.030), time to first defecation (r=0.264, P=0.002), and LOS (r=0.409, P<0.001). There were significant differences in BMI, prealbumin, FFAs, i-FABP, time to first defecation, and time to first fluid diet (P<0.05) among groups based on MNA score. The multivariate logistic analysis showed that D-lactate was an independent risk factor of postoperative complications [odds ratio (OR) =1.354, 95% confidence interval (CI): 1.099-1.669, P=0.004]. Conclusions The preoperative intestinal permeability indicators (i.e., D-lactate and i-FABP) are significantly correlated with some nutritional indicators and postoperative recovery indicators. The preoperative D-lactate level is an independent risk factor of postoperative complications, suggesting that altered gut barrier function before surgery could to some extent influence postoperative recovery in the elderly.
Collapse
Affiliation(s)
- Xining Zhao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Ying Wang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yuying Yang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yan Pan
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jie Liu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shengjin Ge
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
54
|
Abstract
The immune system employs recognition tools to communicate with its microbial evolutionary partner. Among all the methods of microbial perception, T cells enable the widest spectrum of microbial recognition resolution, ranging from the crudest detection of whole groups of microbes to the finest detection of specific antigens. The application of this recognition capability to the crucial task of combatting infections has been the focus of classical immunology. We now appreciate that the coevolution of the immune system and the microbiota has led to development of a lush immunological decision tree downstream of microbial recognition, of which an inflammatory response is but one branch. In this review we discuss known T cell-microbe interactions in the gut and place them in the context of an algorithmic framework of recognition, context-dependent interpretation, and response circuits across multiple levels of microbial recognition resolution. The malleability of T cells in response to the microbiota presents an opportunity to edit immune response cellularity, identity, and functionality by utilizing microbiota-controlled pathways to promote human health.
Collapse
Affiliation(s)
- Ivaylo I Ivanov
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA;
| | - Timur Tuganbaev
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Ashwin N Skelly
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan;
| |
Collapse
|
55
|
Managlia E, Yan X, De Plaen IG. Intestinal Epithelial Barrier Function and Necrotizing Enterocolitis. NEWBORN 2022; 1:32-43. [PMID: 35846894 PMCID: PMC9286028 DOI: 10.5005/jp-journals-11002-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants. NEC is characterized by intestinal tissue inflammation and necrosis. The intestinal barrier is altered in NEC, which potentially contributes to its pathogenesis by promoting intestinal bacterial translocation and stimulating the inflammatory response. In premature infants, many components of the intestinal barrier are immature. This article reviews the different components of the intestinal barrier and how their immaturity contributes to intestinal barrier dysfunction and NEC.
Collapse
Affiliation(s)
- Elizabeth Managlia
- Division of Neonatology, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States; Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s Research Institute, Ann and Robert H Lurie Children’s Hospital of Chicago, Northwestern University, Chicago, Illinois, United States
| | - Xiaocai Yan
- Division of Neonatology, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States; Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s Research Institute, Ann and Robert H Lurie Children’s Hospital of Chicago, Northwestern University, Chicago, Illinois, United States
| | - Isabelle G De Plaen
- Division of Neonatology, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States; Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s Research Institute, Ann and Robert H Lurie Children’s Hospital of Chicago, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
56
|
Shevkani K, Singh N, Patil C, Awasthi A, Paul M. Antioxidative and antimicrobial properties of pulse proteins and their applications in gluten‐free foods and sports nutrition. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Khetan Shevkani
- Department of Applied Agriculture Central University of Punjab Bathinda 151401 India
| | - Narpinder Singh
- Department of Food Science and Technology Guru Nanak Dev University Amritsar 143005 India
| | - Chidanand Patil
- Department of Applied Agriculture Central University of Punjab Bathinda 151401 India
| | - Ankit Awasthi
- Department of Applied Agriculture Central University of Punjab Bathinda 151401 India
| | - Maman Paul
- Department of Physiotherapy Guru Nanak Dev University Amritsar 143005 India
| |
Collapse
|
57
|
Govers C, Calder PC, Savelkoul HFJ, Albers R, van Neerven RJJ. Ingestion, Immunity, and Infection: Nutrition and Viral Respiratory Tract Infections. Front Immunol 2022; 13:841532. [PMID: 35296080 PMCID: PMC8918570 DOI: 10.3389/fimmu.2022.841532] [Citation(s) in RCA: 1] [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: 12/22/2021] [Accepted: 02/02/2022] [Indexed: 12/12/2022] Open
Abstract
Respiratory infections place a heavy burden on the health care system, particularly in the winter months. Individuals with a vulnerable immune system, such as very young children and the elderly, and those with an immune deficiency, are at increased risk of contracting a respiratory infection. Most respiratory infections are relatively mild and affect the upper respiratory tract only, but other infections can be more serious. These can lead to pneumonia and be life-threatening in vulnerable groups. Rather than focus entirely on treating the symptoms of infectious disease, optimizing immune responsiveness to the pathogens causing these infections may help steer towards a more favorable outcome. Nutrition may have a role in such prevention through different immune supporting mechanisms. Nutrition contributes to the normal functioning of the immune system, with various nutrients acting as energy sources and building blocks during the immune response. Many micronutrients (vitamins and minerals) act as regulators of molecular responses of immune cells to infection. It is well described that chronic undernutrition as well as specific micronutrient deficiencies impair many aspects of the immune response and make individuals more susceptible to infectious diseases, especially in the respiratory and gastrointestinal tracts. In addition, other dietary components such as proteins, pre-, pro- and synbiotics, and also animal- and plant-derived bioactive components can further support the immune system. Both the innate and adaptive defense systems contribute to active antiviral respiratory tract immunity. The initial response to viral airway infections is through recognition by the innate immune system of viral components leading to activation of adaptive immune cells in the form of cytotoxic T cells, the production of neutralizing antibodies and the induction of memory T and B cell responses. The aim of this review is to describe the effects of a range different dietary components on anti-infective innate as well as adaptive immune responses and to propose mechanisms by which they may interact with the immune system in the respiratory tract.
Collapse
Affiliation(s)
- Coen Govers
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health Service (NHS) Foundation Trust and University of Southampton, Southampton, United Kingdom
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
| | | | - R. J. Joost van Neerven
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
- Research & Development, FrieslandCampina, Amersfoort, Netherlands
| |
Collapse
|
58
|
Han X, Guo J, Qin Y, Huang W, You Y, Zhan J. Dietary regulation of the SIgA-gut microbiota interaction. Crit Rev Food Sci Nutr 2022; 63:6379-6392. [PMID: 35125055 DOI: 10.1080/10408398.2022.2031097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gut microbiota (GM) is essential for host health, and changes in the GM are related to the development of various diseases. Recently, secretory immunoglobulin A (SIgA), the most abundant immunoglobulin isotype in the intestinal mucosa, has been found to play an essential role in controlling GM. SIgA dysfunction can lead to changes in the GM and is associated with the development of various GM-related diseases. Although in early stage, recent studies have shown that assorted dietary interventions, including vitamins, amino acids, fatty acids, polyphenols, oligo/polysaccharides, and probiotics, can influence the intestinal SIgA response and SIgA-GM interaction. Dietary intervention can enhance the SIgA response by directly regulating it (from top to bottom) or by regulating the GM structure or gene expression (from bottom to top). Furthermore, intensive studies involving the particular influence of dietary intervention on SIgA-binding to the GM and SIgA repertoire and the precise regulation of the SIgA response via dietary intervention are still exceedingly scarce and merit further consideration. This review summarizes the existing knowledge and (possible) mechanisms of the influence of dietary intervention on the SIgA-GM interaction. Key issues are considered, and the approaches in addressing these issues in future studies are also discussed.
Collapse
Affiliation(s)
- Xue Han
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing
| | - Jielong Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yue Qin
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Weidong Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yilin You
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jicheng Zhan
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
59
|
Affiliation(s)
- Michael Camilleri
- Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes, Metabolism, & Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
60
|
Implication of Intestinal Barrier Dysfunction in Gut Dysbiosis and Diseases. Biomedicines 2022; 10:biomedicines10020289. [PMID: 35203499 PMCID: PMC8869546 DOI: 10.3390/biomedicines10020289] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
The intestinal mucosal barrier, also referred to as intestinal barrier, is widely recognized as a critical player in gut homeostasis maintenance as it ensures the complex crosstalk between gut microbes (both commensals and pathogens) and the host immune system. Highly specialized epithelial cells constantly cope with several protective and harmful agents to maintain the multiple physiological functions of the barrier as well as its integrity. However, both genetic defects and environmental factors can break such equilibrium, thus promoting gut dysbiosis, dysregulated immune-inflammatory responses, and even the development of chronic pathological conditions. Here, we review and discuss the molecular and cellular pathways underlying intestinal barrier structural and functional homeostasis, focusing on potential alterations that may undermine this fine balance.
Collapse
|
61
|
Kurabi A, Pak K, Chavez E, Doan J, Ryan AF. A transcytotic transport mechanism across the tympanic membrane. Sci Rep 2022; 12:984. [PMID: 35046419 PMCID: PMC8770641 DOI: 10.1038/s41598-021-04748-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022] Open
Abstract
Drug treatments for middle ear diseases are currently delivered systemically, or locally after opening the impermeable tympanic membrane (TM). We previously used bacteriophage display to discover novel peptides that are actively transported across the intact TM, with a variety of transport rates. Peptide structures were analyzed for evidence regarding the mechanism for this unexpected transport, which was then tested by the application of chemical inhibitors. Primary sequences indicated that trans-TM peptides share one of two amino acid motifs. Secondary structures revealed that linear configurations associate with higher transport rates than coiled structures. Tertiary analysis indicated that the shared sequence motifs are prominently displayed at the free ends of rapidly transported peptide phage. The shared motifs were evaluated for similarity to known motifs. The highest probability matches were for protein motifs involved in transmembrane transport and exosomes. Overall, structural findings suggest that the shared motifs represent binding sequences. They also implicate transcytosis, a polarized cell transport mechanism consisting of endocytosis, transcellular transport, and exocytosis. Inhibitor studies indicated that macropinocytosis, retrograde transport through Golgi and exocytosis participate in transport across the TM, consistent with transcytosis. This process can be harnessed to noninvasively deliver therapeutics to the middle ear.
Collapse
Affiliation(s)
- Arwa Kurabi
- Department of Surgery/Otolaryngology, University of California, 9500 Gilman Drive, La Jolla, CA, 92093-0666, USA.
