51
|
Lindner M, Laporte A, Block S, Elomaa L, Weinhart M. Physiological Shear Stress Enhances Differentiation, Mucus-Formation and Structural 3D Organization of Intestinal Epithelial Cells In Vitro. Cells 2021; 10:2062. [PMID: 34440830 PMCID: PMC8391940 DOI: 10.3390/cells10082062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) mucus plays a pivotal role in the tissue homoeostasis and functionality of the gut. However, due to the shortage of affordable, realistic in vitro GI models with a physiologically relevant mucus layer, studies with deeper insights into structural and compositional changes upon chemical or physical manipulation of the system are rare. To obtain an improved mucus-containing cell model, we developed easy-to-use, reusable culture chambers that facilitated the application of GI shear stresses (0.002-0.08 dyn∙cm-2) to cells on solid surfaces or membranes of cell culture inserts in bioreactor systems, thus making them readily accessible for subsequent analyses, e.g., by confocal microscopy or transepithelial electrical resistance (TEER) measurement. The human mucus-producing epithelial HT29-MTX cell-line exhibited superior reorganization into 3-dimensional villi-like structures with highly proliferative tips under dynamic culture conditions when compared to static culture (up to 180 vs. 80 µm in height). Additionally, the median mucus layer thickness was significantly increased under flow (50 ± 24 vs. 29 ± 14 µm (static)), with a simultaneous accelerated maturation of the cells into a goblet-like phenotype. We demonstrated the strong impact of culture conditions on the differentiation and reorganization of HT29-MTX cells. The results comprise valuable advances towards the improvement of existing GI and mucus models or the development of novel systems using our newly designed culture chambers.
Collapse
Affiliation(s)
- Marcus Lindner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany; (M.L.); (S.B.); (L.E.)
| | - Anna Laporte
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, 30167 Hannover, Germany;
| | - Stephan Block
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany; (M.L.); (S.B.); (L.E.)
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany; (M.L.); (S.B.); (L.E.)
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany; (M.L.); (S.B.); (L.E.)
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, 30167 Hannover, Germany;
| |
Collapse
|
52
|
Neu3 neuraminidase induction triggers intestinal inflammation and colitis in a model of recurrent human food-poisoning. Proc Natl Acad Sci U S A 2021; 118:2100937118. [PMID: 34266954 DOI: 10.1073/pnas.2100937118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Intestinal inflammation is the underlying basis of colitis and the inflammatory bowel diseases. These syndromes originate from genetic and environmental factors that remain to be fully identified. Infections are possible disease triggers, including recurrent human food-poisoning by the common foodborne pathogen Salmonella enterica Typhimurium (ST), which in laboratory mice causes progressive intestinal inflammation leading to an enduring colitis. In this colitis model, disease onset has been linked to Toll-like receptor-4-dependent induction of intestinal neuraminidase activity, leading to the desialylation, reduced half-life, and acquired deficiency of anti-inflammatory intestinal alkaline phosphatase (IAP). Neuraminidase (Neu) inhibition protected against disease onset; however, the source and identity of the Neu enzyme(s) responsible remained unknown. Herein, we report that the mammalian Neu3 neuraminidase is responsible for intestinal IAP desialylation and deficiency. Absence of Neu3 thereby prevented the accumulation of lipopolysaccharide-phosphate and inflammatory cytokine expression in providing protection against the development of severe colitis.
Collapse
|
53
|
New Stable Cell Lines Derived from the Proximal and Distal Intestine of Rainbow Trout ( Oncorhynchus mykiss) Retain Several Properties Observed In Vivo. Cells 2021; 10:cells10061555. [PMID: 34205481 PMCID: PMC8235179 DOI: 10.3390/cells10061555] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/22/2022] Open
Abstract
We derived two novel cell lines from rainbow trout (RT) proximal (RTpi-MI) and distal intestine (RTdi-MI) and compared them with the previously established continuous cell line RTgutGC. Intestinal stem cells, differentiating and differentiated epithelial cells, and connective cells were found in all cell lines. The cell lines formed a polarized barrier, which was not permeable to large molecules and absorbed proline and glucose. High seeding density induced their differentiation into more mature phenotypes, as indicated by the downregulation of intestinal stem cell-related genes (i.e., sox9, hopx and lgr5), whereas alkaline phosphatase activity was upregulated. Other enterocyte markers (i.e., sglt1 and pept1), however, were not regulated as expected. In all cell lines, the presence of a mixed population of epithelial and stromal cells was characterized for the first time. The expression by the stromal component of lgr5, a stem cell niche regulatory molecule, may explain why these lines proliferate stably in vitro. Although most parameters were conserved among the three cell lines, some significant differences were observed, suggesting that characteristics typical of each tract are partly conserved in vitro as well.
Collapse
|
54
|
Tan R, Dong H, Chen Z, Jin M, Yin J, Li H, Shi D, Shao Y, Wang H, Chen T, Yang D, Li J. Intestinal Microbiota Mediates High-Fructose and High-Fat Diets to Induce Chronic Intestinal Inflammation. Front Cell Infect Microbiol 2021; 11:654074. [PMID: 34222037 PMCID: PMC8242949 DOI: 10.3389/fcimb.2021.654074] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/31/2021] [Indexed: 01/14/2023] Open
Abstract
An unhealthy diet has been linked to increased incidence of chronic diseases. To investigate the relationship between diet and intestinal inflammation, mice in two experimental groups were fed on a high-fat diet or high-fructose diet, respectively. The result showed that the defecation volume of the experimental groups was significantly reduced compared with that of the control group, and the levels of pro-inflammatory cytokines (interleukin (IL)-1β and IL-6) and IgG in serum were increased significantly. In addition, inflammatory cell infiltration was observed in intestinal tissue, indicating that a high-fructose or high-fat diet can lead to constipation and inflammation. Further analysis showed that the microbial composition of the experimental groups changed significantly, including a decrease of the Bacteroidetes/Firmicutes ratio and increased levels of Bacteroides, Akkermansia, Lactobacillus, and Ruminococcus, which might be associated with inflammation. The results of pro-inflammatory metabolites analysis showed that the levels of arachidonic acid, stearic acid, and indoxylsulfuric acid were significantly increased in the experimental groups, which were related significantly to Bacteroides, Enterococcus, and Akkermansia. Meanwhile, the content of 5-hydroxytryptamine (5-HT) was significantly decreased, which might cause constipation by reducing intestinal peristalsis. Moreover, transplantation of fecal bacteria from inflammatory mice caused constipation and inflammation in normal mice, which could be relieved by feeding a normal diet. The results of the present study indicated that changes in intestinal microbiota and microbial metabolites may underlie chronic intestinal inflammation and constipation caused by high-fructose and high-fat diets.
Collapse
Affiliation(s)
- Rong Tan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Huiwei Dong
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Zhengshan Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Min Jin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jing Yin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Haibei Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Danyang Shi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Yifan Shao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Huaran Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Tianjiao Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Dong Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Junwen Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
55
|
Nagasaki A, Nagasaki K, Kear BD, Tadesse WD, Thumbigere-Math V, Millán JL, Foster BL, Somerman MJ. Delivery of Alkaline Phosphatase Promotes Periodontal Regeneration in Mice. J Dent Res 2021; 100:993-1001. [PMID: 33840251 DOI: 10.1177/00220345211005677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Factors regulating the ratio of pyrophosphate (PPi) to phosphate (Pi) modulate biomineralization. Tissue-nonspecific alkaline phosphatase (TNAP) is a key promineralization enzyme that hydrolyzes the potent mineralization inhibitor PPi. The goal of this study was to determine whether TNAP could promote periodontal regeneration in bone sialoprotein knockout mice (Ibsp-/- mice), which are known to have a periodontal disease phenotype. Delivery of TNAP was accomplished either systemically (through a lentiviral construct expressing a mineral-targeted TNAP-D10 protein) or locally (through addition of recombinant human TNAP to a fenestration defect model). Systemic TNAP-D10 delivered by intramuscular injection at 5 d postnatal (dpn) increased circulating alkaline phosphatase (ALP) levels in Ibsp-/- mice by 5-fold at 30 dpn, with levels returning to normal by 60 dpn when tissues were evaluated by micro-computed tomography and histology. Local delivery of recombinant human TNAP to fenestration defects in 5-wk-old wild type (WT) and Ibsp-/- mice did not alter long-term circulating ALP levels, and tissues were evaluated by micro-computed tomography and histology at postoperative day 45. Systemic and local delivery of TNAP significantly increased alveolar bone volume (20% and 37%, respectively) and cementum thickness (3- and 42-fold) in Ibsp-/- mice, with evidence for periodontal ligament attachment and bone/cementum marker localization. Local delivery significantly increased regenerated cementum and bone in WT mice. Addition of 100-μg/mL bovine intestinal ALP to culture media to increase ALP in vitro increased media Pi concentration, mineralization, and Spp1 and Dmp1 marker gene expression in WT and Ibsp-/- OCCM.30 cementoblasts. Use of phosphonoformic acid, a nonspecific inhibitor of sodium Pi cotransport, indicated that effects of bovine intestinal ALP on mineralization and marker gene expression were in part through Pi transport. These findings show for the first time through multiple in vivo and in vitro approaches that pharmacologic modulation of Pi/PPi metabolism can overcome periodontal breakdown and accomplish regeneration.
Collapse
Affiliation(s)
- A Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - K Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - B D Kear
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - W D Tadesse
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - V Thumbigere-Math
- Division of Periodontology, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - J L Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - B L Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M J Somerman
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
56
|
Chen VL, Du X, Chen Y, Kuppa A, Handelman SK, Vohnoutka RB, Peyser PA, Palmer ND, Bielak LF, Halligan B, Speliotes EK. Genome-wide association study of serum liver enzymes implicates diverse metabolic and liver pathology. Nat Commun 2021; 12:816. [PMID: 33547301 PMCID: PMC7865025 DOI: 10.1038/s41467-020-20870-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Serum liver enzyme concentrations are the most frequently-used laboratory markers of liver disease, a major cause of mortality. We conduct a meta-analysis of genome-wide association studies of liver enzymes from UK BioBank and BioBank Japan. We identified 160 previously-unreported independent alanine aminotransferase, 190 aspartate aminotransferase, and 199 alkaline phosphatase genome-wide significant associations, with some affecting multiple different enzymes. Associated variants implicate genes that demonstrate diverse liver cell type expression and promote a range of metabolic and liver diseases. These findings provide insight into the pathophysiology of liver and other metabolic diseases that are associated with serum liver enzyme concentrations. Serum liver enzymes are used as markers of liver disease, their concentration influenced in part by genetic factors. Here the authors meta-analyse genome-wide association studies on the UK Biobank and BioBank Japan to evaluate the association of three liver enzymes with liver and other metabolic diseases.
Collapse
Affiliation(s)
- Vincent L Chen
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Xiaomeng Du
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Yanhua Chen
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Annapurna Kuppa
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Samuel K Handelman
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rishel B Vohnoutka
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lawrence F Bielak
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Brian Halligan
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Elizabeth K Speliotes
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA. .,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
57
|
Lannoy V, Côté-Biron A, Asselin C, Rivard N. Phosphatases in toll-like receptors signaling: the unfairly-forgotten. Cell Commun Signal 2021; 19:10. [PMID: 33494775 PMCID: PMC7829650 DOI: 10.1186/s12964-020-00693-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past 2 decades, pattern recognition receptors (PRRs) have been shown to be on the front line of many illnesses such as autoimmune, inflammatory, and neurodegenerative diseases as well as allergies and cancer. Among PRRs, toll-like receptors (TLRs) are the most studied family. Dissecting TLRs signaling turned out to be advantageous to elaborate efficient treatments to cure autoimmune and chronic inflammatory disorders. However, a broad understanding of TLR effectors is required to propose a better range of cures. In addition to kinases and E3 ubiquitin ligases, phosphatases emerge as important regulators of TLRs signaling mediated by NF-κB, type I interferons (IFN I) and Mitogen-Activated Protein Kinases signaling pathways. Here, we review recent knowledge on TLRs signaling modulation by different classes and subclasses of phosphatases. Thus, it becomes more and more evident that phosphatases could represent novel therapeutic targets to control pathogenic TLRs signaling. Video Abstract.
Collapse
Affiliation(s)
- Valérie Lannoy
- Department of Immunology and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201, rue Jean Mignault, Sherbrooke, QC, J1E4K8, Canada
| | - Anthony Côté-Biron
- Department of Immunology and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201, rue Jean Mignault, Sherbrooke, QC, J1E4K8, Canada
| | - Claude Asselin
- Department of Immunology and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201, rue Jean Mignault, Sherbrooke, QC, J1E4K8, Canada
| | - Nathalie Rivard
- Department of Immunology and Cell Biology, Cancer Research Pavilion, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201, rue Jean Mignault, Sherbrooke, QC, J1E4K8, Canada.
| |
Collapse
|
58
|
Martínez-Llorens S, Peruzzi S, Falk-Petersen IB, Godoy-Olmos S, Ulleberg LO, Tomás-Vidal A, Puvanendran V, Odei DK, Hagen Ø, Fernandes JMO, Jobling M. Digestive tract morphology and enzyme activities of juvenile diploid and triploid Atlantic salmon (Salmo salar) fed fishmeal-based diets with or without fish protein hydrolysates. PLoS One 2021; 16:e0245216. [PMID: 33429419 PMCID: PMC7801030 DOI: 10.1371/journal.pone.0245216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/23/2020] [Indexed: 12/25/2022] Open
Abstract
Triploid, sterile Atlantic salmon (Salmo salar) could make a contribution to the development of the farming industry, but uncertainties about the performance and welfare of triploids have limited their adoption by farmers. In this study, we compared the ontogeny of digestive tract morphology and enzyme activities (pepsin, trypsin, chymotrypsin, alkaline phosphatase and aminopeptidase) of diploid and triploid Atlantic salmon. Fish were fed diets based on fishmeal (STD) or a mix of fishmeal and hydrolysed fish proteins (HFM) whilst being reared at low temperature from start-feeding to completion of the parr-smolt transformation. Fish weights for each ploidy and feed combination were used to calculate thermal growth coefficients (TGCs) that spanned this developmental period, and the data were used to examine possible relationships between enzyme activities and growth. At the end of the experiment, faeces were collected and analyzed to determine the apparent digestibility coefficients (ADCs) of the dietary amino acids (AAs). Digestive tract histo-morphology did not differ substantially between ploidies and generally reflected organ maturation and functionality. There were no consistent differences in proteolytic enzyme activities resulting from the inclusion of HFM in the diet, nor was there improved digestibility and AA bioavailability of the HFM feed in either diploid or triploid fish. The triploid salmon had lower ADCs than diploids for most essential and non-essential AAs in both diets (STD and HFM), but without there being any indication of lower intestinal protease activity in triploid fish. When trypsin-to-chymotrypsin activity and trypsin and alkaline phosphatase (ALP) ratios (T:C and T:ALP, respectively) were considered in combination with growth data (TGC) low T:C and T:ALP values coincided with times of reduced fish growth, and vice versa, suggesting that T:C and T:ALP may be used to predict recent growth history and possible growth potential.
Collapse
Affiliation(s)
- Silvia Martínez-Llorens
- Aquaculture and Biodiversity Research Group, Institute of Science and Animal Technology (ICTA), Universitat Politècnica de València, València, Spain
| | - Stefano Peruzzi
- Faculty of Biosciences, Fisheries and Economics, UiT - the Arctic University of Norway, Tromsø, Norway
| | - Inger-Britt Falk-Petersen
- Faculty of Biosciences, Fisheries and Economics, UiT - the Arctic University of Norway, Tromsø, Norway
| | - Sergio Godoy-Olmos
- Aquaculture and Biodiversity Research Group, Institute of Science and Animal Technology (ICTA), Universitat Politècnica de València, València, Spain
| | - Lars Olav Ulleberg
- Faculty of Biosciences, Fisheries and Economics, UiT - the Arctic University of Norway, Tromsø, Norway
| | - Ana Tomás-Vidal
- Aquaculture and Biodiversity Research Group, Institute of Science and Animal Technology (ICTA), Universitat Politècnica de València, València, Spain
| | | | - Derrick Kwame Odei
- Faculty of Biosciences, Fisheries and Economics, UiT - the Arctic University of Norway, Tromsø, Norway
| | - Ørjan Hagen
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Malcolm Jobling
- Faculty of Biosciences, Fisheries and Economics, UiT - the Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
59
|
Alvarenga L, Cardozo LFMF, Lindholm B, Stenvinkel P, Mafra D. Intestinal alkaline phosphatase modulation by food components: predictive, preventive, and personalized strategies for novel treatment options in chronic kidney disease. EPMA J 2020; 11:565-579. [PMID: 33240450 PMCID: PMC7680467 DOI: 10.1007/s13167-020-00228-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
Alkaline phosphatase (AP) is a ubiquitous membrane-bound glycoprotein that catalyzes phosphate monoesters' hydrolysis from organic compounds, an essential process in cell signaling. Four AP isozymes have been described in humans, placental AP, germ cell AP, tissue nonspecific AP, and intestinal AP (IAP). IAP plays a crucial role in gut microbial homeostasis, nutrient uptake, and local and systemic inflammation, and its dysfunction is associated with persistent inflammatory disorders. AP is a strong predictor of mortality in the general population and patients with cardiovascular and chronic kidney disease (CKD). However, little is known about IAP modulation and its possible consequences in CKD, a disease characterized by gut microbiota imbalance and persistent low-grade inflammation. Mitigating inflammation and dysbiosis can prevent cardiovascular complications in patients with CKD, and monitoring factors such as IAP can be useful for predicting those complications. Here, we review IAP's role and the results of nutritional interventions targeting IAP in experimental models to prevent alterations in the gut microbiota, which could be a possible target of predictive, preventive, personalized medicine (PPPM) to avoid CKD complications. Microbiota and some nutrients may activate IAP, which seems to have a beneficial impact on health; however, data on CKD remains scarce.
Collapse
Affiliation(s)
- L. Alvarenga
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
| | - L. F. M. F. Cardozo
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| | - B. Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - P. Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - D. Mafra
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| |
Collapse
|
60
|
Abstract
Nutrient content and nutrient timing are considered key regulators of human health and a variety of diseases and involve complex interactions with the mucosal immune system. In particular, the innate immune system is emerging as an important signaling hub that modulates the response to nutritional signals, in part via signaling through the gut microbiota. In this review we elucidate emerging evidence that interactions between innate immunity and diet affect human metabolic health and disease, including cardiometabolic disorders, allergic diseases, autoimmune disorders, infections, and cancers. Furthermore, we discuss the potential modulatory effects of the gut microbiota on interactions between the immune system and nutrition in health and disease, namely how it relays nutritional signals to the innate immune system under specific physiological contexts. Finally, we identify key open questions and challenges to comprehensively understanding the intersection between nutrition and innate immunity and how potential nutritional, immune, and microbial therapeutics may be developed into promising future avenues of precision treatment.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Niv Zmora
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel;
- Research Center for Digestive Tract and Liver Diseases and Internal Medicine Division, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel;
- Cancer-Microbiome Research Division, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany;
| |
Collapse
|
61
|
Ok M, Yildiz R, Hatipoglu F, Baspinar N, Ider M, Üney K, Ertürk A, Durgut MK, Terzi F. Use of intestine-related biomarkers for detecting intestinal epithelial damage in neonatal calves with diarrhea. Am J Vet Res 2020; 81:139-146. [PMID: 31985285 DOI: 10.2460/ajvr.81.2.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the usefulness of intestinal biomarkers in determining the presence of intestinal epithelial damage in neonatal calves with diarrhea caused by 4 etiologic agents. ANIMALS 40 neonatal calves that were healthy (n = 10) or had diarrhea (30). PROCEDURES The study was a cross-sectional study. Results of hematologic analyses and serum concentrations of intestinal fatty acid-binding protein (I-FABP), liver fatty acid-binding protein (L-FABP), trefoil factor 3 (TFF-3), Claudin-3 (CLDN-3), γ-enteric smooth muscle actin (ACTG2), intestinal alkaline phosphatase (IAP), interleukin-8 (IL-8), platelet-activating factor (PAF), and leptin (LP) were compared among calves grouped according to whether they were healthy (control group; G-1) or had diarrhea caused by K99 Escherichia coli (G-2; n = 10), bovine rota- or coronavirus (G-3; 5 each), or Cryptosporidium spp (G-4; 10). RESULTS Across the 3 time points at which blood samples were obtained and evaluated, the groups of calves with diarrhea generally had markedly higher mean serum concentrations of L-FABP, TFF-3, IAP, IL-8, and LP, compared with the control group. In addition, G-2 also consistently had markedly higher mean serum concentrations of I-FAB and ACTG2 and lower mean serum concentrations of CLDN-3, compared with the control group. CONCLUSIONS AND CLINICAL RELEVANCE Results indicated that degree of intestinal epithelial damage differed among calves grouped by the etiologic agent of diarrhea and that such damage might have been more severe in calves with diarrhea caused by K99 E coli. Additionally, our results indicated that serum concentrations of I-FABP, L-FABP, TFF-3, IAP, IL-8, ACTG2, LP, and CLDN-3 were useful biomarkers of intestinal epithelial damage in calves of the present study.
Collapse
|
62
|
Balasubramanyam M. LPS—Is It a Major Liability Factor for Cancer Risk and Severity? EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2020; 000:1-2. [DOI: 10.14218/erhm.2020.00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
63
|
Akiba Y, Maruta K, Takajo T, Narimatsu K, Said H, Kato I, Kuwahara A, Kaunitz JD. Lipopolysaccharides transport during fat absorption in rodent small intestine. Am J Physiol Gastrointest Liver Physiol 2020; 318:G1070-G1087. [PMID: 32390462 PMCID: PMC7311662 DOI: 10.1152/ajpgi.00079.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharides (LPS) are potent pro-inflammatory molecules that enter the systemic circulation from the intestinal lumen by uncertain mechanisms. We investigated these mechanisms and the effect of exogenous glucagon-like peptide-2 (GLP-2) on LPS transport in the rodent small intestine. Transmucosal LPS transport was measured in Ussing-chambered rat jejunal mucosa. In anesthetized rats, the appearance of fluorescein isothiocyanate (FITC)-LPS into the portal vein (PV) and the mesenteric lymph was simultaneously monitored after intraduodenal perfusion of FITC-LPS with oleic acid and taurocholate (OA/TCA). In vitro, luminally applied LPS rapidly appeared in the serosal solution only with luminal OA/TCA present, inhibited by the lipid raft inhibitor methyl-β-cyclodextrin (MβCD) and the CD36 inhibitor sulfosuccinimidyl oleate (SSO), or by serosal GLP-2. In vivo, perfusion of FITC-LPS with OA/TCA rapidly increased FITC-LPS appearance into the PV, followed by a gradual increase of FITC-LPS into the lymph. Rapid PV transport was inhibited by the addition of MβCD or by SSO, whereas transport into the lymph was inhibited by chylomicron synthesis inhibition. Intraveous injection of the stable GLP-2 analog teduglutide acutely inhibited FITC-LPS transport into the PV, yet accelerated FITC-LPS transport into the lymph via Nω-nitro-l-arginine methyl ester (l-NAME)- and PG97-269-sensitive mechanisms. In vivo confocal microscopy in mouse jejunum confirmed intracellular FITC-LPS uptake with no evidence of paracellular localization. This is the first direct demonstration in vivo that luminal LPS may cross the small intestinal barrier physiologically during fat absorption via lipid raft- and CD36-mediated mechanisms, followed by predominant transport into the PV, and that teduglutide inhibits LPS uptake into the PV in vivo.NEW & NOTEWORTHY We report direct in vivo confirmation of transcellular lipopolysaccharides (LPS) uptake from the intestine into the portal vein (PV) involving CD36 and lipid rafts, with minor uptake via the canonical chylomicron pathway. The gut hormone glucagon-like peptide-2 (GLP-2) inhibited uptake into the PV. These data suggest that the bulk of LPS absorption is via the PV to the liver, helping clarify the mechanism of LPS transport into the PV as part of the "gut-liver" axis. These data do not support the paracellular transport of LPS, which has been implicated in the pathogenesis of the "leaky gut" syndrome.