- San Diego VA Healthcare System, La Jolla, CA, USA.
| | - Kwang Pak
- Department of Surgery/Otolaryngology, University of California, 9500 Gilman Drive, La Jolla, CA, 92093-0666, USA
| | - Eduardo Chavez
- Department of Surgery/Otolaryngology, University of California, 9500 Gilman Drive, La Jolla, CA, 92093-0666, USA
| | - Jennifer Doan
- Department of Biology, University of California, San Diego, USA
| | - Allen F Ryan
- Department of Surgery/Otolaryngology, University of California, 9500 Gilman Drive, La Jolla, CA, 92093-0666, USA
- Department of Neurosciences, University of California, San Diego, USA
- San Diego VA Healthcare System, La Jolla, CA, USA
| |
Collapse
|
62
|
Tena-Garitaonaindia M, Arredondo-Amador M, Mascaraque C, Asensio M, Marin JJG, Martínez-Augustin O, Sánchez de Medina F. MODULATION OF INTESTINAL BARRIER FUNCTION BY GLUCOCORTICOIDS: LESSONS FROM PRECLINICAL MODELS. Pharmacol Res 2022; 177:106056. [PMID: 34995794 DOI: 10.1016/j.phrs.2022.106056] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/22/2021] [Accepted: 01/01/2022] [Indexed: 12/15/2022]
Abstract
Glucocorticoids (GCs) are widely used drugs for their anti-inflammatory and immunosuppressant effects, but they are associated with multiple adverse effects. Despite their frequent oral administration, relatively little attention has been paid to the effects of GCs on intestinal barrier function. In this review, we present a summary of the published studies on this matter carried out in animal models and cultured cells. In cultured intestinal epithelial cells, GCs have variable effects in basal conditions and generally enhance barrier function in the presence of inflammatory cytokines such as tumor necrosis factor (TNF). In turn, in rodents and other animals, GCs have been shown to weaken barrier function, with increased permeability and lower production of IgA, which may account for some features observed in stress models. When given to animals with experimental colitis, barrier function may be debilitated or strengthened, despite a positive anti-inflammatory activity. In sepsis models, GCs have a barrier-enhancing effect. These effects are probably related to the inhibition of epithelial cell proliferation and wound healing, modulation of the microbiota and mucus production, and interference with the mucosal immune system. The available information on underlying mechanisms is described and discussed.
Collapse
Affiliation(s)
- Mireia Tena-Garitaonaindia
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Arredondo-Amador
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Mascaraque
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Maitane Asensio
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
63
|
Gertie JA, Zhang B, Liu EG, Hoyt LR, Yin X, Xu L, Long LL, Soldatenko A, Gowthaman U, Williams A, Eisenbarth SC. Oral anaphylaxis to peanut in a mouse model is associated with gut permeability but not with Tlr4 or Dock8 mutations. J Allergy Clin Immunol 2022; 149:262-274. [PMID: 34051223 PMCID: PMC8626534 DOI: 10.1016/j.jaci.2021.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The etiology of food allergy is poorly understood; mouse models are powerful systems to discover immunologic pathways driving allergic disease. C3H/HeJ mice are a widely used model for the study of peanut allergy because, unlike C57BL/6 or BALB/c mice, they are highly susceptible to oral anaphylaxis. However, the immunologic mechanism of this strain's susceptibility is not known. OBJECTIVE We aimed to determine the mechanism underlying the unique susceptibility to anaphylaxis in C3H/HeJ mice. We tested the role of deleterious Toll-like receptor 4 (Tlr4) or dedicator of cytokinesis 8 (Dock8) mutations in this strain because both genes have been associated with food allergy. METHODS We generated C3H/HeJ mice with corrected Dock8 or Tlr4 alleles and sensitized and challenged them with peanut. We then characterized the antibody response to sensitization, anaphylaxis response to both oral and systemic peanut challenge, gut microbiome, and biomarkers of gut permeability. RESULTS In contrast to C3H/HeJ mice, C57BL/6 mice were resistant to anaphylaxis after oral peanut challenge; however, both strains undergo anaphylaxis with intraperitoneal challenge. Restoring Tlr4 or Dock8 function in C3H/HeJ mice did not protect from anaphylaxis. Instead, we discovered enhanced gut permeability resulting in ingested allergens in the bloodstream in C3H/HeJ mice compared to C57BL/6 mice, which correlated with an increased number of goblet cells in the small intestine. CONCLUSIONS Our work highlights the potential importance of gut permeability in driving anaphylaxis to ingested food allergens; it also indicates that genetic loci outside of Tlr4 and Dock8 are responsible for the oral anaphylactic susceptibility of C3H/HeJ mice.
Collapse
Affiliation(s)
- Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Singapore Immunology Network (SIgN), Singapore
| | - Elise G Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, Conn
| | - Laura R Hoyt
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Xiangyun Yin
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Lauren L Long
- The Jackson Laboratory for Genomic Medicine, Farmington, Conn
| | - Arielle Soldatenko
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Department of Pathology, University of Massachusetts Medical School, Worcester, Mass
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Farmington, Conn; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Conn.
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, Conn.
| |
Collapse
|
64
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
65
|
Wilson CG, Aarons L, Augustijns P, Brouwers J, Darwich AS, De Waal T, Garbacz G, Hansmann S, Hoc D, Ivanova A, Koziolek M, Reppas C, Schick P, Vertzoni M, García-Horsman JA. Integration of advanced methods and models to study drug absorption and related processes: An UNGAP perspective. Eur J Pharm Sci 2021; 172:106100. [PMID: 34936937 DOI: 10.1016/j.ejps.2021.106100] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.
Collapse
Affiliation(s)
- Clive G Wilson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, Glasgow, U.K.
| | | | | | | | | | | | | | | | | | | | - Mirko Koziolek
- NCE Formulation Sciences, Abbvie Deutschland GmbH & Co. KG, Germany
| | | | - Philipp Schick
- Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, Germany
| | | | | |
Collapse
|
66
|
Elz AS, Trevaskis NL, Porter CJH, Bowen JM, Prestidge CA. Smart design approaches for orally administered lipophilic prodrugs to promote lymphatic transport. J Control Release 2021; 341:676-701. [PMID: 34896450 DOI: 10.1016/j.jconrel.2021.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/22/2022]
Abstract
Challenges to effective delivery of drugs following oral administration has attracted growing interest over recent decades. Small molecule drugs (<1000 Da) are generally absorbed across the gastrointestinal tract into the portal blood and further transported to the systemic circulation via the liver. This can result in a significant reduction to the oral bioavailability of drugs that are metabolically labile and ultimately lead to ineffective exposure and treatment. Targeting drug delivery to the intestinal lymphatics is attracting increased attention as an alternative route of drug transportation providing multiple benefits. These include bypassing hepatic first-pass metabolism and selectively targeting disease reservoirs residing within the lymphatic system. The particular physicochemical requirements for drugs to be able to access the lymphatics after oral delivery include high lipophilicity (logP>5) and high long-chain triglyceride solubility (> 50 mg/g), properties required to enable drug association with the lipoprotein transport pathway. The majority of small molecule drugs, however, are not this lipophilic and therefore not substantially transported via the intestinal lymph. This has contributed to a growing body of investigation into prodrug approaches to deliver drugs to the lymphatic system by chemical manipulation. Optimised lipophilic prodrugs have the potential to increase lymphatic transport thereby improving oral pharmacokinetics via a reduction in first pass metabolism and may also target of disease-specific reservoirs within the lymphatics. This may provide advantages for current pharmacotherapy approaches for a wide array of pathological conditions, e.g. immune disease, cancer and metabolic disease, and also presents a promising approach for advanced vaccination strategies. In this review, specific emphasis is placed on medicinal chemistry strategies that have been successfully employed to design lipophilic prodrugs to deliberately enable lymphatic transport. Recent progress and opportunities in medicinal chemistry and drug delivery that enable new platforms for efficacious and safe delivery of drugs are critically evaluated.
Collapse
Affiliation(s)
- Aurelia S Elz
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Christopher J H Porter
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Joanne M Bowen
- School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
67
|
Lee Y, Kamada N, Moon JJ. Oral nanomedicine for modulating immunity, intestinal barrier functions, and gut microbiome. Adv Drug Deliv Rev 2021; 179:114021. [PMID: 34710529 PMCID: PMC8665886 DOI: 10.1016/j.addr.2021.114021] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract (GIT) affects not only local diseases in the GIT but also various systemic diseases. Factors that can affect the health and disease of both GIT and the human body include 1) the mucosal immune system composed of the gut-associated lymphoid tissues and the lamina propria, 2) the intestinal barrier composed of mucus and intestinal epithelium, and 3) the gut microbiota. Selective delivery of drugs, including antigens, immune-modulators, intestinal barrier enhancers, and gut-microbiome manipulators, has shown promising results for oral vaccines, immune tolerance, treatment of inflammatory bowel diseases, and other systemic diseases, including cancer. However, physicochemical and biological barriers of the GIT present significant challenges for successful translation. With the advances of novel nanomaterials, oral nanomedicine has emerged as an attractive option to not only overcome these barriers but also to selectively deliver drugs to the target sites in GIT. In this review, we discuss the GIT factors and physicochemical and biological barriers in the GIT. Furthermore, we present the recent progress of oral nanomedicine for oral vaccines, immune tolerance, and anti-inflammation therapies. We also discuss recent advances in oral nanomedicine designed to fortify the intestinal barrier functions and modulate the gut microbiota and microbial metabolites. Finally, we opine about the future directions of oral nano-immunotherapy.
Collapse
Affiliation(s)
- Yonghyun Lee
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea.
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA.
| |
Collapse
|
68
|
Barbara G, Barbaro MR, Fuschi D, Palombo M, Falangone F, Cremon C, Marasco G, Stanghellini V. Corrigendum: Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier. Front Nutr 2021; 8:790387. [PMID: 34790692 PMCID: PMC8591313 DOI: 10.3389/fnut.2021.790387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Affiliation(s)
- Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Marta Palombo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
69
|
Ilchmann-Diounou H, Buleon M, Bacquie V, Theodorou V, Denis C, Menard S. Revisiting definition and assessment of intestinal trans-epithelial passage. Cell Mol Life Sci 2021; 78:8157-8164. [PMID: 34731253 PMCID: PMC8629865 DOI: 10.1007/s00018-021-04000-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/27/2021] [Accepted: 10/18/2021] [Indexed: 10/26/2022]
Abstract
This study aims to remind that Intestinal Passage (IP) measurement is a complex task that cannot be achieved by a unique measure of an orally given exogenous marker in blood or urine. This will be illustrated in the case of NOD mice. Indeed, various methods have been proposed to measure IP. Among them ex vivo measurement in Ussing chambers of luminal to serosal fluxes of exogenous markers and in vivo measurement of exogenous markers in blood or urine after oral gavage are the more commonly used. Even though they are commonly used indifferently, they do not give the same information and can provide contradictory results. Published data showed that diabetic status in female Non Obese Diabetic (NOD) mice increased FD4 concentration in blood after gavage but did not modify FD4 fluxes in Ussing chamber. We observed the same results in our experimental conditions and tracked FD4 concentrations in blood over a kinetic study (Area Under the Curve-AUC). In vivo measurements are a dynamic process and address not only absorption (IP and intestinal surface) but also distribution, metabolism and excretion (ADME). Diabetic status in NOD mice was associated with an increase of intestinal length (absorptive surface), itself positively correlated with AUC of FD4 in blood. We concluded that increased intestinal length induced by diabetic status will extend the absorptive surface and increase FD4 concentration in plasma (in vivo measurement) despite no modification on IP of FD4 (ex vivo measurement). In addition, this study characterized intestinal function in diabetic NOD mice. Diabetic status in NOD female mice increases intestinal length and decreases paracellular IP (FSS) without affecting transcellular IP (HRP, FD4). Histological studies of small and large intestine did not show any modification of intestinal circumference nor villi and crypt size. Finally, diabetic status was not associated with intestinal inflammation (ELISA).
Collapse
Affiliation(s)
- Hanna Ilchmann-Diounou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Marie Buleon
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Disease, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Valérie Bacquie
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Vassilia Theodorou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Colette Denis
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Disease, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Sandrine Menard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France. .,IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, 31024, Toulouse, France.
| |
Collapse
|
70
|
Mu C, Hao X, Zhang X, Zhao J, Zhang J. Effects of high-concentrate diet supplemented with grape seed procyanidins on the colonic fermentation, colonic morphology, and inflammatory response in lambs. Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.115118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
71
|
Alizadeh A, Akbari P, Garssen J, Fink-Gremmels J, Braber S. Epithelial integrity, junctional complexes, and biomarkers associated with intestinal functions. Tissue Barriers 2021; 10:1996830. [PMID: 34719339 PMCID: PMC9359365 DOI: 10.1080/21688370.2021.1996830] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An intact intestinal barrier is crucial for immune homeostasis and its impairment activates the immune system and may result in chronic inflammation. The epithelial cells of the intestinal barrier are connected by tight junctions, which form an anastomosing network sealing adjacent epithelial cells. Tight junctions are composed of transmembrane and cytoplasmic scaffolding proteins. Transmembrane tight junction proteins at the apical-lateral membrane of the cell consist of occludin, claudins, junctional adhesion molecules, and tricellulin. Cytoplasmic scaffolding proteins, including zonula occludens, cingulin and afadin, provide a direct link between transmembrane tight junction proteins and the intracellular cytoskeleton. Each individual component of the tight junction network closely interacts with each other to form an efficient intestinal barrier. This review aims to describe the molecular structure of intestinal epithelial tight junction proteins and to characterize their organization and interaction. Moreover, clinically important biomarkers associated with impairment of gastrointestinal integrity are discussed.
Collapse
Affiliation(s)
- Arash Alizadeh
- Division of Pharmacology and Toxicology, Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Peyman Akbari
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Johanna Fink-Gremmels
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
72
|
Alotaibi BS, Buabeid M, Ibrahim NA, Kharaba ZJ, Ijaz M, Murtaza G. Recent strategies driving oral biologic administration. Expert Rev Vaccines 2021; 20:1587-1601. [PMID: 34612121 DOI: 10.1080/14760584.2021.1990044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION High patient compliance, noninvasiveness, and self-administration are the leading features of vaccine delivery through the oral route. The implementation of swift mass vaccination campaigns in pandemic outbreaks fascinates the use of oral vaccination. This approach can elicit both mucosal and systemic immune responses to protect against infection at the surface of the mucosa. AREA COVERED As pathogen entry and spread mainly occurs through the gastrointestinal tract (GIT) mucosal surfaces, oral vaccination may protect and limit disease spread. Oral vaccines target various potential mucosal inductive sites in the GIT, such as the oral cavity, gastric area, and small intestine. Orally delivered vaccines having subunit and nucleic acid pass through various GIT-associated risks, such as the biodegradation of biologics and their reduced absorption. This article presents a summarized review of the existing technologies and prospects for oral vaccination. EXPERT OPINION The intestinal mucosa focuses on current approaches, while future strategies target new mucosal sites, i.e. oral cavity and stomach. Recent developments in biologic delivery through the oral route and their potential use in future oral vaccination are mainly considered.
Collapse
Affiliation(s)
- Badriyah Shadid Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Nihal Abdalla Ibrahim
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore, 54000, Pakistan
| |
Collapse
|
73
|
Barbara G, Barbaro MR, Fuschi D, Palombo M, Falangone F, Cremon C, Marasco G, Stanghellini V. Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier. Front Nutr 2021; 8:718356. [PMID: 34589512 PMCID: PMC8475765 DOI: 10.3389/fnut.2021.718356] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
The intestinal epithelial barrier (IEB) is one of the largest interfaces between the environment and the internal milieu of the body. It is essential to limit the passage of harmful antigens and microorganisms and, on the other side, to assure the absorption of nutrients and water. The maintenance of this delicate equilibrium is tightly regulated as it is essential for human homeostasis. Luminal solutes and ions can pass across the IEB via two main routes: the transcellular pathway or the paracellular pathway. Tight junctions (TJs) are a multi-protein complex responsible for the regulation of paracellular permeability. TJs control the passage of antigens through the IEB and have a key role in maintaining barrier integrity. Several factors, including cytokines, gut microbiota, and dietary components are known to regulate intestinal TJs. Gut microbiota participates in several human functions including the modulation of epithelial cells and immune system through the release of several metabolites, such as short-chain fatty acids (SCFAs). Mediators released by immune cells can induce epithelial cell damage and TJs dysfunction. The subsequent disruption of the IEB allows the passage of antigens into the mucosa leading to further inflammation. Growing evidence indicates that dysbiosis, immune activation, and IEB dysfunction have a role in several diseases, including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and gluten-related conditions. Here we summarize the interplay between the IEB and gut microbiota and mucosal immune system and their involvement in IBS, IBD, and gluten-related disorders.
Collapse
Affiliation(s)
- Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Marta Palombo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
74
|
Moroni I, Garcia-Bennett AE. Effects of Absorption Kinetics on the Catabolism of Melatonin Released from CAP-Coated Mesoporous Silica Drug Delivery Vehicles. Pharmaceutics 2021; 13:1436. [PMID: 34575512 PMCID: PMC8464897 DOI: 10.3390/pharmaceutics13091436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
Melatonin (MLT) is a pineal hormone involved in the regulation of the sleep/wake cycle. The efficacy of exogenous MLT for the treatment of circadian and sleep disorders is variable due to a strong liver metabolism effect. In this work, MLT is encapsulated in mesoporous silica (AMS-6) with a loading capacity of 28.8 wt%, and the mesopores are blocked using a coating of cellulose acetate phthalate (CAP) at 1:1 and 1:2 AMS-6/MLT:CAP ratios. The release kinetics of MLT from the formulations is studied in simulated gastrointestinal fluids. The permeability of the MLT released from the formulations and its 6-hydroxylation are studied in an in vitro model of the intestinal tract (Caco-2 cells monolayer). The release of MLT from AMS-6/MLT:CAP 1:2 is significantly delayed in acidic environments up to 40 min, while remaining unaffected in neutral environments. The presence of CAP decreases the absorption of melatonin and increases its catabolism into 6-hydroxylation by the cytochrome P450 enzyme CYP1A2. The simple confinement of melatonin into AMS-6 pores slightly affects the permeability and significantly decreases melatonin 6-hydroxylation. Measurable amounts of silicon in the basolateral side of the Caco-2 cell monolayer might suggest the dissolution of AMS-6 during the experiment.
Collapse
Affiliation(s)
- Irene Moroni
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia;
| | - Alfonso E. Garcia-Bennett
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia;
- ARC Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
75
|
Kumar P, Kedaria D, Mahapatra C, Mohandas M, Chatterjee K. A designer cell culture insert with a nanofibrous membrane toward engineering an epithelial tissue model validated by cellular nanomechanics. NANOSCALE ADVANCES 2021; 3:4714-4725. [PMID: 36134314 PMCID: PMC9419865 DOI: 10.1039/d1na00280e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/04/2021] [Indexed: 05/13/2023]
Abstract
Engineered platforms for culturing cells of the skin and other epithelial tissues are useful for the regeneration and development of in vitro tissue models used in drug screening. Recapitulating the biomechanical behavior of the cells is one of the important hallmarks of successful tissue generation on these platforms. The biomechanical behavior of cells profoundly affects the physiological functions of the generated tissue. In this work, a designer nanofibrous cell culture insert (NCCI) device was developed, consisting of a free-hanging polymeric nanofibrous membrane. The free-hanging nanofibrous membrane has a well-tailored architecture, stiffness, and topography to better mimic the extracellular matrix of any soft tissue than conventional, flat tissue culture polystyrene (TCPS) surfaces. Human keratinocytes (HaCaT cells) cultured on the designer NCCIs exhibited a 3D tissue-like phenotype compared to the cells cultured on TCPS. Furthermore, the biomechanical characterization by bio-atomic force microscopy (Bio-AFM) revealed a markedly altered cellular morphology and stiffness of the cellular cytoplasm, nucleus, and cell-cell junctions. The nuclear and cytoplasmic moduli were reduced, while the stiffness of the cellular junctions was enhanced on the NCCI compared to cells on TCPS, which are indicative of the fluidic state and migratory phenotype on the NCCI. These observations were corroborated by immunostaining, which revealed enhanced cell-cell contact along with a higher expression of junction proteins and enhanced migration in a wound-healing assay. Taken together, these results underscore the role of the novel designer NCCI device as an in vitro platform for epithelial cells with several potential applications, including drug testing, disease modeling, and tissue regeneration.