Collapse
Affiliation(s)
- Yasutada Akiba
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
- Brentwood Biomedical Research Institute, Los Angeles, California
| | - Koji Maruta
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Takeshi Takajo
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Kazuyuki Narimatsu
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Hyder Said
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Ikuo Kato
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Atsukazu Kuwahara
- Research Unit for Epithelial Physiology, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Japan
| | - Jonathan D Kaunitz
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
- Department of Surgery, University of California, School of Medicine, Los Angeles, California
- Brentwood Biomedical Research Institute, Los Angeles, California
| |
Collapse
|
64
|
Cytoskeletal Organization and Cell Polarity in the Pathogenesis of Crohn’s Disease. Clin Rev Allergy Immunol 2020; 60:164-174. [DOI: 10.1007/s12016-020-08795-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
65
|
Tabasi M, Ashrafian F, Khezerloo JK, Eshghjoo S, Behrouzi A, Javadinia SA, Poursadegh F, Eybpoosh S, Ahmadi S, Radmanesh A, Soroush A, Siadat SD. Changes in Gut Microbiota and Hormones After Bariatric Surgery: a Bench-to-Bedside Review. Obes Surg 2020; 29:1663-1674. [PMID: 30793228 DOI: 10.1007/s11695-019-03779-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Overweight and obesity are among the most prevalent non-communicable diseases which are generally treated successfully by bariatric or sleeve surgery. There are evidences affirming that sleeve surgery can manipulate the pH of the stomach and interact with the metabolism of fatty acids, carbohydrates, and bile acid transfer, leading to the overgrowth of gut microbiota. Therefore, this study aims to review the changes in gut microbiota and hormones after bariatric surgery.
Collapse
Affiliation(s)
- Mohsen Tabasi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave., Tehran, 13164, Iran
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Fatemeh Ashrafian
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Jamil Kheirvari Khezerloo
- Young Researchers and Elite Club, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Eshghjoo
- Microbial Pathogenesis and Immunology Department, Texas A&M University, Bryan, TX, USA
| | - Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Alireza Javadinia
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| | - Farid Poursadegh
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| | - Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Sara Ahmadi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Amin Radmanesh
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
66
|
The effects of different high-fat (lard, soybean oil, corn oil or olive oil) diets supplemented with fructo-oligosaccharides on colonic alkaline phosphatase activity in rats. Eur J Nutr 2020; 60:89-99. [PMID: 32193633 DOI: 10.1007/s00394-020-02219-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE We recently reported that fermentable non-digestible carbohydrates including fructo-oligosaccharides (FOS) commonly elevate colonic alkaline phosphatase (ALP) activity and the expression of IAP-I, an ALP gene, in rats fed a high-fat (HF) diet, and also elevate gut mucins and modulate gut microbiota. This study aims to investigate whether dietary fat types influence the effect of FOS on colonic ALP activity and the luminal environment in HF-fed rats. METHODS Male Sprague-Dawley rats were fed a diet containing 30% soybean oil, corn oil, olive oil or lard with or without 4% FOS for 2 weeks. Colon ALP activity, gene expression, and gut luminal variables including mucins and microbiota were measured. RESULTS In the lard diet groups, dietary FOS significantly elevated colonic ALP activity and the expression of IAP-I. The elevating effect of FOS on colonic ALP activity was also observed in the olive oil diet groups, although here the IAP-I expression was not changed. However, the soybean oil and corn oil diet groups did not exhibit the elevating effect of FOS on colon ALP. Fecal ALP and mucins were significantly elevated by dietary FOS regardless of dietary fat types, and the effect of FOS was prominent in the lard diet groups. The number of Lactobacillus spp. observed in fecal matter was significantly increased by dietary FOS in the lard and olive oil diet groups, but not in the soybean oil and corn oil diets groups. CONCLUSION This study suggests that dietary fat types may change the effect of FOS on the colonic luminal environment including the ALP activity in rats fed a high-fat diet.
Collapse
|
67
|
Lallès JP. Recent advances in intestinal alkaline phosphatase, inflammation, and nutrition. Nutr Rev 2020; 77:710-724. [PMID: 31086953 DOI: 10.1093/nutrit/nuz015] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In recent years, much new data on intestinal alkaline phosphatase (IAP) have been published, and major breakthroughs have been disclosed. The aim of the present review is to critically analyze the publications released over the last 5 years. These breakthroughs include, for example, the direct implication of IAP in intestinal tight junction integrity and barrier function maintenance; chronic intestinal challenge with low concentrations of Salmonella generating long-lasting depletion of IAP and increased susceptibility to inflammation; the suggestion that genetic mutations in the IAP gene in humans contribute to some forms of chronic inflammatory diseases and loss of functional IAP along the gut and in stools; stool IAP as an early biomarker of incipient diabetes in humans; and omega-3 fatty acids as direct inducers of IAP in intestinal tissue. Many recent papers have also explored the prophylactic and therapeutic potential of IAP and other alkaline phosphatase (AP) isoforms in various experimental settings and diseases. Remarkably, nearly all data confirm the potent anti-inflammatory properties of (I)AP and the negative consequences of its inhibition on health. A simplified model of the body AP system integrating the IAP compartment is provided. Finally, the list of nutrients and food components stimulating IAP has continued to grow, thus emphasizing nutrition as a potent lever for limiting inflammation.
Collapse
Affiliation(s)
- Jean-Paul Lallès
- Institut National de la Recherche Agronomique (INRA), Human Nutrition Division, Clermont-Ferrand, France, and the Centre de Recherche en Nutrition Humaine Ouest, Nantes, France
| |
Collapse
|
68
|
Zaher DM, El‐Gamal MI, Omar HA, Aljareh SN, Al‐Shamma SA, Ali AJ, Zaib S, Iqbal J. Recent advances with alkaline phosphatase isoenzymes and their inhibitors. Arch Pharm (Weinheim) 2020; 353:e2000011. [DOI: 10.1002/ardp.202000011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Dana M. Zaher
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
| | - Mohammed I. El‐Gamal
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
- College of PharmacySharjah United Arab Emirates
- Department of Medicinal ChemistryFaculty of PharmacyMansoura Egypt
| | - Hany A. Omar
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
- College of PharmacySharjah United Arab Emirates
- Department of PharmacologyFaculty of PharmacyBeni‐Suef Egypt
| | | | | | - Aya J. Ali
- College of PharmacySharjah United Arab Emirates
| | - Sumera Zaib
- Centre for Advanced Drug ResearchCOMSATS University Islamabad Abbottabad Campus Abbottabad Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug ResearchCOMSATS University Islamabad Abbottabad Campus Abbottabad Pakistan
| |
Collapse
|
69
|
Yang T, Li H, Oliveira AC, Goel R, Richards EM, Pepine CJ, Raizada MK. Transcriptomic signature of gut microbiome-contacting cells in colon of spontaneously hypertensive rats. Physiol Genomics 2020; 52:121-132. [PMID: 31869283 PMCID: PMC7099409 DOI: 10.1152/physiolgenomics.00087.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023] Open
Abstract
Fecal matter transfer from hypertensive patients and animals into normotensive animals increases blood pressure, strengthening the evidence for gut-microbiota interactions in the control of blood pressure. However, cellular and molecular events involved in gut dysbiosis-associated hypertension remain poorly understood. Therefore, our objective in this study was to use gene expression profiling to characterize the gut epithelium layer in the colon in hypertension. We observed significant suppression of components of T cell receptor (TCR) signaling in the colonic epithelium of spontaneously hypertensive rats (SHR) when compared with Wistar Kyoto (WKY) normotensive rats. Western blot analysis confirmed lower expression of key proteins including T cell surface glycoprotein CD3 gamma chain (Cd3g) and lymphocyte cytosolic protein 2 (Lcp2). Furthermore, lower expression of cytokines and receptors responsible for lymphocyte proliferation, differentiation, and activation (e.g., Il12r, Il15ra, Il7, Il16, Tgfb1) was observed in the colonic epithelium of the SHR. Finally, Alpi and its product, intestinal alkaline phosphatase, primarily localized in the epithelial cells, were profoundly lower in the SHR. These observations demonstrate that the colonic epithelium undergoes functional changes linked to altered immune, barrier function, and dysbiosis in hypertension.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Hongbao Li
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Aline C Oliveira
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Ruby Goel
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Elaine M Richards
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
70
|
Plaeke P, De Man JG, Smet A, Malhotra-Kumar S, Pintelon I, Timmermans JP, Nullens S, Jorens PG, Hubens G, De Winter BY. Effects of intestinal alkaline phosphatase on intestinal barrier function in a cecal ligation and puncture (CLP)-induced mouse model for sepsis. Neurogastroenterol Motil 2020; 32:e13754. [PMID: 31751495 DOI: 10.1111/nmo.13754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sepsis is a severe pathological condition associated with systemic inflammation, intestinal inflammation, and gastrointestinal barrier dysfunction. Intestinal alkaline phosphatase (IAP) has been demonstrated to detoxify lipopolysaccharide, an important mediator in the pathophysiology of sepsis. We investigated the effect of treatment with IAP on intestinal permeability, intestinal inflammation, and bacterial translocation. METHODS OF-1 mice were divided into 4 groups (n = 12/group), undergoing either a sham or cecal ligation and puncture (CLP) procedure to induce sepsis. Mice received IAP or a vehicle intraperitoneally 5 minutes prior to the onset of the CLP or sham procedure, which was repeated every 12 hours for two consecutive days. After two days, in vivo intestinal permeability, intestinal inflammation, and bacterial translocation were determined. KEY RESULTS CLP-induced sepsis resulted in significantly more weight loss, worse clinical disease scores, bacterial translocation, and elevated inflammatory cytokines. Intestinal permeability was increased up to 5-fold (P < .001). IAP activity was significantly increased in septic animals. Treatment with IAP had no effect on clinical outcomes but reduced the increased permeability of the small intestine by 50% (P = .005). This reduction in permeability was accompanied by a modified gene expression of claudin-1 (P = .025), claudin-14 (P = .035), and interleukin 12 (P = .015). A discriminant analysis showed that treatment with IAP is linked to modified mRNA levels of several tight junction proteins and cytokines. CONCLUSIONS AND INFERENCES Treatment with IAP diminished CLP-induced intestinal barrier disruption, associated with modified expression of several cytokines and claudins. Nevertheless, this effect did not translate into better clinical outcomes in our experimental setup.
Collapse
Affiliation(s)
- Philip Plaeke
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | | | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sara Nullens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Philippe G Jorens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium.,Department of Intensive Care Medicine, Antwerp University Hospital, Edegem (Antwerp), Belgium
| | - Guy Hubens
- Department of Abdominal Surgery, Antwerp University Hospital, Edegem (Antwerp), Belgium.,Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium.,Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
71
|
Development of chimeric peptides to facilitate the neutralisation of lipopolysaccharides during bactericidal targeting of multidrug-resistant Escherichia coli. Commun Biol 2020; 3:41. [PMID: 31974490 PMCID: PMC6978316 DOI: 10.1038/s42003-020-0761-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 12/02/2019] [Indexed: 11/16/2022] Open
Abstract
Pathogenic Escherichia coli can cause fatal diarrheal diseases in both animals and humans. However, no antibiotics or antimicrobial peptides (AMPs) can adequately kill resistant bacteria and clear bacterial endotoxin, lipopolysaccharide (LPS) which leads to inflammation and sepsis. Here, the LPS-targeted smart chimeric peptides (SCPs)-A6 and G6 are generated by connecting LPS-targeting peptide-LBP14 and killing domain-N6 via different linkers. Rigid and flexible linkers retain the independent biological activities from each component. SCPs-A6 and G6 exert low toxicity and no bacterial resistance, and they more rapidly kill multiple-drug-resistant E. coli and more effectively neutralize LPS toxicity than N6 alone. The SCPs can enhance mouse survival more effectively than N6 or polymyxin B and alleviate lung injuries by blocking mitogen-activated protein kinase and nuclear factor kappa-B p65 activation. These findings uniquely show that SCPs-A6 and G6 may be promising dual-function candidates as improved antibacterial and anti-endotoxin agents to treat bacterial infection and sepsis. Wang ZL and Wang XM design bactericidal peptides in which an antimicrobial domain is fused to a domain that facilitates the neutralisation of lipoplysaccaride (LPS) to prevent inflammation associated with the targeting of Gram-negative bacteria. They characterise their properties and structures, and show their efficiency in vitro and in vivo.