Collapse
Affiliation(s)
- Prasoon Kumar
- Department of Materials Engineering, Indian Institute of Science C.V. Raman Avenue Bangalore 560012 India +91-80-22933408
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela 769008 India
| | - Dhaval Kedaria
- Department of Materials Engineering, Indian Institute of Science C.V. Raman Avenue Bangalore 560012 India +91-80-22933408
| | - Chinmaya Mahapatra
- Department of Materials Engineering, Indian Institute of Science C.V. Raman Avenue Bangalore 560012 India +91-80-22933408
- School of Chemical Engineering, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Monisha Mohandas
- Centre for BioSystems Science and Engineering, Indian Institute of Science C.V. Raman Avenue Bangalore 560012 India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science C.V. Raman Avenue Bangalore 560012 India +91-80-22933408
- Centre for BioSystems Science and Engineering, Indian Institute of Science C.V. Raman Avenue Bangalore 560012 India
| |
Collapse
|
76
|
Um CY, Hodge RA, Tran HQ, Campbell PT, Gewirtz AT, McCullough ML. Association of Emulsifier and Highly Processed Food Intake with Circulating Markers of Intestinal Permeability and Inflammation in the Cancer Prevention Study-3 Diet Assessment Sub-Study. Nutr Cancer 2021; 74:1701-1711. [PMID: 34353196 DOI: 10.1080/01635581.2021.1957947] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Compelling animal studies report increased intestinal permeability, inflammation, and colorectal carcinogenesis with exposure to certain emulsifiers commonly added to processed foods, but human data are lacking. Highly processed food consumption is also associated with obesity and higher risk of chronic diseases. We cross-sectionally examined the association of emulsifier and highly processed food consumption estimated from six 24-h dietary recalls among 588 U.S. men and women over one year, with biomarkers of intestinal permeability and inflammation measured from two fasting blood samples collected six months apart. In multivariable-adjusted generalized linear models, greater emulsifier intake (g/d) was not associated with antibodies to flagellin (P-trend = 0.88), lipopolysaccharide (LPS) (P-trend = 0.56), or the combined total thereof (P-trend = 0.65) but was positively associated with an inflammatory biomarker, glycoprotein acetyls (GlycA) (P-trend = 0.02). Highly processed food intake (% kcal/d) was associated with higher anti-LPS antibodies (P-trend = 0.001) and total anti-flagellin and anti-LPS antibodies (P-trend = 0.005) but not with other biomarkers, whereas processed food intake expressed as % g/d was associated with higher GlycA (P-trend = 0.02). Our findings suggest that, broadly, highly processed food consumption may be associated with intestinal permeability biomarkers, and both emulsifier and highly processed food intakes may be associated with inflammation. Additional studies are warranted to further evaluate these relationships.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1957947.
Collapse
Affiliation(s)
- Caroline Y Um
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Rebecca A Hodge
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Hao Q Tran
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Peter T Campbell
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | |
Collapse
|
77
|
Xu Z, Chen W, Deng Q, Huang Q, Wang X, Yang C, Huang F. Flaxseed oligosaccharides alleviate DSS-induced colitis through modulation of gut microbiota and repair of the intestinal barrier in mice. Food Funct 2021; 11:8077-8088. [PMID: 32856645 DOI: 10.1039/d0fo01105c] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intestinal epithelial barrier dysfunction with dysbiosis of gut microbiota contributes to the occurrence and acceleration of colitis. This study aimed to evaluate the effect of flaxseed oligosaccharides (FOSs) on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mice and to elucidate the underlying mechanisms. UC was induced in mice by administering 2% DSS in drinking water for 8 days. Then, FOS (50 mg kg-1 d-1, 100 mg kg-1 d-1 and 200 mg kg-1 d-1) was administered by gavage for 14 days. The results showed that FOS treatment (200 mg kg-1 d-1) significantly ameliorated colitis by decreasing disease activity index (DAI), increasing colon length and improving colonic histology. FOS treatment (200 mg kg-1 d-1) down-regulated the critical markers of oxidative stresses, including malondialdehyde (MDA) and myeloperoxidase (MPO). Furthermore, FOS (200 mg kg-1 d-1) significantly suppressed the levels of pro-inflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and interleukin (IL)-1β but increased that of anti-inflammatory cytokine interleukin (IL)-10. The 16S rDNA gene high-throughput sequencing results indicated that FOS treatment increased the gut microbial diversity and inhibited the proliferation of inflammation-related bacteria such as unidentified_Clostridiales. An increase in total short-chain fatty acids (SCFAs), propionic acid and butyric acid, was also observed by FOS supplementation. FOS (200 mg kg-1d-1) also protected the intestinal barrier by increasing the protein levels of Claudin1 and Occludin. In conclusion, FOS attenuated DSS-induced colitis by modulating the gut microbiota and repairing the intestinal barrier.
Collapse
Affiliation(s)
- Zhenxia Xu
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China.
| | - Wenchao Chen
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China.
| | - Qianchun Deng
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China.
| | - Qingde Huang
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China.
| | - Xu Wang
- Huazhong Agricultural University, No. 1 Shizishan Street, Wuhan 430070, China
| | - Chen Yang
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China.
| | - Fenghong Huang
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, No. 2 Xudong 2nd Road, Wuhan 430062, China. and Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan 250100, China
| |
Collapse
|
78
|
Usuda H, Okamoto T, Wada K. Leaky Gut: Effect of Dietary Fiber and Fats on Microbiome and Intestinal Barrier. Int J Mol Sci 2021; 22:ijms22147613. [PMID: 34299233 PMCID: PMC8305009 DOI: 10.3390/ijms22147613] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Intestinal tract is the boundary that prevents harmful molecules from invading into the mucosal tissue, followed by systemic circulation. Intestinal permeability is an index for intestinal barrier integrity. Intestinal permeability has been shown to increase in various diseases-not only intestinal inflammatory diseases, but also systemic diseases, including diabetes, chronic kidney dysfunction, cancer, and cardiovascular diseases. Chronic increase of intestinal permeability is termed 'leaky gut' which is observed in the patients and animal models of these diseases. This state often correlates with the disease state. In addition, recent studies have revealed that gut microbiota affects intestinal and systemic heath conditions via their metabolite, especially short-chain fatty acids and lipopolysaccharides, which can trigger leaky gut. The etiology of leaky gut is still unknown; however, recent studies have uncovered exogenous factors that can modulate intestinal permeability. Nutrients are closely related to intestinal health and permeability that are actively investigated as a hot topic of scientific research. Here, we will review the effect of nutrients on intestinal permeability and microbiome for a better understanding of leaky gut and a possible mechanism of increase in intestinal permeability.
Collapse
Affiliation(s)
- Haruki Usuda
- Correspondence: (H.U.); (T.O.); Tel.: +81-853-20-3067 (H.U.)
| | | | | |
Collapse
|
79
|
Krasulova K, Illes P. Intestinal interplay of quorum sensing molecules and human receptors. Biochimie 2021; 189:108-119. [PMID: 34186126 DOI: 10.1016/j.biochi.2021.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/03/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022]
Abstract
Human gut is in permanent contact with microorganisms that play an important role in many physiological processes including metabolism and immunologic activity. These microorganisms communicate and manage themself by the quorum sensing system (QS) that helps to coordinate optimal growth and subsistence by activating signaling pathways that regulate bacterial gene expression. Diverse QS molecules produced by pathogenic as well as resident microbiota have been found throughout the human gut. However, even a host can by affected by these molecules. Intestinal and immune cells possess a range of molecular targets for QS. Our present knowledge on bacteria-cell communication encompasses G-protein-coupled receptors, nuclear receptors and receptors for bacterial cell-wall components. The QS of commensal bacteria has been approved as a protective factor with favourable effects on intestinal homeostasis and immunity. Signaling molecules of QS interacting with above-mentioned receptors thus parcipitate on maintaining of barrier functions, control of inflammation processes and increase of resistance to pathogen colonization in host organisms. Pathogens QS molecules can have a dual function. Host cells are able to detect the ongoing infection by monitoring the presence and changes in concentrations of QS molecules. Such information can help to set the most effective immune defence to prevent or overcome the infection. Contrary, pathogens QS signals can target the host receptors to deceive the immune system to get the best conditions for growth. However, our knowledge about communication mediated by QS is still limited and detailed understanding of molecular mechanisms of QS signaling is desired.
Collapse
Affiliation(s)
- Kristyna Krasulova
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic.
| | - Peter Illes
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic
| |
Collapse
|
80
|
Fan J, Zhao XH, Zhao JR, Li BR. Galangin and Kaempferol Alleviate the Indomethacin-Caused Cytotoxicity and Barrier Loss in Rat Intestinal Epithelial (IEC-6) Cells Via Mediating JNK/Src Activation. ACS OMEGA 2021; 6:15046-15056. [PMID: 34151085 PMCID: PMC8210432 DOI: 10.1021/acsomega.1c01167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/21/2021] [Indexed: 05/16/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) like indomethacin and others are widely used in clinics, but they have the potential to cause severe gastrointestinal damage including intestinal barrier dysfunction. Thus, two flavonols galangin and kaempferol with or without heat treatment (100 °C, 30 min) were assessed for their effect on indomethacin-damaged rat intestine epithelial (IEC-6) cells. In total, the cell exposure of 300 μmol/L indomethacin for 24 h caused cell toxicity efficiently, resulting in decreased cell viability, enhanced lactate dehydrogenase (LDH) release or reactive oxygen species (ROS) production, and obvious barrier loss. Meanwhile, pretreatment of the cells with these flavonols for 24 and 48 h before the indomethacin exposure could alleviate cytotoxicity and especially barrier loss, resulting in increased cell viability and transepithelial resistance, decreased LDH release, ROS production, and paracellular permeability, together with the promoted expression of three tight junction proteins zonula occluden-1, occludin, and claudin-1. Moreover, the intracellular Ca2+ concentration and expression levels of p-JNK and p-Src arisen from the indomethacin damage were also reduced by the flavonols, suggesting an inhibited calcium-mediated JNK/Src activation. Consistently, galangin showed higher activity than kaempferol to the cells, while the heated flavonols were less efficient than the unheated counterparts. It is thus highlighted that the two flavonols could alleviate indomethacin cytotoxicity and combat against the indomethacin-induced barrier loss in IEC-6 cells, but heat treatment of the flavonols would weaken the two beneficial functions.
Collapse
Affiliation(s)
- Jing Fan
- School
of Biological and Food Engineering, Guangdong
University of Petrochemical Technology, 525000 Maoming, Guangdong, P. R. China
- Key
Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, 150030 Harbin, P. R. China
| | - Xin-Huai Zhao
- School
of Biological and Food Engineering, Guangdong
University of Petrochemical Technology, 525000 Maoming, Guangdong, P. R. China
- Maoming
Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong University of Petrochemical Technology, 525000 Maoming, P. R. China
| | - Jun-Ren Zhao
- School
of Biological and Food Engineering, Guangdong
University of Petrochemical Technology, 525000 Maoming, Guangdong, P. R. China
| | - Bai-Ru Li
- School
of Biological and Food Engineering, Guangdong
University of Petrochemical Technology, 525000 Maoming, Guangdong, P. R. China
| |
Collapse
|
81
|
Advances in Intestinal Barrier Preservation and Restoration in the Allogeneic Hematopoietic Cell Transplantation Setting. J Clin Med 2021; 10:jcm10112508. [PMID: 34204044 PMCID: PMC8201017 DOI: 10.3390/jcm10112508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
The intestinal barrier consists of an epithelial lining covered with specialized mucus inhabited by intestinal microbiota. An intact gut barrier ensures a resistance to bacteria and toxins translocation. On the other hand, gut permeability allows the absorption of essential nutrients, fluids and ions. This balance is achieved only by the complex structure and functional characteristics of the intestinal barrier. Allogenic hematopoietic cell transplantation remains the only curative treatment for many hematological diseases, but its application is limited because of possible transplant-related mortality mainly due to graft-versus-host disease and infectious complications. The intestinal barrier has been extensively studied in recent years as the primary site of graft-versus-host disease initiation and propagation. In the present review, we focused on the physiological structure and function of the gut barrier and the evidence of how the disruption of the gut barrier and increased intestinal permeability affects transplant recipients. Finally, therapeutic strategies aiming at intestinal barrier protection with a special focus on microbiome preservation and restoration in the allogenic hematopoietic cell transplantation setting are discussed.
Collapse
|
82
|
Mulberroside A from Cortex Mori Enhanced Gut Integrity in Diabetes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6655555. [PMID: 34104203 PMCID: PMC8159636 DOI: 10.1155/2021/6655555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/07/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022]
Abstract
Background Diabetic endotoxemia has been recognized as one of the hallmarks of type 2 diabetes mellitus (T2DM). Recent findings suggest that gut leak plays a pivotal role in diabetic endotoxemia. Cortex Mori (CM) has been widely applied in China to ameliorate development of T2DM, but its effect on endotoxemia is unknown. Methods The study was constructed with two parts: (1) in vivo study of CM on diabetic endotoxemia in db/db mice. Eight C57BL/6 mice were set as normal control; (2) in vitro study of mulberroside A (MBA) from CM on diabetic endotoxemia. Potential mechanism of MBA on ameliorating diabetic endotoxemia was also explored. Results The present study found that CM water extract decreased levels of blood glucose, ameliorated liver and renal damage in db/db mice, and ameliorated diabetic endotoxemia (p < 0.01). We also found that the water extract enhanced gut integrity and decreased gut inflammatory protein ICAM-1 expression in db/db mice as detected by H&E staining and immunohistochemistry methods. In the in vitro study, MBA decreased levels of MDA and ROS induced by LPS (p < 0.01) and enhanced the integrity of gut epithelial barrier (p < 0.01). Conclusions We found that Cortex Mori and its active component mulberroside A could ameliorate diabetic endotoxemia by preserving gut integrity.
Collapse
|
83
|
Lerner A. The intestinal luminal sources of α-synuclein: a gastroenterologist perspective. Nutr Rev 2021; 80:282-293. [PMID: 33942062 DOI: 10.1093/nutrit/nuab024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease is characterized by nonmotor/motor dysfunction, midbrain dopaminergic neuronal death, and α-synuclein (aSN) deposits. The current hypothesis is that aSN accumulates in the enteric nervous system to reach the brain. However, invertebrate, vertebrate, and nutritional sources of aSN reach the luminal compartment. Submitted to local amyloidogenic forces, the oligomerized proteins' cargo can be sensed and sampled by a specialized mucosal cell to be transmitted to the adjacent enteric nervous system, starting their upward journey to the brain. The present narrative review extends the current mucosal origin of Parkinson's disease, presenting the possibility that the disease starts in the intestinal lumen. If substantiated, eliminating the nutritional sources of aSN (eg, applying a vegetarian diet) might revolutionize the currently used dopaminergic pharmacologic therapy.
Collapse
Affiliation(s)
- Aaron Lerner
- A. Lerner is with the Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
84
|
Nagu P, Parashar A, Behl T, Mehta V. Gut Microbiota Composition and Epigenetic Molecular Changes Connected to the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2021; 71:1436-1455. [PMID: 33829390 DOI: 10.1007/s12031-021-01829-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and its pathogenesis is not fully known. Although there are several hypotheses, such as neuroinflammation, tau hyperphosphorylation, amyloid-β plaques, neurofibrillary tangles, and oxidative stress, none of them completely explain the origin and progression of AD. Emerging evidence suggests that gut microbiota and epigenetics can directly influence the pathogenesis of AD via their effects on multiple pathways, including neuroinflammation, oxidative stress, and amyloid protein. Various gut microbes such as Actinobacteria, Bacteroidetes, E. coli, Firmicutes, Proteobacteria, Tenericutes, and Verrucomicrobia are known to play a crucial role in the pathogenesis of AD. These microbes and their metabolites modulate various physiological processes that contribute to AD pathogenesis, such as neuroinflammation and other inflammatory processes, amyloid deposition, cytokine storm syndrome, altered BDNF and NMDA signaling, impairing neurodevelopmental processes. Likewise, epigenetic markers associated with AD mainly include histone modifications and DNA methylation, which are under the direct control of a variety of enzymes, such as acetylases and methylases. The activity of these enzymes is dependent upon the metabolites generated by the host's gut microbiome, suggesting the significance of epigenetics in AD pathogenesis. It is interesting to know that both gut microbiota and epigenetics are dynamic processes and show a high degree of variation according to diet, stressors, and environmental factors. The bidirectional relation between the gut microbiota and epigenetics suggests that they might work in synchrony to modulate AD representation, its pathogenesis, and progression. They both also provide numerous targets for early diagnostic biomarkers and for the development of AD therapeutics. This review discusses the gut microbiota and epigenetics connection in the pathogenesis of AD and aims to highlight vast opportunities for diagnosis and therapeutics of AD.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmaceutics, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.,Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Arun Parashar
- Faculty of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.
| |
Collapse
|
85
|
Villard A, Boursier J, Andriantsitohaina R. Microbiota-derived extracellular vesicles and metabolic syndrome. Acta Physiol (Oxf) 2021; 231:e13600. [PMID: 33319492 DOI: 10.1111/apha.13600] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
AIM Metabolic syndrome is a major health problem concerning approximately 25% of worldwide population. Metabolic syndrome regroups a cluster of five metabolic abnormalities predisposing to Type 2 Diabetes mellitus. Dysbiotic gut microbiota is accompanied by an increase of both intestinal permeability and pathogen-associated molecular patterns translocation into blood circulation to induce metabolic endotoxemia responsible for the low-grade systemic inflammation and insulin resistance in metabolic syndrome. Among pathogen-associated molecular patterns, bacterial extracellular vesicles are gaining growing attention. The latter are produced by eukaryotic and prokaryotic cells and are vectors of communication between gut microbiota and its host The present review brings evidence to the importance of the control of the balance between the different subsets of gut microbiota in the development of metabolic diseases including metabolic syndrome. RESULTS The ability of bacteria, including gut bacteria, to release extracellular vesicles implicated in host metabolic homeostasis is highlighted with their plethora of actions on intestinal barrier, inflammation and insulin resistance. CONCLUSION Bacterial extracellular vesicles can be considered as key players in the pathophysiological of metabolic diseases and may represent an interesting strategy for specific manipulations of microbiome for promoting host health.
Collapse
Affiliation(s)
- Alexandre Villard
- INSERM UMR1063 Stress Oxydant et Pathologies Métaboliques Faculté de Santé Université d’AngersUniversité Bretagne Loire Angers France
- Hémodynamique Interaction Fibrose et Invasivité Tumorales Hépatiques (HIFIH) Angers France
| | - Jérôme Boursier
- Hémodynamique Interaction Fibrose et Invasivité Tumorales Hépatiques (HIFIH) Angers France
| | - Ramaroson Andriantsitohaina
- INSERM UMR1063 Stress Oxydant et Pathologies Métaboliques Faculté de Santé Université d’AngersUniversité Bretagne Loire Angers France
| |
Collapse
|
86
|
Balamtekin N, Erdal H, Gençkardeşler E, Arslan M. Reliability of anti-tissue transglutaminase antibodies in children with malnutrition. Scand J Gastroenterol 2021; 56:378-381. [PMID: 33590788 DOI: 10.1080/00365521.2021.1882554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Serological markers are used in the diagnosis of celiac disease. Among these, the most widely used are tissue transglutaminase antibodies (anti-TG2 antibodies). It has been suggested that the mechanisms that are set in motion by malnutrition cause the tight connections between enterocytes to expand, which allows gluten-derived peptides to pass through the epithelium. This causes the production of anti-TG2 antibodies without the presence of celiac disease. METHODS The patients who were examined for malnutrition and had their anti-TG2 antibody levels measured at the same time, were accepted into the study. The patients who were investigated for suspected celiac disease, showed no signs of malnutrition, and had their anti-TG2 antibody levels measured were accepted into a control group. RESULTS The study population consisted of 126 children with mild malnutrition (54.8% female, 7.44 ± 5.38 years); 89 children with moderate malnutrition (54.8% female, 7.62 ± 5.43 years), and a control group of 200 children (53.2% female, 7.72 ± 5.05 years). According to the results, anti-TG2 IgG levels were significantly higher among patients in the mild and moderate malnutrition groups compared to patients in the control group (p = .02 and p = .01, respectively). However, there was no significant difference between the mild and moderate malnutrition groups (p > .05). CONCLUSIONS Malnutrition does not affect anti-TG2 IgA levels in children. However, anti-TG2 IgG levels increase in children suffering from malnutrition. When examining celiac disease, especially in people with a background IgA deficiency, doctors should consider whether malnutrition may be the cause of the increase in serum anti-TG2 IgG levels without celiac disease.