Collapse
|
72
|
Wan J, Tian Z, Yao BY, Liu C, He JN, Yin X, Shi Y. Role of intestinal alkaline phosphatase in intestinal mucosal barrier. Shijie Huaren Xiaohua Zazhi 2019; 27:1441-1445. [DOI: 10.11569/wcjd.v27.i23.1441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intestinal alkaline phosphatase (IAP) is an alkaline phosphatase that plays an important role in maintaining the stability of the bowel function and the intestinal mucosal barrier, including adjusting the duodenal pH, participating the development of the intestinal tract, regulating the absorption ability of intestinal epithelial cells, reducing the toxicity of lipopolysaccharide, preventing and reducing the intestinal inflammation, regulating intestinal flora, improving intestinal calcium absorption, etc. In this paper, we will review the role of IAP in intestinal mucosal barrier.
Collapse
Affiliation(s)
- Jun Wan
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Zhong Tian
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Bai-Yu Yao
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Chong Liu
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Jing-Ni He
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Xin Yin
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Yang Shi
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| |
Collapse
|
73
|
Moron R, Galvez J, Colmenero M, Anderson P, Cabeza J, Rodriguez-Cabezas ME. The Importance of the Microbiome in Critically Ill Patients: Role of Nutrition. Nutrients 2019; 11:E3002. [PMID: 31817895 PMCID: PMC6950228 DOI: 10.3390/nu11123002] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022] Open
Abstract
Critically ill patients have an alteration in the microbiome in which it becomes a disease-promoting pathobiome. It is characterized by lower bacterial diversity, loss of commensal phyla, like Firmicutes and Bacteroidetes, and a domination of pathogens belonging to the Proteobacteria phylum. Although these alterations are multicausal, many of the treatments administered to these patients, like antibiotics, play a significant role. Critically ill patients also have a hyperpermeable gut barrier and dysregulation of the inflammatory response that favor the development of the pathobiome, translocation of pathogens, and facilitate the emergence of sepsis. In order to restore the homeostasis of the microbiome, several nutritional strategies have been evaluated with the aim to improve the management of critically ill patients. Importantly, enteral nutrition has proven to be more efficient in promoting the homeostasis of the gut microbiome compared to parenteral nutrition. Several nutritional therapies, including prebiotics, probiotics, synbiotics, and fecal microbiota transplantation, are currently being used, showing variable results, possibly due to the unevenness of clinical trial conditions and the fact that the beneficial effects of probiotics are specific to particular species or even strains. Thus, it is of great importance to better understand the mechanisms by which nutrition and supplement therapies can heal the microbiome in critically ill patients in order to finally implement them in clinical practice with optimal safety and efficacy.
Collapse
Affiliation(s)
- Rocio Moron
- Servicio Farmacia Hospitalaria, Hospital Universitario Clínico San Cecilio, 18016-Granada, Spain; (R.M.); (J.C.)
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18012 Granada, Spain; (M.C.); (P.A.); (M.E.R.-C.)
| | - Julio Galvez
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18012 Granada, Spain; (M.C.); (P.A.); (M.E.R.-C.)
- Department of Pharmacology, CIBER-ehd, Center of Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain
| | - Manuel Colmenero
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18012 Granada, Spain; (M.C.); (P.A.); (M.E.R.-C.)
- Servicio de Medicina Intensiva, Hospital Universitaro Clinico San Cecilio, 18016 Granada, Spain
| | - Per Anderson
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18012 Granada, Spain; (M.C.); (P.A.); (M.E.R.-C.)
- Servicio de Análisis Clínicos e Inmunologia, UGC Laboratorio Clínico, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| | - José Cabeza
- Servicio Farmacia Hospitalaria, Hospital Universitario Clínico San Cecilio, 18016-Granada, Spain; (R.M.); (J.C.)
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18012 Granada, Spain; (M.C.); (P.A.); (M.E.R.-C.)
| | - Maria Elena Rodriguez-Cabezas
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18012 Granada, Spain; (M.C.); (P.A.); (M.E.R.-C.)
- Department of Pharmacology, CIBER-ehd, Center of Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain
| |
Collapse
|
74
|
Hinman SS, Wang Y, Allbritton NL. Photopatterned Membranes and Chemical Gradients Enable Scalable Phenotypic Organization of Primary Human Colon Epithelial Models. Anal Chem 2019; 91:15240-15247. [PMID: 31692334 DOI: 10.1021/acs.analchem.9b04217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Biochemical gradients across the intestinal epithelium play a major role in governing intestinal stem cell compartmentalization, differentiation dynamics, and organ-level self-renewal. However, scalable platforms that recapitulate the architecture and gradients present in vivo are absent. We present a platform in which individually addressable arrays of chemical gradients along the intestinal crypt long axis can be generated, enabling scalable culture of primary in vitro colonic epithelial replicas. The platform utilizes standardized well plate spacing, maintains access to basal and luminal compartments, and relies on a photopatterned porous membrane to act as diffusion windows while supporting the in vitro crypts. Simultaneous fabrication of 3875 crypts over a single membrane was developed. Growth factor gradients were modeled and then experimentally optimized to promote long-term health and self-renewal of the crypts which were assayed in situ by confocal fluorescence microscopy. The cultured in vitro crypt arrays successfully recapitulated the architecture and luminal-to-basal phenotypic polarity observed in vivo. Furthermore, known signaling regulators (e.g., butyrate and DAPT) produced measurable and predictable effects on the organized cell compartments, each decreasing crypt proliferation in the basal regions to negligible values. This platform is readily adaptable to the screening of tissue from individual patients to assay the impact of food and bacterial metabolites and/or drugs on colonic crypt dynamics. Importantly, the cassette is compatible with a wide range of sensing/detection modalities, and the developed fabrication methods should find applications for other cell and tissue types.
Collapse
Affiliation(s)
- Samuel S Hinman
- Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Yuli Wang
- Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Nancy L Allbritton
- Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States.,Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States , and North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
75
|
Komazin G, Maybin M, Woodard RW, Scior T, Schwudke D, Schombel U, Gisch N, Mamat U, Meredith TC. Substrate structure-activity relationship reveals a limited lipopolysaccharide chemotype range for intestinal alkaline phosphatase. J Biol Chem 2019; 294:19405-19423. [PMID: 31704704 DOI: 10.1074/jbc.ra119.010836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Lipopolysaccharide (LPS) from the Gram-negative bacterial outer membrane potently activates the human innate immune system. LPS is recognized by the Toll-like receptor 4/myeloid differentiation factor-2 (TLR4/MD2) complex, leading to the release of pro-inflammatory cytokines. Alkaline phosphatase (AP) is currently being investigated as an anti-inflammatory agent for detoxifying LPS through dephosphorylating lipid A, thus providing a potential treatment for managing both acute (sepsis) and chronic (metabolic endotoxemia) pathologies wherein aberrant TLR4/MD2 activation has been implicated. Endogenous LPS preparations are chemically heterogeneous, and little is known regarding the LPS chemotype substrate range of AP. Here, we investigated the activity of AP on a panel of structurally defined LPS chemotypes isolated from Escherichia coli and demonstrate that calf intestinal AP (cIAP) has only minimal activity against unmodified enteric LPS chemotypes. Pi was only released from a subset of LPS chemotypes harboring spontaneously labile phosphoethanolamine (PEtN) modifications connected through phosphoanhydride bonds. We demonstrate that the spontaneously hydrolyzed O-phosphorylethanolamine is the actual substrate for AP. We found that the 1- and 4'-lipid A phosphate groups critical in TLR4/MD2 signaling become susceptible to hydrolysis only after de-O-acylation of ester linked primary acyl chains on lipid A. Furthermore, PEtN modifications on lipid A specifically enhanced hTLR4 agonist activity of underacylated LPS preparations. Computational binding models are proposed to explain the limitation of AP substrate specificity imposed by the acylation state of lipid A, and the mechanism of PEtN in enhancing hTLR4/MD2 signaling.
Collapse
Affiliation(s)
- Gloria Komazin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Michael Maybin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Ronald W Woodard
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Thomas Scior
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico
| | - Dominik Schwudke
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Ursula Schombel
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Nicolas Gisch
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Uwe Mamat
- Cellular Microbiology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Timothy C Meredith
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
76
|
Ying M, Yu Q, Zheng B, Wang H, Wang J, Chen S, Gu Y, Nie S, Xie M. Cultured Cordyceps sinensis polysaccharides attenuate cyclophosphamide-induced intestinal barrier injury in mice. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103523] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
77
|
Molina-Castro SE, Tiffon C, Giraud J, Boeuf H, Sifre E, Giese A, Belleannée G, Lehours P, Bessède E, Mégraud F, Dubus P, Staedel C, Varon C. The Hippo Kinase LATS2 Controls Helicobacter pylori-Induced Epithelial-Mesenchymal Transition and Intestinal Metaplasia in Gastric Mucosa. Cell Mol Gastroenterol Hepatol 2019; 9:257-276. [PMID: 31669263 PMCID: PMC6957828 DOI: 10.1016/j.jcmgh.2019.10.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Gastric carcinoma is related mostly to CagA+-Helicobacter pylori infection, which disrupts the gastric mucosa turnover and elicits an epithelial-mesenchymal transition (EMT) and preneoplastic transdifferentiation. The tumor suppressor Hippo pathway controls stem cell homeostasis; its core, constituted by the large tumor suppressor 2 (LATS2) kinase and its substrate Yes-associated protein 1 (YAP1), was investigated in this context. METHODS Hippo, EMT, and intestinal metaplasia marker expression were investigated by transcriptomic and immunostaining analyses in human gastric AGS and MKN74 and nongastric immortalized RPE1 and HMLE epithelial cell lines challenged by H pylori, and on gastric tissues of infected patients and mice. LATS2 and YAP1 were silenced using small interfering RNAs. A transcriptional enhanced associated domain (TEAD) reporter assay was used. Cell proliferation and invasion were evaluated. RESULTS LATS2 and YAP1 appear co-overexpressed in the infected mucosa, especially in gastritis and intestinal metaplasia. H pylori via CagA stimulates LATS2 and YAP1 in a coordinated biphasic pattern, characterized by an early transient YAP1 nuclear accumulation and stimulated YAP1/TEAD transcription, followed by nuclear LATS2 up-regulation leading to YAP1 phosphorylation and targeting for degradation. LATS2 and YAP1 reciprocally positively regulate each other's expression. Loss-of-function experiments showed that LATS2 restricts H pylori-induced EMT marker expression, invasion, and intestinal metaplasia, supporting a role of LATS2 in maintaining the epithelial phenotype of gastric cells and constraining H pylori-induced preneoplastic changes. CONCLUSIONS H pylori infection engages a number of signaling cascades that alienate mucosa homeostasis, including the Hippo LATS2/YAP1/TEAD pathway. In the host-pathogen conflict, which generates an inflammatory environment and perturbations of the epithelial turnover and differentiation, Hippo signaling appears as a protective pathway, limiting the loss of gastric epithelial cell identity that precedes gastric carcinoma development.