Collapse
Affiliation(s)
- Necati Balamtekin
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Turkey
| | - Harun Erdal
- Department of Internal Medicine, Division of Gastroenterology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Turkey
| | - Emira Gençkardeşler
- Department of Pediatrics, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Turkey
| | - Melike Arslan
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
87
|
Roos Ljungberg K, Martinsson K, Wetterö J, Svärd A, Kastbom A. Circulating anti-citrullinated protein antibodies containing secretory component are prognostic for arthritis onset in at-risk patients. Clin Exp Immunol 2021; 204:344-351. [PMID: 33675063 PMCID: PMC8119868 DOI: 10.1111/cei.13591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022] Open
Abstract
Autoantibodies related to rheumatoid arthritis (RA), such as anti‐citrullinated protein antibodies (ACPA), are often detectable in the preclinical period years before arthritis onset. However, events triggering arthritis development remain incompletely known. We aimed to determine whether ACPA isotype levels are prognostic for arthritis development in patients presenting with immunoglobulin (Ig)G ACPA and musculoskeletal pain. Study participants (n = 82) had musculoskeletal pain of any sort and duration and a positive IgG ACPA test. None of the patients had arthritis upon clinical examination at baseline, but during follow‐up (mean = 6 years), 48% developed at least one arthritic joint. IgG, IgA, IgM and secretory component (SC)‐containing ACPA was measured in longitudinally collected serum samples. Cox regression analysis was performed to test the prognostic value of baseline antibody levels and changes over time. All analysed ACPA isotype levels were associated with arthritis development in univariable Cox regression analysis. In multivariable analysis, baseline SC ACPA levels were independently prognostic for arthritis development in multivariable analysis [hazard ratio (HR) = 1·006, 95% confidence interval (CI) = 1·001–1·010, P = 0·012]. There were no significant changes in ACPA isotype levels over time, and no significant association between changes over time and arthritis development. In this prospective longitudinal study, baseline serum SC ACPA levels, but neither IgG, IgA nor IgM ACPA are prognostic for future arthritis development. Repeated measurement of ACPA isotypes do not bring additional prognostic value. The results reinforce a mucosal connection in RA development and encourage further exploration of the mechanisms underlying secretory ACPA formation as a trigger for arthritis development.
Collapse
Affiliation(s)
- K Roos Ljungberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Center for Clinical Research Dalarna, Uppsala University, Uppsala, Sweden
| | - K Martinsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - J Wetterö
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - A Svärd
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Center for Clinical Research Dalarna, Uppsala University, Uppsala, Sweden
| | - A Kastbom
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Rheumatology in Östergötland, Linköping University Hospital, Linköping, Sweden
| |
Collapse
|
88
|
Mokhtar DM, Abdelhafez EA. An overview of the structural and functional aspects of immune cells in teleosts. Histol Histopathol 2021; 36:399-414. [PMID: 33415722 DOI: 10.14670/hh-18-302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The immune system of fish consists of two main components, innate and adaptive immunities. Innate immunity is non-specific and acts as the primary line of protection against pathogen invasion, while adaptive immunity is more specific to a certain pathogen/following adaptation. The adaptive immune system consists of the humoral and cellular components. Cytotoxic T-lymphocyte cells are the major component of the cellular immunity that frequently kills viral-, bacterial- or parasitic-infected cells. According to the anatomical location, the mucosal-associated lymphoid tissue (MALT) in teleost fish subdivides into gut-associated lymphoid tissue (GALT), gill-associated lymphoid tissue (GIALT), and skin-associated lymphoid tissue (SALT). The MALTs contain various leukocytes; including, but not limited to, lymphocytes (T and B cells), plasma cells, macrophages, and granulocytes. Macrophages are multifunctional cells that are mainly involved in the immune response, including; phagocytosis and degradation of foreign antigens, tissue remodeling, and production of cytokines, chemokines and growth factors. An interesting feature of teleost macrophages is their ability to form melanomacrophage centers (MMC) in the hemopoietic tissues. Dendritic cells, rodlet cells, mast cells, eosinophilic granular cells (ECGs), telocytes, osteoclasts, club cells, as well as, barrier cells have been recorded in many fish species and have many immunological roles. This paper aims to summarize the current knowledge of the immune cells present in fish tissues serving as anatomical and physiological barriers against external hazards. Increased knowledge of fish immune systems will facilitate the development of novel vaccination strategies in fish.
Collapse
Affiliation(s)
- Doaa M Mokhtar
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt.
| | - Enas A Abdelhafez
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
89
|
Coffey JW, Gaiha GD, Traverso G. Oral Biologic Delivery: Advances Toward Oral Subunit, DNA, and mRNA Vaccines and the Potential for Mass Vaccination During Pandemics. Annu Rev Pharmacol Toxicol 2021; 61:517-540. [PMID: 32466690 PMCID: PMC8057107 DOI: 10.1146/annurev-pharmtox-030320-092348] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oral vaccination enables pain-free and self-administrable vaccine delivery for rapid mass vaccination during pandemic outbreaks. Furthermore, it elicits systemic and mucosal immune responses. This protects against infection at mucosal surfaces, which may further enhance protection and minimize the spread of disease. The gastrointestinal (GI) tract presents a number of prospective mucosal inductive sites for vaccine targeting, including the oral cavity, stomach, and small intestine. However, currently available oral vaccines are effectively limited to live-attenuated and inactivated vaccines against enteric diseases. The GI tract poses a number of challenges,including degradative processes that digest biologics and mucosal barriers that limit their absorption. This review summarizes the approaches currently under development and future opportunities for oral vaccine delivery to established (intestinal) and relatively new (oral cavity, stomach) mucosal targets. Special consideration is given to recent advances in oral biologic delivery that offer promise as future platforms for the administration of oral vaccines.
Collapse
Affiliation(s)
- Jacob William Coffey
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunology, University of Melbourne, Victoria, 3000, Australia
| | - Gaurav Das Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
90
|
Muehler A, Slizgi JR, Kohlhof H, Groeppel M, Peelen E, Vitt D. Clinical relevance of intestinal barrier dysfunction in common gastrointestinal diseases. World J Gastrointest Pathophysiol 2020; 11:114-130. [PMID: 33362939 PMCID: PMC7739114 DOI: 10.4291/wjgp.v11.i6.114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/07/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
The intestinal barrier is a complex and well-controlled physiological construct designed to separate luminal contents from the bowel wall. In this review, we focus on the intestinal barrier’s relationship with the host’s immune system interaction and the external environment, specifically the microbiome. The bowel allows the host to obtain nutrients vital to survival while protecting itself from harmful pathogens, luminal antigens, or other pro-inflammatory factors. Control over barrier function and the luminal milieu is maintained at the biochemical, cellular, and immunological level. However, disruption to this highly regulated environment can cause disease. Recent advances to the field have progressed the mechanistic understanding of compromised intestinal barrier function in the context of gastrointestinal pathology. There are numerous examples where bowel barrier dysfunction and the resulting interaction between the microbiome and the immune system has disease-triggering consequences. The purpose of this review is to summarize the clinical relevance of intestinal barrier dysfunction in common gastrointestinal and related diseases. This may help highlight the importance of restoring barrier function as a therapeutic mechanism of action in gastrointestinal pathology.
Collapse
|
91
|
Lewis CV, Taylor WR. Intestinal barrier dysfunction as a therapeutic target for cardiovascular disease. Am J Physiol Heart Circ Physiol 2020; 319:H1227-H1233. [PMID: 32986965 PMCID: PMC7792706 DOI: 10.1152/ajpheart.00612.2020] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023]
Abstract
The gut microbiome and intestinal dysfunction have emerged as potential contributors to the development of cardiovascular disease (CVD). Alterations in gut microbiome are well documented in hypertension, atherosclerosis, and heart failure and have been investigated as a therapeutic target. However, a perhaps underappreciated but related role for intestinal barrier function has become evident. Increased intestinal permeability is observed in patients and mouse models of CVD. This increased intestinal permeability can enhance systemic inflammation, alter gut immune function, and has been demonstrated as predictive of adverse cardiovascular outcomes. The goal of this review is to examine the evidence supporting a role for intestinal barrier function in cardiovascular disease and its prospect as a novel therapeutic target. We outline key studies that have investigated intestinal permeability in hypertension, coronary artery disease, atherosclerosis, heart failure, and myocardial infarction. We highlight the central mechanisms involved in the breakdown of barrier function and look at emerging evidence for restored barrier function as a contributor to promising treatment strategies such as short chain fatty acid, probiotic, and renin angiotensin system-targeted therapeutics. Recent studies of more selective targeting of the intestinal barrier to improve disease outcomes are also examined. We suggest that although current data supporting a contribution of intestinal permeability to CVD pathogenesis are largely associative, it appears to be a promising avenue for further investigation. Additional studies of the mechanisms of barrier restoration in CVD and testing of intestinal barrier-targeted compounds will be required to confirm their potential as a new class of CVD therapeutic.
Collapse
Affiliation(s)
- Caitlin V Lewis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Cardiology Division, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
92
|
Schoos AMM, Bullens D, Chawes BL, Costa J, De Vlieger L, DunnGalvin A, Epstein MM, Garssen J, Hilger C, Knipping K, Kuehn A, Mijakoski D, Munblit D, Nekliudov NA, Ozdemir C, Patient K, Peroni D, Stoleski S, Stylianou E, Tukalj M, Verhoeckx K, Zidarn M, van de Veen W. Immunological Outcomes of Allergen-Specific Immunotherapy in Food Allergy. Front Immunol 2020; 11:568598. [PMID: 33224138 PMCID: PMC7670865 DOI: 10.3389/fimmu.2020.568598] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022] Open
Abstract
IgE-mediated food allergies are caused by adverse immunologic responses to food proteins. Allergic reactions may present locally in different tissues such as skin, gastrointestinal and respiratory tract and may result is systemic life-threatening reactions. During the last decades, the prevalence of food allergies has significantly increased throughout the world, and considerable efforts have been made to develop curative therapies. Food allergen immunotherapy is a promising therapeutic approach for food allergies that is based on the administration of increasing doses of culprit food extracts, or purified, and sometime modified food allergens. Different routes of administration for food allergen immunotherapy including oral, sublingual, epicutaneous and subcutaneous regimens are being evaluated. Although a wealth of data from clinical food allergen immunotherapy trials has been obtained, a lack of consistency in assessed clinical and immunological outcome measures presents a major hurdle for evaluating these new treatments. Coordinated efforts are needed to establish standardized outcome measures to be applied in food allergy immunotherapy studies, allowing for better harmonization of data and setting the standards for the future research. Several immunological parameters have been measured in food allergen immunotherapy, including allergen-specific immunoglobulin levels, basophil activation, cytokines, and other soluble biomarkers, T cell and B cell responses and skin prick tests. In this review we discuss different immunological parameters and assess their applicability as potential outcome measures for food allergen immunotherapy that may be included in such a standardized set of outcome measures.
Collapse
Affiliation(s)
- Ann-Marie Malby Schoos
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Sygehus, Slagelse, Denmark
| | - Dominique Bullens
- Allergy and Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Clinical Division of Pediatrics, UZ Leuven, Leuven, Belgium
| | - Bo Lund Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Joana Costa
- REQUIMTE-LAQV, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Liselot De Vlieger
- Allergy and Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Audrey DunnGalvin
- School of Applied Psychology, University College Cork, Cork, Ireland
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child’s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Michelle M. Epstein
- Experimental Allergy Laboratory, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Centre of Excellence Immunology, Danone Nutricia research, Utrecht, Netherlands
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Karen Knipping
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Centre of Excellence Immunology, Danone Nutricia research, Utrecht, Netherlands
| | - Annette Kuehn
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Dragan Mijakoski
- Institute of Occupational Health of RNM, Skopje, North Macedonia
- Faculty of Medicine, Ss. Cyril and Methodius, University in Skopje, Skopje, North Macedonia
| | - Daniel Munblit
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child’s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Inflammation, Repair and Development Section, NHLI, Imperial College London, London, United Kingdom
| | - Nikita A. Nekliudov
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child’s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Cevdet Ozdemir
- Institute of Child Health, Department of Pediatric Basic Sciences, Istanbul University, Istanbul, Turkey
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Karine Patient
- SPI—Food Allergy Unit, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, CEA, INRAE, Gif-sur-Yvette, France
| | - Diego Peroni
- Section of Pediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sasho Stoleski
- Institute of Occupational Health of RNM, Skopje, North Macedonia
- Faculty of Medicine, Ss. Cyril and Methodius, University in Skopje, Skopje, North Macedonia
| | - Eva Stylianou
- Regional Unit for Asthma, Allergy and Hypersensitivity, Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Mirjana Tukalj
- Children’s Hospital, Department of Allergology and Pulmonology, Zagreb, Croatia
- Faculty of Medicine, University of Osijek, Osijek, Croatia
- Catholic University of Croatia, Zagreb, Croatia
| | - Kitty Verhoeckx
- Division of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mihaela Zidarn
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| |
Collapse
|
93
|
Soy bioactive peptides and the gut microbiota modulation. Appl Microbiol Biotechnol 2020; 104:9009-9017. [PMID: 32945899 DOI: 10.1007/s00253-020-10799-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
The balance of protein, carbohydrate, and fat affect the composition and functions of the gut microbiota. The complexities involved thereof require insights into the roles and impacts of individual dietary components due to the difficulty of defining such in a group of others. Peptides and proteins from several animal and plant sources have been widely explored in relation to the gut microbiome modulation, but the effects of soy peptides and other soy derivatives on the gut microbiota are largely unexplored. This piece considered an overview of the production and interventions of soy bioactive peptides on gut, as they affect the composition and functions of the gut microorganisms. A mini review on the production of soy protein hydrolysates/peptides and highlights of the most recent knowledge regarding their physiological effects on host's gut microbiota cum health were investigated. Overall deductions and research gaps were critically evaluated for futuristic interventions and relevance. Key points • Diet affects the composition of gut microorganisms. • Modulation of the gut microbiota by soy biopeptides is described. • Critical deductions on personal and commercial use are provided.
Collapse
|
94
|
Rajendran A, Poncet N, Oparija-Rogenmozere L, Herzog B, Verrey F. Tissue-specific deletion of mouse basolateral uniporter LAT4 (Slc43a2) reveals its crucial role in small intestine and kidney amino acid transport. J Physiol 2020; 598:5109-5132. [PMID: 32841365 PMCID: PMC7693055 DOI: 10.1113/jp280234] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/19/2020] [Indexed: 01/16/2023] Open
Abstract
Key points LAT4 is a broadly expressed uniporter selective for essential branched chain amino acids, methionine and phenylalanine, which are involved in epithelial transport. Its global deletion leads to an early malnutrition‐like phenotype and death within 10 days after birth. Here, we tested the impact of deleting LAT4 selectively in the mouse intestine. This affected slightly the absorption of amino acids (AAs) and delayed gastrointestinal motility; however, it had no major phenotypic effect, even when combined with aromatic AA uniporter TAT1 knockout (KO). Conversely, kidney tubule‐selective deletion of LAT4 led to a substantial aminoaciduria that strongly increased under a high protein diet. Combining a partial tubular LAT4 deletion with TAT1 KO implicated their synergistic action on AA reabsorption. These results show that LAT4 plays an important role for kidney AA reabsorption, but that its functional role in intestinal AA absorption is largely dispensable.
Abstract Amino acid (AA) transporter LAT4 (Slc43a2) functions as facilitated diffusion uniporter for essential neutral AAs and is highly expressed at the basolateral membrane of small intestine (SI) and kidney tubule epithelia. Previously, we showed that LAT4 global knockout (KO) mice were born at the expected Mendelian ratio but died within 10 days. Their failure to gain weight and a severe malnutrition‐like phenotype contrasted with apparently normal feeding, suggesting a severe intestinal AA absorption defect. In the present study, using conditional global and tissue‐specific LAT4 KO mouse models, we nullified this hypothesis, demonstrating that the selective lack of intestinal LAT4 does not impair postnatal development, although it leads to an absorption defect accompanied by delayed gastrointestinal motility. Kidney tubule‐specific LAT4 KO led to a substantial aminoaciduria as a result of a reabsorption defect of AAs transported by LAT4 and of other AAs that are substrates of the antiporter LAT2, demonstrating, in vivo, the functional co‐operation of these two transporters. The major role played by basolateral uniporters in the kidney was further supported by the observation that, in mice lacking TAT1, another neutral AA uniporter, a partial LAT4 KO led to a synergistic increase of urinary AA loss. Surprisingly in the SI, the same combined KO induced no major effect, suggesting yet unknown compensatory mechanisms. Taken together, the lethal malnutrition‐like phenotype observed previously in LAT4 global KO pups is suggested to be the consequence of a combinatorial effect of LAT4 deletion in the SI, kidney and presumably other tissues. LAT4 is a broadly expressed uniporter selective for essential branched chain amino acids, methionine and phenylalanine, which are involved in epithelial transport. Its global deletion leads to an early malnutrition‐like phenotype and death within 10 days after birth. Here, we tested the impact of deleting LAT4 selectively in the mouse intestine. This affected slightly the absorption of amino acids (AAs) and delayed gastrointestinal motility; however, it had no major phenotypic effect, even when combined with aromatic AA uniporter TAT1 knockout (KO). Conversely, kidney tubule‐selective deletion of LAT4 led to a substantial aminoaciduria that strongly increased under a high protein diet. Combining a partial tubular LAT4 deletion with TAT1 KO implicated their synergistic action on AA reabsorption. These results show that LAT4 plays an important role for kidney AA reabsorption, but that its functional role in intestinal AA absorption is largely dispensable.
Collapse
Affiliation(s)
| | - Nadège Poncet
- Institute of Physiology University of Zurich, Zurich, Switzerland
| | | | - Brigitte Herzog
- Institute of Physiology University of Zurich, Zurich, Switzerland
| | - François Verrey
- Institute of Physiology University of Zurich, Zurich, Switzerland.,NCCR Kidney. CH, University of Zurich, Zurich, Switzerland
| |
Collapse
|
95
|
Choi JH, Park SE, Yeo SH, Kim S. Anti-Inflammatory and Cytotoxicity Effects of Cudrania tricuspidata Fruits Vinegar in a Co-Culture System with RAW264.7 Macrophages and 3T3-L1 Adipocytes. Foods 2020; 9:foods9091232. [PMID: 32899648 PMCID: PMC7554877 DOI: 10.3390/foods9091232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/26/2022] Open
Abstract
Vinegar has been found to have in vitro improvement effect on inflammatory biomarkers, and clinically used to improve inflammation and obesity-related diseases. This study was designed to analyze in vitro anti-inflammatory effects of Cudrania tricuspidata fruits vinegar (CTFV) in a co-culture system with macrophages and adipocytes. We analyzed the physicochemical properties and polyphenolic ingredients of CTFV, and investigated in vitro anti-inflammatory effects of CTFV in a co-culture system with macrophages and adipocytes. The cells were cultured in the presence of CTFV for 24 h in contact with each other, then, harvested. The levels of monocyte chemoattractant protein (MCP)-1, tumour necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS), nitric oxide (NO), and interleukin (IL)-6 were evaluated by using the Griess reagent, western blot, or enzyme-linked immunosorbent assay assays. We found that increasing levels for NO, iNOS, TNF-α, IL-6 and MCP-1 were caused by LPS treatment and co-culture using the contact method, whereas CTFV efficaciously attenuated inflammatory response by improving inflammatory parameters including NO, iNOS, TNF-α, IL-6 and MCP-1. The present study indicates that CTFV might provide a nutraceutical product or functional food resource for improving inflammation processed via the interaction of adipocytes and macrophages.
Collapse
Affiliation(s)
- Jun-Hui Choi
- Department of Food Science and Biotechnology, Gwangju University, Gwangju 503-703, Korea; (J.-H.C.); (S.-E.P.)
| | - Se-Eun Park
- Department of Food Science and Biotechnology, Gwangju University, Gwangju 503-703, Korea; (J.-H.C.); (S.-E.P.)
| | - Soo-Hwan Yeo
- Department of Agro-food Resource, National Academy of Agricultural Science, RDA, Suwon 441-853, Korea;
| | - Seung Kim
- Department of Food Science and Biotechnology, Gwangju University, Gwangju 503-703, Korea; (J.-H.C.); (S.-E.P.)