Collapse
Affiliation(s)
- Silvia Elena Molina-Castro
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,University of Costa Rica, San José, Costa Rica
| | - Camille Tiffon
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | - Julie Giraud
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | - Hélène Boeuf
- INSERM, UMR1026, Bioingénierie tissulaire (BioTis), University of Bordeaux, Bordeaux, France
| | - Elodie Sifre
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | - Alban Giese
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | | | - Philippe Lehours
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Emilie Bessède
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Francis Mégraud
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Pierre Dubus
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Cathy Staedel
- INSERM, UMR1212, University of Bordeaux, Bordeaux, France,Cathy Staedel, PhD, INSERM U1212, “ARN: Régulations naturelle et artificielle” (ARNA)-Unités Mixtes de Recherche (UMR) Centre national de la recherche scientifique (CNRS) 5320, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France. fax: +33 5 57 57 10 15.
| | - Christine Varon
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Correspondence Address correspondence to: Christine Varon, PhD, INSERM U1053 Bordeaux Research in Translational Oncology (BaRITOn), University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France. fax: +33 5 56 79 60 18.
| |
Collapse
|
78
|
Opriessnig T, Karuppannan AK, Beckler D, Ait-Ali T, Cubas-Atienzar A, Halbur PG. Bacillus pumilus probiotic feed supplementation mitigates Lawsonia intracellularis shedding and lesions. Vet Res 2019; 50:85. [PMID: 31640784 PMCID: PMC6806562 DOI: 10.1186/s13567-019-0696-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/11/2019] [Indexed: 01/18/2023] Open
Abstract
The causative agent of ileitis, Lawsonia intracellularis, is commonly associated with diarrhea and reduced weight gain in growing pigs. The effect of in-feed probiotics on L. intracellularis infection dynamics was evaluated. In brief, 70 2.5-week-old-pigs were randomly divided into six groups with 10–20 pigs each. All pigs were fed an age appropriate base ration for the duration of the study, which was supplemented with one of three Bacillus strains including B. amyloliquefaciens (T01), B. licheniformis (T02) and B. pumilus (T03). Another group was orally vaccinated with a commercial live L. intracellularis vaccine (VAC) at 3 weeks of age. At 7 weeks of age, T01-LAW, T02-LAW, T03-LAW, VAC-LAW and the POS-CONTROL groups were challenged with L. intracellularis while the NEG-CONTROL pigs were not challenged. All pigs were necropsied 16 days later. By the time of inoculation, all VAC-LAW pigs had seroconverted and at necropsy 10–65% of the pigs in all other challenged groups were also seropositive. The results indicate a successful L. intracellularis challenge with highest bacterial DNA levels in POS-CONTROL pigs, VAC-LAW pigs and T01-LAW pigs. There was a delay in onset of shedding in T02-LAW and T03-LAW groups, which was reflected in less severe macroscopic and microscopic lesions, reduced intralesional L. intracellularis antigen levels and a lower area under the curve for bacterial shedding. Under the study conditions, two of the probiotics tested suppressed L. intracellularis infection. The obtained findings show the potential of probiotics in achieving antibiotic-free control of L. intracellularis.
Collapse
Affiliation(s)
- Tanja Opriessnig
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK. .,Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| | - Anbu K Karuppannan
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | | | - Tahar Ait-Ali
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Ana Cubas-Atienzar
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Patrick G Halbur
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| |
Collapse
|
79
|
Murdoch CC, Rawls JF. Commensal Microbiota Regulate Vertebrate Innate Immunity-Insights From the Zebrafish. Front Immunol 2019; 10:2100. [PMID: 31555292 PMCID: PMC6742977 DOI: 10.3389/fimmu.2019.02100] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022] Open
Abstract
Microbial communities populate the mucosal surfaces of all animals. Metazoans have co-evolved with these microorganisms, forming symbioses that affect the molecular and cellular underpinnings of animal physiology. These microorganisms, collectively referred to as the microbiota, are found on many distinct body sites (including the skin, nasal cavity, and urogenital tract), however the most densely colonized host tissue is the intestinal tract. Although spatially confined within the intestinal lumen, the microbiota and associated products shape the development and function of the host immune system. Studies comparing gnotobiotic animals devoid of any microbes (germ free) with counterparts colonized with selected microbial communities have demonstrated that commensal microorganisms are required for the proper development and function of the immune system at homeostasis and following infectious challenge or injury. Animal model systems have been essential for defining microbiota-dependent shifts in innate immune cell function and intestinal physiology during infection and disease. In particular, the zebrafish has emerged as a powerful vertebrate model organism with unparalleled capacity for in vivo imaging, a full complement of genetic approaches, and facile methods to experimentally manipulate microbial communities. Here we review key insights afforded by the zebrafish into the impact of microbiota on innate immunity, including evidence that the perception of and response to the microbiota is evolutionarily conserved. We also highlight opportunities to strengthen the zebrafish model system, and to gain new insights into microbiota-innate immune interactions that would be difficult to achieve in mammalian models.
Collapse
Affiliation(s)
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
80
|
Zhong JR, Feng L, Jiang WD, Wu P, Liu Y, Jiang J, Kuang SY, Tang L, Zhou XQ. Phytic acid disrupted intestinal immune status and suppressed growth performance in on-growing grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2019; 92:536-551. [PMID: 31247320 DOI: 10.1016/j.fsi.2019.06.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/23/2019] [Accepted: 06/23/2019] [Indexed: 06/09/2023]
Abstract
Phytic acid (PA) is one of the most common anti-nutritional factors in plant-derived protein feeds, and it poses considerable threats to aquaculture production. However, little is known about the effects of PA on fish intestinal health. This study aimed to investigate the impacts of PA on intestinal immune function in on-growing grass carp. To achieve this goal, a growth trial was conducted for 60 days by feeding 540 fish (120.56 ± 0.51 g) with six semi-purified diets containing graded levels of PA (0, 0.8, 1.6, 2.4, 3.2 and 4.0%). Then fish were challenged with Aeromonas hydrophila for 6 days. The results indicated that, compared with the control group (0% PA), PA did the following: (1) suppressed fish growth performance (percentage weight gain and feed efficiency) and reduced their ability to resist enteritis; (2) decreased fish intestinal antimicrobial ability by reducing intestinal lysozyme (LZ) activities, the contents of complement 3 (C3), C4 and immunoglobulin M (IgM), and downregulating the mRNA levels of hepcidin, liver-expressed antimicrobial peptide 2A (LEAP-2A), LEAP-2B, and β-defensin-1; and (3) aggravated fish intestinal inflammation responses by upregulating the mRNA levels of pro-inflammatory cytokines including tumour necrosis factor α (TNF-α), interleukin 1β (IL-1β) (except in the DI), interferon γ2 (IFN-γ2), IL-8, IL-12p40, IL-15 (except in the DI) and IL-17D, which is partly related to the nuclear factor kappa B (NF-κB) signalling pathway, whereas downregulating the mRNA levels of anti-inflammatory cytokines including transforming growth factor β1 (TGF-β1), IL-4/13A, IL-4/13B, IL-10 and IL-11, which is partially associated with the target of rapamycin (TOR) signalling pathway. The possible reasons for some distinctive gene expression patterns in fish three intestinal segments were discussed. Finally, based on the percent weight gain, enteritis morbidity, IgM content and LZ activity in the PI, the maximum tolerance levels of PA for on-growing grass carp were estimated to be 2.17, 1.68, 1.47 and 1.18% of the diet, respectively.
Collapse
Affiliation(s)
- Jing-Ren Zhong
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China.
| |
Collapse
|
81
|
Gonzalez CG, Tankou SK, Cox LM, Casavant EP, Weiner HL, Elias JE. Latent-period stool proteomic assay of multiple sclerosis model indicates protective capacity of host-expressed protease inhibitors. Sci Rep 2019; 9:12460. [PMID: 31462662 PMCID: PMC6713765 DOI: 10.1038/s41598-019-48495-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/02/2019] [Indexed: 01/20/2023] Open
Abstract
Diseases are often diagnosed once overt symptoms arise, ignoring the prior latent period when effective prevention may be possible. Experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, exhibits such disease latency, but the molecular processes underlying this asymptomatic period remain poorly characterized. Gut microbes also influence EAE severity, yet their impact on the latent period remains unknown. Here, we show the latent period between immunization and EAE's overt symptom onset is characterized by distinct host responses as measured by stool proteomics. In particular, we found a transient increase in protease inhibitors that inversely correlated with disease severity. Vancomycin administration attenuated both EAE symptoms and protease inhibitor induction potentially by decreasing immune system reactivity, supporting a subset of the microbiota's role in modulating the host's latent period response. These results strengthen previous evidence of proteases and their inhibitors in EAE and highlight the utility stool-omics for revealing complex, dynamic biology.
Collapse
Affiliation(s)
- Carlos G Gonzalez
- Chemical and Systems Biology Department, Stanford University School of Medicine, Stanford, USA
| | - Stephanie K Tankou
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard School of Medicine, Boston, MA, USA
- Department Of Neurology, Icahn School Of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School Of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School Of Medicine at Mount Sinai, New York, NY, USA
| | - Laura M Cox
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard School of Medicine, Boston, MA, USA
| | - Ellen P Casavant
- Chemical and Systems Biology Department, Stanford University School of Medicine, Stanford, USA
| | - Howard L Weiner
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard School of Medicine, Boston, MA, USA
| | - Joshua E Elias
- Chemical and Systems Biology Department, Stanford University School of Medicine, Stanford, USA.
- Chan Zuckerberg Biohub, San Francisco, California, USA.
| |
Collapse
|
82
|
Yang WH, Heithoff DM, Aziz PV, Haslund-Gourley B, Westman JS, Narisawa S, Pinkerton AB, Millán JL, Nizet V, Mahan MJ, Marth JD. Accelerated Aging and Clearance of Host Anti-inflammatory Enzymes by Discrete Pathogens Fuels Sepsis. Cell Host Microbe 2019; 24:500-513.e5. [PMID: 30308156 DOI: 10.1016/j.chom.2018.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/09/2018] [Accepted: 09/16/2018] [Indexed: 12/29/2022]
Abstract
Sepsis is a life-threatening inflammatory syndrome accompanying a bloodstream infection. Frequently secondary to pathogenic bacterial infections, sepsis remains difficult to treat as a singular disease mechanism. We compared the pathogenesis of murine sepsis experimentally elicited by five bacterial pathogens and report similarities among host responses to Gram-negative Salmonella and E. coli. We observed that a host protective mechanism involving de-toxification of lipopolysaccharide by circulating alkaline phosphatase (AP) isozymes was incapacitated during sepsis caused by Salmonella or E. coli through activation of host Toll-like receptor 4, which triggered Neu1 and Neu3 neuraminidase induction. Elevated neuraminidase activity accelerated the molecular aging and clearance of AP isozymes, thereby intensifying disease. Mice deficient in the sialyltransferase ST3Gal6 displayed increased disease severity, while deficiency of the endocytic lectin hepatic Ashwell-Morell receptor was protective. AP augmentation or neuraminidase inhibition diminished inflammation and promoted host survival. This study illuminates distinct routes of sepsis pathogenesis, which may inform therapeutic development.
Collapse
Affiliation(s)
- Won Ho Yang
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Douglas M Heithoff
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Peter V Aziz
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA; Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Benjamin Haslund-Gourley
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA; Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Julia S Westman
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Sonoko Narisawa
- Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Anthony B Pinkerton
- Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - José Luis Millán
- Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Michael J Mahan
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Jamey D Marth
- Center for Nanomedicine, University of California Santa Barbara, Santa Barbara, California 93106, USA; Sanford-Burham-Prebys Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, California 93106, USA; Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106, USA.
| |
Collapse
|
83
|
Zhao B, Che D, Adams S, Guo N, Han R, Zhang C, Qin G, Farouk MH, Jiang H. N-Acetyl-d-galactosamine prevents soya bean agglutinin-induced intestinal barrier dysfunction in intestinal porcine epithelial cells. J Anim Physiol Anim Nutr (Berl) 2019; 103:1198-1206. [PMID: 30934149 DOI: 10.1111/jpn.13091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/23/2022]
Abstract
Soya bean agglutinin (SBA) is a glycoprotein and the main anti-nutritional component in most soya bean feedstuffs. It is mainly a non-fibre carbohydrate-based protein and represents about 10% of soya bean-based anti-nutritional effects. In this study, we sought to determine the effects of N-Acetyl-D-galactosamine (GalNAc or D-GalNAc) on the damage induced by SBA on the membrane permeability and tight junction proteins of piglet intestinal epithelium (IPEC-J2) cells. The IPEC-J2 cells were pre-cultured with 0, 0.125 × 10-4 , 0.25 × 10-4 , 0.5 × 10-4 , 1.0 × 10-4 and 2.0 × 10-4 mmol/L GalNAc at different time period (1, 2, 4 and 8 hr) before being exposed to 0.5 mg/ml SBA for 24 hr. The results indicate that pre-incubation with GalNAc mitigates the mechanical barrier injury as reflected by a significant increase in trans-epithelial electric resistance (TEER) value and a decrease in alkaline phosphatase (ALP) activity in cell culture medium pre-treated with GalNAc before incubation with SBA as both indicate a reduction in cellular membrane permeability. In addition, mRNA levels of the tight junction proteins occludin and claudin-3 were lower in the SBA-treated groups without pre-treatment with GalNAc. The mRNA expression of occludin was reduced by 17.3% and claudin-3 by 42% (p < 0.01). Moreover, the corresponding protein expression levels were lowered by 17.8% and 43.5% (p < 0.05) respectively. However, in the GalNAc pre-treated groups, occludin and claudin-3 mRNAs were reduced by 1.6% (p > 0.05) and 2.7% (p < 0.01), respectively, while the corresponding proteins were reduced by 4.3% and 7.2% (p < 0.05). In conclusion, GalNAc may prevent the effect of SBA on membrane permeability and tight junction proteins on IPEC-J2s.
Collapse
Affiliation(s)
- Bao Zhao
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Dongsheng Che
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Seidu Adams
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Nan Guo
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Rui Han
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chun Zhang
- Department of Animal Science and Technology, Changchun University of science and technology, Changchun, China
| | - Guixin Qin
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Mohammed Hamdy Farouk
- Department of Animal Production, Faculty of Agriculture, Al-Azhar University, Cairo, Egypt
| | - Hailong Jiang
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
84
|
Boyd GW, Drew M, Ward S, Baird M, Connaboy C, Graham SM. The effect of the branched-chain amino acids on the in-vitro activity of bovine intestinal alkaline phosphatase. Appl Physiol Nutr Metab 2019; 44:632-636. [DOI: 10.1139/apnm-2018-0449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Branched-chain amino acids (BCAA) are used as nutritional support for patients with a range of conditions including liver cirrhosis and in-born errors of amino acid metabolism, and they are commonly used “sports” or exercise supplements. The effects of the BCAA on the in-vitro activity of calf intestinal alkaline phosphatase (EC. 3.1.3.1) were studied. All three BCAA were found to be uncompetitive inhibitors of the enzyme with L-leucine being the most potent ([Formula: see text] = 24.9 mmol/L) and L-valine, the least potent ([Formula: see text] = 37 mmol/L). Mixed BCAA are able to act in combination to inhibit the enzyme. Given the important role of intestinal alkaline phosphatase in gut homeostasis, these findings have potential implications for those taking high levels of BCAA as supplements.
Collapse
Affiliation(s)
- Gary William Boyd
- School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Marion Drew
- School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Shannon Ward
- School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Marianne Baird
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK
| | - Christopher Connaboy
- School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Scott Murray Graham
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK
| |
Collapse
|
85
|
Attili D, McClintock SD, Rizvi AH, Pandya S, Rehman H, Nadeem DM, Richter A, Thomas D, Dame MK, Turgeon DK, Varani J, Aslam MN. Calcium-induced differentiation in normal human colonoid cultures: Cell-cell / cell-matrix adhesion, barrier formation and tissue integrity. PLoS One 2019; 14:e0215122. [PMID: 30995271 PMCID: PMC6469792 DOI: 10.1371/journal.pone.0215122] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022] Open
Abstract
Background and aims The goal of the study was to assess calcium alone and Aquamin, a multi-mineral natural product that contains magnesium and detectable levels of 72 trace elements in addition to calcium, for capacity to affect growth and differentiation in colonoid cultures derived from histologically-normal human colon tissue. Methods Colonoid cultures were maintained in a low-calcium (0.25 mM) medium or in medium supplemented with an amount of calcium (1.5–3.0 mM), either from calcium alone or Aquamin for a period of two weeks. This was shown in a previous study to induce differentiation in colonoids derived from large adenomas. Changes in growth, morphological features and protein expression profile were assessed at the end of the incubation period using a combination of phase-contrast and scanning electron microscopy, histology and immunohistology, proteomic assessment and transmission electron microscopy. Results Unlike the previously-studied tumor-derived colonoids (which remained un-differentiated in the absence of calcium-supplementation), normal tissue colonoids underwent differentiation as indicated by gross and microscopic appearance, a low proliferative index and high-level expression of cytokeratin 20 in the absence of intervention (i.e., in control condition). Only modest additional changes were seen in these parameters with either calcium alone or Aquamin (providing up to 3.0 mM calcium). In spite of this, proteomic analysis and immunohistochemistry revealed that both interventions induced strong up-regulation of proteins that promote cell-cell and cell-matrix adhesive functions, barrier formation and tissue integrity. Transmission electron microscopy revealed an increase in desmosomes in response to intervention. Conclusions These findings demonstrate that colonoids derived from histologically normal human tissue can undergo differentiation in the presence of a low ambient calcium concentration. However, higher calcium levels induce elaboration of proteins that promote cell-cell and cell-matrix adhesion. These changes could lead to improved barrier function and improved colon tissue health.
Collapse
Affiliation(s)
- Durga Attili
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Shannon D. McClintock
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Areeba H. Rizvi
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Shailja Pandya
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Humza Rehman
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Daniyal M. Nadeem
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Aliah Richter
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Dafydd Thomas
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Michael K. Dame
- Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Danielle Kim Turgeon
- Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - James Varani
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Muhammad N. Aslam
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
86
|
Chandra L, Borcherding DC, Kingsbury D, Atherly T, Ambrosini YM, Bourgois-Mochel A, Yuan W, Kimber M, Qi Y, Wang Q, Wannemuehler M, Ellinwood NM, Snella E, Martin M, Skala M, Meyerholz D, Estes M, Fernandez-Zapico ME, Jergens AE, Mochel JP, Allenspach K. Derivation of adult canine intestinal organoids for translational research in gastroenterology. BMC Biol 2019; 17:33. [PMID: 30975131 PMCID: PMC6460554 DOI: 10.1186/s12915-019-0652-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022] Open
Abstract
Background Large animal models, such as the dog, are increasingly being used for studying diseases including gastrointestinal (GI) disorders. Dogs share similar environmental, genomic, anatomical, and intestinal physiologic features with humans. To bridge the gap between commonly used animal models, such as rodents, and humans, and expand the translational potential of the dog model, we developed a three-dimensional (3D) canine GI organoid (enteroid and colonoid) system. Organoids have recently gained interest in translational research as this model system better recapitulates the physiological and molecular features of the tissue environment in comparison with two-dimensional cultures. Results Organoids were derived from tissue of more than 40 healthy dogs and dogs with GI conditions, including inflammatory bowel disease (IBD) and intestinal carcinomas. Adult intestinal stem cells (ISC) were isolated from whole jejunal tissue as well as endoscopically obtained duodenal, ileal, and colonic biopsy samples using an optimized culture protocol. Intestinal organoids were comprehensively characterized using histology, immunohistochemistry, RNA in situ hybridization, and transmission electron microscopy, to determine the extent to which they recapitulated the in vivo tissue characteristics. Physiological relevance of the enteroid system was defined using functional assays such as optical metabolic imaging (OMI), the cystic fibrosis transmembrane conductance regulator (CFTR) function assay, and Exosome-Like Vesicles (EV) uptake assay, as a basis for wider applications of this technology in basic, preclinical and translational GI research. We have furthermore created a collection of cryopreserved organoids to facilitate future research. Conclusions We establish the canine GI organoid systems as a model to study naturally occurring intestinal diseases in dogs and humans, and that can be used for toxicology studies, for analysis of host-pathogen interactions, and for other translational applications. Electronic supplementary material The online version of this article (10.1186/s12915-019-0652-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lawrance Chandra
- Departments of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | | | - Dawn Kingsbury
- Departments of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | - Todd Atherly
- Departments of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | | | | | - Wang Yuan
- Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Michael Kimber
- Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Yijun Qi
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Qun Wang
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Michael Wannemuehler
- Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | | | | | | | - Melissa Skala
- Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - David Meyerholz
- Division of Comparative Pathology, University of Iowa Carver College of Medicine, Iowa City, USA
| | - Mary Estes
- Baylor College of Medicine, Houston, TX, USA
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Albert E Jergens
- Departments of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | | | - Karin Allenspach
- Departments of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA.
| |
Collapse
|
87
|
Dosh RH, Jordan-Mahy N, Sammon C, Le Maitre CL. Long-term in vitro 3D hydrogel co-culture model of inflammatory bowel disease. Sci Rep 2019; 9:1812. [PMID: 30755679 PMCID: PMC6372635 DOI: 10.1038/s41598-019-38524-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/13/2018] [Indexed: 12/26/2022] Open
Abstract
The in vitro study of the pathogenesis of inflammatory bowel disease (IBD) requires a cell model which closely reflects the characteristics of the in vivo intestinal epithelium. This study aimed to investigate the application of L-pNIPAM hydrogel as a scaffold to develop a long-term 3D co-culture model of Caco-2 and HT29-MTX cells under conditions analogous to inflammation, to determine its potential use in studying IBD. Monocultures and co-cultures were layered on L-pNIPAM hydrogel scaffolds and maintained under dynamic culture conditions for up to 12 weeks. Treatments with IL-1β, TNFα, and hypoxia for 1 week were used to create an inflammatory environment. Following prolonged culture, the metabolic activity of Caco-2 monoculture and 90% Caco-2/10% HT29-MTX co-cultures on L-pNIPAM hydrogels were increased, and finger-like structures, similar in appearance to villi were observed. Following treatment with IL-1β, TNFα and hypoxia, ALP and ZO-1 were decreased, MUC2 increased, and MUC5AC remained unchanged. ADAMTS1 was increased in response to hypoxia. Caspase 3 expression was increased in response to TNFα and hypoxic conditions. In conclusion, L-pNIPAM hydrogel supported long-term co-culture within a 3D model. Furthermore, stimulation with factors seen during inflammation recapitulated features seen during IBD.
Collapse
Affiliation(s)
- Rasha H Dosh
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB, UK.,Department of Anatomy and Histology, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Nicola Jordan-Mahy
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB, UK
| | - Christopher Sammon
- Materials and Engineering Research Institute, Sheffield Hallam University, Sheffield, S1 1WB, UK
| | - Christine L Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB, UK.
| |
Collapse
|
88
|
Zhou W, Davis EA, Dailey MJ. Obesity, independent of diet, drives lasting effects on intestinal epithelial stem cell proliferation in mice. Exp Biol Med (Maywood) 2019; 243:826-835. [PMID: 29932373 DOI: 10.1177/1535370218777762] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The intestinal epithelium plays an essential role in nutrient absorption, hormone release, and barrier function. Maintenance of the epithelium is driven by continuous cell renewal by intestinal epithelial stem cells located in the intestinal crypts. Obesity affects this process and results in changes in the size and function of the tissue. Because both the amount of food intake and the composition of the diet are contributing factors to developing and maintaining obesity, it is necessary to tease apart the separate contributions of obesity versus the type/amount of diet in driving the epithelial changes. C57BL/6J mice were fed a 60% high-fat diet versus a 10% low-fat diet for three months. A pair fed group was included (mice were fed with high-fat diet, but in equal kcal as that eaten by the low-fat diet- fed mice to keep them lean). We investigated the differences in (1) crypt-villus morphology in vivo, (2) the number and function of differentiated epithelial cell types in vivo, and (3) lasting effects on intestinal epithelial stem cell proliferation and growth in vitro. We found that high-fat diet-induced obesity, independent of the high-fat diet, increased crypt depth, villus height, the number of intestinal epithelial stem cells and goblet cells in vivo, and enhanced the size of the enterospheres developed from isolated IESCs in vitro. In addition, there is an interaction of obesity, type of diet, and availability of the diet (pair fed versus ad libitum) on protein and mRNA expression of alkaline phosphatase (an enzyme of enterocytes). These results suggest that high-fat diet-induced obesity, independent of the high-fat diet, induces lasting effects on intestinal epithelial stem cell proliferation, and drives the differentiation into goblet cells, but an interaction of obesity and diet drives alterations in the function of the enterocytes. Impact statement This study investigates whether obesity or the type/amount of diet differentially alters the proliferation, differentiation, growth, and function of the intestinal epithelial tissue. Although diet-induced obesity is known to alter the growth and function of the epithelium in vivo and cause lasting effects in intestinal epithelial stem cells (IESCs) in vitro, we are the first to tease apart the separate contributions of obesity versus the type/amount of diet in these processes. We found that high-fat diet (HFD)-induced obesity, independent of the HFD, drives lasting effects on IESC proliferation and differentiation into goblet cells, which may contribute to the growth of the epithelium. In addition, there is an interaction of obesity, type of diet, and availability of the diet (PF versus ad libitum) on the function of enterocytes. Identification of the factors driving the epithelial changes may provide new therapeutic strategies to control altered tissue growth and function associated with obesity.