- Correspondence: ; Tel.: +82-62-670-2718; Fax: +82-62-670-2761
| |
Collapse
|
96
|
Ilchmann-Diounou H, Menard S. Psychological Stress, Intestinal Barrier Dysfunctions, and Autoimmune Disorders: An Overview. Front Immunol 2020; 11:1823. [PMID: 32983091 PMCID: PMC7477358 DOI: 10.3389/fimmu.2020.01823] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Autoimmune disorders (ADs) are multifactorial diseases involving, genetic, epigenetic, and environmental factors characterized by an inappropriate immune response toward self-antigens. In the past decades, there has been a continuous rise in the incidence of ADs, which cannot be explained by genetic factors alone. Influence of psychological stress on the development or the course of autoimmune disorders has been discussed for a long time. Indeed, based on epidemiological studies, stress has been suggested to precede AD occurrence and to exacerbate symptoms. Furthermore, compiling data showed that most of ADs are associated with gastrointestinal symptoms, that is, microbiota dysbiosis, intestinal hyperpermeability, and intestinal inflammation. Interestingly, social stress (acute or chronic, in adult or in neonate) is a well-described intestinal disrupting factor. Taken together, those observations question a potential role of stress-induced defect of the intestinal barrier in the onset and/or the course of ADs. In this review, we aim to present evidences supporting the hypothesis for a role of stress-induced intestinal barrier disruption in the onset and/or the course of ADs. We will mainly focus on autoimmune type 1 diabetes, multiple sclerosis and systemic lupus erythematosus, ADs for which we could find sufficient circumstantial data to support this hypothesis. We excluded gastrointestinal (GI) ADs like coeliac disease to privilege ADs not focused on intestinal disorders to avoid confounding factors. Indeed, GIADs are characterized by antibodies directed against intestinal barrier actors.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/epidemiology
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/microbiology
- Autoimmunity
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/microbiology
- Dysbiosis
- Gastrointestinal Microbiome
- Host-Pathogen Interactions
- Humans
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Lupus Erythematosus, Systemic/epidemiology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/microbiology
- Multiple Sclerosis/epidemiology
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/microbiology
- Permeability
- Risk Factors
- Stress, Psychological/epidemiology
- Stress, Psychological/immunology
- Stress, Psychological/metabolism
- Stress, Psychological/microbiology
Collapse
Affiliation(s)
| | - Sandrine Menard
- Neuro-Gastroenterology and Nutrition Team, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
97
|
Di Ciaula A, Baj J, Garruti G, Celano G, De Angelis M, Wang HH, Di Palo DM, Bonfrate L, Wang DQH, Portincasa P. Liver Steatosis, Gut-Liver Axis, Microbiome and Environmental Factors. A Never-Ending Bidirectional Cross-Talk. J Clin Med 2020; 9:E2648. [PMID: 32823983 PMCID: PMC7465294 DOI: 10.3390/jcm9082648] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide and parallels comorbidities such as obesity, metabolic syndrome, dyslipidemia, and diabetes. Recent studies describe the presence of NAFLD in non-obese individuals, with mechanisms partially independent from excessive caloric intake. Increasing evidences, in particular, point towards a close interaction between dietary and environmental factors (including food contaminants), gut, blood flow, and liver metabolism, with pathways involving intestinal permeability, the composition of gut microbiota, bacterial products, immunity, local, and systemic inflammation. These factors play a critical role in the maintenance of intestinal, liver, and metabolic homeostasis. An anomalous or imbalanced gut microbial composition may favor an increased intestinal permeability, predisposing to portal translocation of microorganisms, microbial products, and cell wall components. These components form microbial-associated molecular patterns (MAMPs) or pathogen-associated molecular patterns (PAMPs), with potentials to interact in the intestine lamina propria enriched in immune cells, and in the liver at the level of the immune cells, i.e., Kupffer cells and stellate cells. The resulting inflammatory environment ultimately leads to liver fibrosis with potentials to progression towards necrotic and fibrotic changes, cirrhosis. and hepatocellular carcinoma. By contrast, measures able to modulate the composition of gut microbiota and to preserve gut vascular barrier might prevent or reverse NAFLD.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Gabriella Garruti
- Section of Endocrinology, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Giuseppe Celano
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Maria De Angelis
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Helen H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Domenica Maria Di Palo
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| |
Collapse
|
98
|
Dotsenko V, Oittinen M, Taavela J, Popp A, Peräaho M, Staff S, Sarin J, Leon F, Isola J, Mäki M, Viiri K. Genome-Wide Transcriptomic Analysis of Intestinal Mucosa in Celiac Disease Patients on a Gluten-Free Diet and Postgluten Challenge. Cell Mol Gastroenterol Hepatol 2020; 11:13-32. [PMID: 32745639 PMCID: PMC7593586 DOI: 10.1016/j.jcmgh.2020.07.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Gluten challenge studies are instrumental in understanding the pathophysiology of celiac disease. Our aims in this study were to reveal early gluten-induced transcriptomic changes in duodenal biopsies and to find tools for clinics. METHODS Duodenal biopsies were collected from 15 celiac disease patients on a strict long-term gluten-free diet (GFD) prior to and post a gluten challenge (PGC) and from 6 healthy control individuals (DC). Biopsy RNA was subjected to genome-wide 3' RNA-Seq. Sequencing data was used to determine the differences between the three groups and was compared to sequencing data from the public repositories. The biopsies underwent morphometric analyses. RESULTS In DC vs. GFD group comparisons, 167 differentially expressed genes were identified with 117 genes downregulated and 50 genes upregulated. In PGC vs. GFD group comparisons, 417 differentially expressed genes were identified with 195 genes downregulated and 222 genes upregulated. Celiac disease patients on a GFD were not "healthy". In particular, genes encoding proteins for transporting small molecules were expressed less. In addition to the activation of immune response genes, a gluten challenge induced hyperactive intestinal wnt-signaling and consequent immature crypt gene expression resulting in less differentiated epithelium. Biopsy gene expression in response to a gluten challenge correlated with the extent of the histological damage. Regression models using only four gene transcripts described 97.2% of the mucosal morphology and 98.0% of the inflammatory changes observed. CONCLUSIONS Our gluten challenge trial design provided an opportunity to study the transition from health to disease. The results show that even on a strict GFD, despite being deemed healthy, patients reveal patterns of ongoing disease. Here, a transcriptomic regression model estimating the extent of gluten-induced duodenal mucosal injury is presented.
Collapse
Affiliation(s)
- Valeriia Dotsenko
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland
| | - Mikko Oittinen
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland
| | - Juha Taavela
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland,Department of Internal Medicine, Central Finland Central Hospital, Jyväskylä, Finland
| | - Alina Popp
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland,Pediatric Department, Carol Davila University of Medicine and Pharmacy, Alessandrescu-Rusescu National Institute for Mother and Child Health, Bucharest, Romania
| | - Markku Peräaho
- Department of Internal Medicine, Central Finland Central Hospital, Jyväskylä, Finland
| | - Synnöve Staff
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University, Tampere, Finland
| | - Jani Sarin
- Laboratory of Cancer Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland,Jilab Inc, Tampere, Finland
| | | | - Jorma Isola
- Laboratory of Cancer Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland,Jilab Inc, Tampere, Finland
| | - Markku Mäki
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland
| | - Keijo Viiri
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University Hospital, Tampere University Tampere, Finland,Correspondence Address correspondence to: Keijo Viiri, PhD, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Arvo Ylpön katu 34, Tampere, FIN-33520, Finland; fax: +35833641369.
| |
Collapse
|
99
|
Bile acid transporter-mediated oral drug delivery. J Control Release 2020; 327:100-116. [PMID: 32711025 DOI: 10.1016/j.jconrel.2020.07.034] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/12/2022]
Abstract
Bile acids are synthesized in the liver, stored in the gallbladder, and secreted into the duodenum at meals. Apical sodium-dependent bile acid transporter (ASBT), an ileal Na+-dependent transporter, plays the leading role of bile acid absorption into enterocytes, where bile acids are delivered to basolateral side by ileal bile acid binding protein (IBABP) and then released by organic solute transporter OSTα/β. The absorbed bile acids are delivered to the liver via portal vein. In this process called "enterohepatic recycling", only 5% of the bile acid pool (~3 g in human) is excreted in feces, indicating the large recycling capacity and high transport efficacy of ASBT-mediated absorption. Therefore, bile acid transporter-mediated oral drug delivery has been regarded as a feasible and potential strategy to improve the oral bioavailability. This review introduces the key factors in enterohepatic recycling, especially the mechanism of bile acid uptake by ASBT, and the development of bile acid-based oral drug delivery for ASBT-targeting, including bile acid-based prodrugs, bile acid/drug electrostatic complexation and bile acid-containing nanocarriers. Furthermore, the specific transport pathways of bile acid in enterocytes are described and the recent finding of lymphatic delivery of bile acid-containing nanocarriers is discussed.
Collapse
|
100
|
Coméra C, Cartier C, Gaultier E, Catrice O, Panouille Q, El Hamdi S, Tirez K, Nelissen I, Théodorou V, Houdeau E. Jejunal villus absorption and paracellular tight junction permeability are major routes for early intestinal uptake of food-grade TiO 2 particles: an in vivo and ex vivo study in mice. Part Fibre Toxicol 2020; 17:26. [PMID: 32527323 PMCID: PMC7345522 DOI: 10.1186/s12989-020-00357-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/29/2020] [Indexed: 01/13/2023] Open
Abstract
Background Food-grade TiO2 (E171 in the EU) is widely used as a coloring agent in foodstuffs, including sweets. Chronic dietary exposure raises concerns for human health due to proinflammatory properties and the ability to induce and promote preneoplastic lesions in the rodent gut. Characterization of intestinal TiO2 uptake is essential for assessing the health risk in humans. We studied in vivo the gut absorption kinetics of TiO2 in fasted mice orally given a single dose (40 mg/kg) to assess the ability of intestinal apical surfaces to absorb particles when available without entrapment in the bolus. The epithelial translocation pathways were also identified ex vivo using intestinal loops in anesthetized mice. Results The absorption of TiO2 particles was analyzed in gut tissues by laser-reflective confocal microscopy and ICP-MS at 4 and 8 h following oral administration. A bimodal pattern was detected in the small intestine: TiO2 absorption peaked at 4 h in jejunal and ileal villi before returning to basal levels at 8 h, while being undetectable at 4 h but significantly present at 8 h in the jejunal Peyer’s patches (PP). Lower absorption occurred in the colon, while TiO2 particles were clearly detectable by confocal microscopy in the blood at 4 and 8 h after treatment. Ex vivo, jejunal loops were exposed to the food additive in the presence and absence of pharmacological inhibitors of paracellular tight junction (TJ) permeability or of transcellular (endocytic) passage. Thirty minutes after E171 addition, TiO2 absorption by the jejunal villi was decreased by 66% (p < 0.001 vs. control) in the presence of the paracellular permeability blocker triaminopyrimidine; the other inhibitors had no significant effect. Substantial absorption through a goblet cell (GC)-associated pathway, insensitive to TJ blockade, was also detected. Conclusions After a single E171 dose in mice, early intestinal uptake of TiO2 particles mainly occurred through the villi of the small intestine, which, in contrast to the PP, represent the main absorption surface in the small intestine. A GC-associated passage and passive diffusion through paracellular TJ spaces between enterocytes appeared to be major absorption routes for transepithelial uptake of dietary TiO2.
Collapse
Affiliation(s)
- Christine Coméra
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France.
| | - Christel Cartier
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France
| | - Eric Gaultier
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France
| | - Olivier Catrice
- LIPM, Université de Toulouse, INRAE, CNRS, Castanet-Tolosan, France
| | - Quentin Panouille
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France
| | - Sarah El Hamdi
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France
| | - Kristof Tirez
- VITO (Flemish Institute for Technological Research), Mol, Belgium
| | - Inge Nelissen
- VITO (Flemish Institute for Technological Research), Mol, Belgium
| | - Vassilia Théodorou
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France
| | - Eric Houdeau
- Toxalim (Research Centre in Food Toxicology), Toulouse University, INRAE, ENVT, INP, Toulouse, France
| |
Collapse
|