Collapse
Affiliation(s)
- Weinan Zhou
- 1 Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Elizabeth A Davis
- 2 Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Megan J Dailey
- 1 Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,2 Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
89
|
Arabi Y, Jawdat D, Bouchama A, Tamim H, Tamimi W, Al-Balwi M, Al-Dorzi HM, Sadat M, Afesh L, Abdullah ML, Mashaqbeh W, Sakhija M, Hussein MA, ElObeid A, Al-Dawood A. Permissive underfeeding, cytokine profiles and outcomes in critically ill patients. PLoS One 2019; 14:e0209669. [PMID: 30615631 PMCID: PMC6322779 DOI: 10.1371/journal.pone.0209669] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 12/10/2018] [Indexed: 01/26/2023] Open
Abstract
Background During critical illness in humans, the effects of caloric restriction on the inflammatory response are not well understood. The aim of this study is to examine the associations of caloric restriction, inflammatory response profiles and outcomes in critically ill patients. Methods This is a sub-study of the PermiT trial (Permissive Underfeeding or Standard Enteral Feeding in Critically Ill Adults Trial- ISRCTN68144998). Serum samples were collected on study days 1, 3, 5, 7 and 14 and analyzed for a panel of 29 cytokines. We used principal component analysis to convert possibly correlated variables (cytokine levels) into a limited number of linearly uncorrelated variables (principal components). We constructed repeated measures mixed linear models to assess whether permissive underfeeding compared to standard feeding was associated with difference cytokine levels over time. Results A total of 72 critically ill patients were enrolled in this study (permissive underfeeding n = 36 and standard feeding n = 36). Principal component analysis identified 6 components that were responsible for 78% of the total variance. When adjusted to principal components, permissive underfeeding was not associated with 90-day mortality (adjusted odds ratio 1.75, 95% confidence interval 0.44, 6.95, p = 0.43) or with incident renal replacement therapy. The cytokines did not differ with time between permissive underfeeding and standard feeding groups. Conclusions The association of permissive underfeeding compared to standard feeding with mortality was not influenced by the inflammatory profile. Permissive underfeeding compared to standard feeding was not associated with differences in the serum levels of cytokines in critically ill patients.
Collapse
Affiliation(s)
- Yaseen Arabi
- Intensive Care Department, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- * E-mail: ,
| | - Dunia Jawdat
- Cord Blood Bank, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Abderrezak Bouchama
- Department of Experimental Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Hani Tamim
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Waleed Tamimi
- Department of Clinical Laboratory, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Mohammed Al-Balwi
- Department of Clinical Laboratory, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Hasan M. Al-Dorzi
- Intensive Care Department, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Musharaf Sadat
- Intensive Care Department, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Lara Afesh
- Intensive Care Department, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Mashan L. Abdullah
- Department of Experimental Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Walid Mashaqbeh
- Cord Blood Bank, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Maram Sakhija
- Intensive Care Department, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Mohamed A. Hussein
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Adila ElObeid
- Department of Biobank, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Abdulaziz Al-Dawood
- Intensive Care Department, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
90
|
Evaluation of Intestinal Damage Biomarkers in Calves with Atresia Coli. J Vet Res 2018; 62:379-384. [PMID: 30584620 PMCID: PMC6295999 DOI: 10.2478/jvetres-2018-0054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/23/2018] [Indexed: 11/20/2022] Open
Abstract
Introduction Intestinal obstruction such as atresia coli causes pathophysiological changes in gastrointestinal tissue due to the rise of intra-abdominal pressure. The aim of this study is to determine the intestinal damage with intestinal biomarkers in calves with atresia coli. Material and Methods The study was conducted on 40 Holstein calves diagnosed with atresia coli with mild to moderate abdominal distention and 10 healthy Holstein calves which served as the control. Blood samples were collected from all calves, and then serum concentrations of intestinal biomarkers were estimated, namely intestinal fatty acid binding protein (IFABP), liver fatty acid binding protein (LFABP), trefoil factor 3 (TFF3), and intestinal alkaline phosphatase (IAP), using commercially available specific bovine ELISA kits. An automatic blood gas analyser was employed for determining the lactate concentration. Results The concentrations of serum LFABP (P < 0.01), IFABP, TFF3, IAP, and blood lactate (P < 0.001) were significantly higher in calves with atresia coli than in healthy calves. Conclusion The calves affected with atresia coli exhibited severe intestinal damage, and IFABP, LFABP, and TFF3 have significant diagnostic importance and play a useful role in determining the intestinal damage due to intestinal obstruction. High levels of IAP and lactate may serve as a signal for the development of intestinal injury.
Collapse
|
91
|
Iqbal J, Ejaz SA, Ibrar A, Umar MI, Lecka J, Sévigny J, Saeed A. Expanding the Alkaline Phosphatase Inhibition, Cytotoxic and Proapoptotic Profile of Biscoumarin‐Iminothiazole and Coumarin‐Triazolothiadiazine Conjugates. ChemistrySelect 2018. [DOI: 10.1002/slct.201801863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jamshed Iqbal
- Centre for Advanced Drug ResearchCOMSATS University IslamabadAbbottabad Campus Abbottabad-22060 Pakistan
| | - Syeda Abida Ejaz
- Centre for Advanced Drug ResearchCOMSATS University IslamabadAbbottabad Campus Abbottabad-22060 Pakistan
| | - Aliya Ibrar
- Department of ChemistryQuaid-i-Azam University Islamabad-45320 Pakistan
| | - Muhammad Ihtisham Umar
- Department of PharmacyCOMSATS University IslamabadLahore Campus Defence Road Lahore-54000 Pakistan
| | - Joanna Lecka
- Département de microbiologie-infectiologie et d'immunologieFaculté de MédecineUniversité Laval, Québec, QC G1 V 0 A6 Canada
- Centre de Recherche du CHU de Québec – Université LavalQuébec, QC G1 V 4G2 Canada
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologieFaculté de MédecineUniversité Laval, Québec, QC G1 V 0 A6 Canada
- Centre de Recherche du CHU de Québec – Université LavalQuébec, QC G1 V 4G2 Canada
| | - Aamer Saeed
- Department of ChemistryQuaid-i-Azam University Islamabad-45320 Pakistan
| |
Collapse
|
92
|
Abstract
PURPOSE OF REVIEW To summarize the current understanding and recent advances on the genetic aetiology in the pathogenesis of very early onset inflammatory bowel disease (VEO-IBD). RECENT FINDINGS IBD is a chronic disorder of the gastrointestinal tract whose manifestation is a result of complex interactions between genetics, environment, immune system and microbial flora. Over 230 IBD risk loci have been reported in genome wide association studies but the genetic contribution of the majority of these loci in the manifestation of IBD is very low. Patients with VEO-IBD present with a more severe disease than older patients, characterized by poor prognosis and failure of conventional therapy. Recent studies have reported several monogenic diseases with high penetrance that present with IBD and IBD-like intestinal manifestations and overlap with primary immunodeficiencies. Increasing body of evidence supports a prominent role of genetics in the onset of VEO-IBD. New genetic variants and diagnoses in VEO-IBD are reviewed and current challenges in therapy with potential strategy to manage the disease are discussed. SUMMARY Functional analysis of the genes implicated in monogenic IBD has increased the understanding of the underlying pathobiological mechanism of the disease. This knowledge can be used to personalize medicine for specific patients, improving the standard of care and quality of life.
Collapse
Affiliation(s)
- Vritika Batura
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Institute for Medical Science and Biochemistry, University of Toronto, Hospital for Sick Children,Toronto, Ontario, Canada
| | - Aleixo M Muise
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Institute for Medical Science and Biochemistry, University of Toronto, Hospital for Sick Children,Toronto, Ontario, Canada
| |
Collapse
|
93
|
Moreno-Olivas F, Tako E, Mahler GJ. ZnO nanoparticles affect nutrient transport in an in vitro model of the small intestine. Food Chem Toxicol 2018; 124:112-127. [PMID: 30503572 DOI: 10.1016/j.fct.2018.11.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/31/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022]
Abstract
Nano-sized zinc oxide (ZnO) is present in food packaging, putting consumers at risk of ingestion. There is little information on the amount of ZnO nanoparticles (NP) present in food packaging and the effects of ZnO NP ingestion on intestinal function. To estimate physiologically relevant ZnO NP exposures from food that are commonly packaged with ZnO NP, food samples were analyzed with inductively coupled plasma mass spectrometry (ICP-MS). An in vitro model of the small intestine was used to investigate the effects of ZnO NP exposure. Cells were exposed to pristine NP in culture medium and to NP subjected to an in vitro digestion process to better reflect the transformation that the NP undergo in the human gastrointestinal (GI) tract. The findings show that a physiologically relevant dose of ZnO NP can cause a significant decrease in glucose transport, which is consistent with gene expression changes for the basolateral glucose transporter GLUT2. There is also evidence that the ZnO NP affect the microvilli of the intestinal cells, therefore reducing the amount of surface area available to absorb nutrients. These results suggest that the ingestion of ZnO NP can alter nutrient absorption in an in vitro model of the human small intestine.
Collapse
Affiliation(s)
- Fabiola Moreno-Olivas
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Elad Tako
- Plant, Soil and Nutrition Laboratory, Agricultural Research Services, U.S. Department of Agriculture, Ithaca, NY, 14850, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902, USA.
| |
Collapse
|
94
|
Pereira MT, Malik M, Nostro JA, Mahler GJ, Musselman LP. Effect of dietary additives on intestinal permeability in both Drosophila and a human cell co-culture. Dis Model Mech 2018; 11:dmm034520. [PMID: 30504122 PMCID: PMC6307910 DOI: 10.1242/dmm.034520] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/06/2018] [Indexed: 12/13/2022] Open
Abstract
Increased intestinal barrier permeability has been correlated with aging and disease, including type 2 diabetes, Crohn's disease, celiac disease, multiple sclerosis and irritable bowel syndrome. The prevalence of these ailments has risen together with an increase in industrial food processing and food additive consumption. Additives, including sugar, metal oxide nanoparticles, surfactants and sodium chloride, have all been suggested to increase intestinal permeability. We used two complementary model systems to examine the effects of food additives on gut barrier function: a Drosophila in vivo model and an in vitro human cell co-culture model. Of the additives tested, intestinal permeability was increased most dramatically by high sugar. High sugar also increased feeding but reduced gut and overall animal size. We also examined how food additives affected the activity of a gut mucosal defense factor, intestinal alkaline phosphatase (IAP), which fluctuates with bacterial load and affects intestinal permeability. We found that high sugar reduced IAP activity in both models. Artificial manipulation of the microbiome influenced gut permeability in both models, revealing a complex relationship between the two. This study extends previous work in flies and humans showing that diet can play a role in the health of the gut barrier. Moreover, simple models can be used to study mechanisms underlying the effects of diet on gut permeability and function.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Matthew T Pereira
- Department of Biological Sciences, Binghamton University, Binghamton, New York 13902, USA
| | - Mridu Malik
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York 13902, USA
| | - Jillian A Nostro
- Department of Biological Sciences, Binghamton University, Binghamton, New York 13902, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York 13902, USA
| | | |
Collapse
|
95
|
Consumption of non-digestible oligosaccharides elevates colonic alkaline phosphatase activity by up-regulating the expression of IAP-I, with increased mucins and microbial fermentation in rats fed a high-fat diet. Br J Nutr 2018; 121:146-154. [PMID: 30400998 DOI: 10.1017/s0007114518003082] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We have recently reported that soluble dietary fibre, glucomannan, increased colonic alkaline phosphatase (ALP) activity and the gene expression without affecting the small-intestinal activity and that colonic ALP was correlated with gut mucins (index of intestinal barrier function). We speculated that dietary fermentable carbohydrates including oligosaccharides commonly elevate colonic ALP and gene expression as well as increase mucin secretion and microbial fermentation. To test this hypothesis, male Sprague-Dawley rats were fed a diet containing 30 % lard with or without 4 % fructo-oligosaccharides (FOS), galacto-oligosaccharides (GOS), raffinose (RAF) and lactulose (LAC), which are non-digestible oligosaccharides or isomalto-oligosaccharides (IMOS; some digestible oligosaccharides) for 2 weeks. Colon ALP activity, the gene expression and gut luminal variables including mucins, organic acids and microbiota were measured. Colonic ALP was significantly elevated in the FOS, RAF and LAC groups, and a similar trend was observed in the GOS group. Colonic expression of intestinal alkaline phosphatase (IAP -I), an ALP gene, was significantly elevated in the FOS, GOS and RAF groups and tended to be increased in the LAC group. Dietary FOS, GOS, RAF and LAC significantly elevated faecal mucins, caecal n-butyrate and faecal ratio of Bifidobacterium spp. Dietary IMOS had no effect on colonic ALP, mucins, organic acids and microbiota. Colon ALP was correlated with mucins, caecal n-butyrate and faecal Bifidobacterium spp. This study demonstrated that non-digestible and fermentable oligosaccharides commonly elevate colonic ALP activity and the expression of IAP-I, with increasing mucins and microbial fermentation, which might be important for protection of gut epithelial homoeostasis.
Collapse
|
96
|
Monaco MH, Wang M, Pan X, Li Q, Richards JD, Chichlowski M, Berg BM, Dilger RN, Donovan SM. Evaluation of Sialyllactose Supplementation of a Prebiotic-Containing Formula on Growth, Intestinal Development, and Bacterial Colonization in the Neonatal Piglet. Curr Dev Nutr 2018; 2:nzy067. [PMID: 30443641 PMCID: PMC6226774 DOI: 10.1093/cdn/nzy067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/06/2018] [Accepted: 07/26/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Sialyllactose (SL) is a highly abundant oligosaccharide in human milk that has been shown to influence intestinal maturation and cognitive development and exert bifidogenic effects on the gut microbiota. The SL content of infant formula is significantly less than that of human milk, therefore there is interest in determining the effect of supplementing SL to infant formula at the levels in human milk on neonatal outcomes. OBJECTIVE The aim of this study was to investigate the effect of varying doses of dietary SL compared with a milk replacer formula on weight gain, gastrointestinal development, and microbiota composition in piglets. METHODS Thirty-eight intact male piglets were randomly assigned to 1 of 4 experimental diets from 2 to 32-33 d of age. Diets were formulated to contain SL at 0 mg/L (CON), 130 mg/L (LOW), 380 mg/L (MOD), or 760 mg/L (HIGH). At 32-33 d of age, blood was collected for serum chemistry and blood cellular analyses, and coagulation time. Immediately after humane killing, the small intestine was excised and intestinal segments fixed for quantification of mucin-producing goblet cells and morphologic analysis. In addition, mucosal disaccharide activity was assessed. Colonic luminal contents and feces were collected for measurement of pH, dry matter, volatile fatty acids, and the microbiota. RESULTS SL at ≤760 mg/L supported normal growth, intestinal development, and enzyme activity as well as serum chemistries and hematology (P > 0.05). In addition, SL supplementation did not affect overall microbiota structure and diversity in ascending colon contents and feces, but had minor effects on the relative abundances of specific microbes. CONCLUSIONS The findings in this study demonstrate that SL addition to a prebiotic-containing formula was well-tolerated by neonatal piglets, supported normal growth, and did not result in any adverse effects on serum chemistries or intestinal development.
Collapse
Affiliation(s)
- Marcia H Monaco
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL
| | - Xiao Pan
- Mead Johnson Pediatric Nutrition Institute, Evansville, IN
| | - Qian Li
- Mead Johnson Pediatric Nutrition Institute, Evansville, IN
| | | | | | - Brian M Berg
- Division of Nutritional Sciences, University of Illinois, Urbana, IL
- Mead Johnson Pediatric Nutrition Institute, Evansville, IN
| | - Ryan N Dilger
- Division of Nutritional Sciences, University of Illinois, Urbana, IL
- Department of Animal Sciences, University of Illinois, Urbana, IL
| | - Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL
- Division of Nutritional Sciences, University of Illinois, Urbana, IL
| |
Collapse
|
97
|
Brun LR, Lombarte M, Roma S, Perez F, Millán JL, Rigalli A. Increased calcium uptake and improved trabecular bone properties in intestinal alkaline phosphatase knockout mice. J Bone Miner Metab 2018; 36:661-667. [PMID: 29234952 PMCID: PMC6338327 DOI: 10.1007/s00774-017-0887-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 11/15/2017] [Indexed: 01/02/2023]
Abstract
Previous studies have demonstrated a negative correlation between intestinal alkaline phosphatase (IAP) activity and calcium (Ca) absorption in the gut, as IAP acts as a protective mechanism inhibiting high Ca entry into enterocytes, preventing Ca overload. Here we evaluated Ca absorption and bone properties in knockout mice (KO) completely devoid of duodenal IAP (Akp3 -/- mice). Female C57BL/6 control mice (WT, n = 7) and KO mice (n = 10) were used to determine Ca absorption in vivo and by in situ isolated duodenal loops followed by histomorphometric analysis of duodenal villi and crypts. Bone mineral density, morphometry, histomorphometry and trabecular connectivity and biomechanical properties were measured on bones. We observed mild atrophy of the villi with lower absorption surface and a significantly higher Ca uptake in KO mice. While no changes were seen in cortical bone, we found better trabecular connectivity and biomechanical properties in the femurs of KO mice compared to WT mice. Our data indicate that IAP KO mice display higher intestinal Ca uptake, which over time appears to correlate with a positive effect on the biomechanical properties of trabecular bone.
Collapse
Affiliation(s)
- Lucas R Brun
- Bone Biology Laboratory, Cátedra de Química Biológica, Facultad de Ciencias Médicas, School of Medicine, Rosario National University, Santa Fe 3100, 2000, Rosario, Argentina.
- National Council of Scientific and Technical Research (CONICET), Rosario, Argentina.
| | - M Lombarte
- Bone Biology Laboratory, Cátedra de Química Biológica, Facultad de Ciencias Médicas, School of Medicine, Rosario National University, Santa Fe 3100, 2000, Rosario, Argentina
- National Council of Scientific and Technical Research (CONICET), Rosario, Argentina
| | - S Roma
- Histology and Embryology Department, School of Medicine, Rosario National University, Rosario, Argentina
| | - F Perez
- Histology and Embryology Department, School of Medicine, Rosario National University, Rosario, Argentina
| | - J L Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - A Rigalli
- Bone Biology Laboratory, Cátedra de Química Biológica, Facultad de Ciencias Médicas, School of Medicine, Rosario National University, Santa Fe 3100, 2000, Rosario, Argentina
- National Council of Scientific and Technical Research (CONICET), Rosario, Argentina
- Rosario National University Research Council, Rosario, Argentina
| |
Collapse
|
98
|
Gámez-Belmonte R, Hernández-Chirlaque C, Sánchez de Medina F, Martínez-Augustin O. Experimental acute pancreatitis is enhanced in mice with tissue nonspecific alkaline phoshatase haplodeficiency due to modulation of neutrophils and acinar cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3769-3779. [DOI: 10.1016/j.bbadis.2018.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/31/2018] [Accepted: 09/09/2018] [Indexed: 01/13/2023]
|
99
|
Presbitero A, Mancini E, Brands R, Krzhizhanovskaya VV, Sloot PMA. Supplemented Alkaline Phosphatase Supports the Immune Response in Patients Undergoing Cardiac Surgery: Clinical and Computational Evidence. Front Immunol 2018; 9:2342. [PMID: 30364262 PMCID: PMC6193081 DOI: 10.3389/fimmu.2018.02342] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/20/2018] [Indexed: 01/29/2023] Open
Abstract
Alkaline phosphatase (AP) is an enzyme that exhibits anti-inflammatory effects by dephosphorylating inflammation triggering moieties (ITMs) like bacterial lipopolysaccharides and extracellular nucleotides. AP administration aims to prevent and treat peri- and post-surgical ischemia reperfusion injury in cardiothoracic surgery patients. Recent studies reported that intravenous bolus administration and continuous infusion of AP in patients undergoing coronary artery bypass grafting with cardiac valve surgery induce an increased release of liver-type “tissue non-specific alkaline phosphatase” (TNAP) into the bloodstream. The release of liver-type TNAP into circulation could be the body's way of strengthening its defense against a massive ischemic insult. However, the underlying mechanism behind the induction of TNAP is still unclear. To obtain a deeper insight into the role of AP during surgery, we developed a mathematical model of systemic inflammation that clarifies the relation between supplemented AP and TNAP and describes a plausible induction mechanism of TNAP in patients undergoing cardiothoracic surgery. The model was validated against clinical data from patients treated with bovine Intestinal AP (bIAP treatment) or without AP (placebo treatment), in addition to standard care procedures. We performed additional in-silico experiments adding a secondary source of ITMs after surgery, as observed in some patients with complications, and predicted the response to different AP treatment regimens. Our results show a strong protective effect of supplemented AP for patients with complications. The model provides evidence of the existence of an induction mechanism of liver-type tissue non-specific alkaline phosphatase, triggered by the supplementation of AP in patients undergoing cardiac surgery. To the best of our knowledge this is the first time that a quantitative and validated numerical model of systemic inflammation under clinical treatment conditions is presented.
Collapse
Affiliation(s)
- Alva Presbitero
- High Performance Computing Department, ITMO University, Saint Petersburg, Russia
| | - Emiliano Mancini
- Institute for Advanced Studies and Computational Science Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Ruud Brands
- Complexity Institute, Nanyang Technological University, Singapore, Singapore.,Alloksys Life Sciences BV, Wageningen, Netherlands
| | - Valeria V Krzhizhanovskaya
- High Performance Computing Department, ITMO University, Saint Petersburg, Russia.,Institute for Advanced Studies and Computational Science Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Peter M A Sloot
- High Performance Computing Department, ITMO University, Saint Petersburg, Russia.,Institute for Advanced Studies and Computational Science Laboratory, University of Amsterdam, Amsterdam, Netherlands.,Complexity Institute, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
100
|
Chandrupatla DMSH, Molthoff CFM, Ritsema WIGR, Vos R, Elshof E, Matsuyama T, Low PS, Musters RJP, Hammond A, Windhorst AD, Lammertsma AA, van der Laken CJ, Brands R, Jansen G. Prophylactic and therapeutic activity of alkaline phosphatase in arthritic rats: single-agent effects of alkaline phosphatase and synergistic effects in combination with methotrexate. Transl Res 2018; 199:24-38. [PMID: 29802817 DOI: 10.1016/j.trsl.2018.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/09/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022]
Abstract
Alkaline phosphatase (AP) is a gate-keeper of innate immune system responses by detoxifying inflammation triggering moieties released from endogenous and external sources. We examined whether AP's broad mechanism of action constitutes a safe therapeutic, either as single agent or combined with methotrexate (MTX), for chronic inflammatory disorders, for example, rheumatoid arthritis (RA). A rat model for RA was used with repeated intra-articular methylated bovine serum albumin (mBSA) injections in 1 knee ("arthritic" knee), with the contralateral knee serving as internal control. AP (200 µg, subcut) was administered before mBSA injections (prophylactic setting) or after arthritis induction (therapeutic setting) or combined with MTX (0.3 mg/kg or 1 mg/kg; intraperitoneally). As end point of treatment outcome, macrophage infiltration in knees, liver, and spleen was assessed by immunohistochemistry (ED1 and ED2 expression), immunofluoresence (macrophage marker folate receptor-β [FRβ]), and [18F]fluoro-polyethylene glycol-folate positron emission tomography (PET) (macrophage imaging) and ex vivo tissue distribution. Single-agent AP treatment and combinations with MTX were well tolerated. Both prophylactic and therapeutic AP markedly reduced synovial macrophage infiltration in arthritic knees (ED1: 3.5- to 4-fold; ED2: 3.5- to 6-fold), comparable with MTX treatment. AP-MTX combinations slightly improved on single agent effects. PET monitoring and ex vivo tissue distribution studies corroborated the impact of AP, MTX, and AP-MTX on reducing synovial macrophage infiltration. Beyond localized articular effects, AP also revealed systemic anti-inflammatory effects by a 2-fold reduction of ED1, ED2, and FRβ+ macrophages in liver and spleen of arthritic rats. Collectively, single-agent AP and AP combined with MTX elicited local and systemic anti-arthritic activity in arthritic rats.
Collapse
Affiliation(s)
- Durga M S H Chandrupatla
- Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Carla F M Molthoff
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Wayne I G R Ritsema
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Ricardo Vos
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Eline Elshof
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Takami Matsuyama
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - René J P Musters
- Department of Physiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anthony Hammond
- Department of Rheumatology, KIMS Hospital, Kent, United Kingdom
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Conny J van der Laken
- Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Gerrit Jansen
- Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|