1
|
Han S, Luo Z, Bao S, Xiao Z, Xu W, Xie T, Shi C, Wang J, Shan J. Effects of excessive Platycodon grandiflorus root on gut microbiota and host co-metabolism in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119577. [PMID: 40058476 DOI: 10.1016/j.jep.2025.119577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Platycodon grandiflorus root, is a widely used herb in East Asia for treating respiratory diseases, but research on its oral safety is limited. AIM OF THE STUDY This study examines the potential adverse gastrointestinal reactions resulting from excessive consumption of Platycodon grandiflorus root (PR) and its effects on gut microbiota and host co-metabolism. MATERIALS AND METHODS This study evaluated the effects of different doses (1.5, 4.5, and 7.5 g/kg/day) of PR on ICR mice through gavage. Select the 7.5 g/kg/day dosage group and the control group to assess intestinal morphology and conduct histopathological studies. Examine inflammation-related factors and tight junction proteins using WB, qPCR, and ELISA. Additionally, perform 16S rDNA sequencing and metabolomic analyses to evaluate changes in gut microbiota and endogenous metabolites. Finally, the clearance of gut microbiota with antibiotics, the effects of excessive PR on mice were investigated. RESULTS Excessive intake of PR can lead to mortality in mice, as well as symptoms such as intestinal flatulence and slowed intestinal transit, suggesting the occurrence of chronic intestinal pseudo-obstruction accompanied by endotoxemia. It altered both α-diversity and β-diversity in the gut microbiota of mice, with increased relative abundances of Pseudomonadota, Verrucomicrobiota, Escherichia-Shigella, Akkermansia, Bacteroides, and Klebsiella, closely linked to intestinal obstruction and bacterial overgrowth. Excessive intake of PR also resulted in metabolic disturbances in mice, particularly in the levels of metabolites such as bate-hydroxybutyrate, 5,6-dihydrouracil, uridine, isoleucine, mannitol, bate-alanine, L-cysteine, L-tyrosine, and orotic acid, which may provide insights into the side effects associated with excessive consumption of PR. Clearing the gut microbiota significantly mitigated adverse effects on the intestines and restored metabolite levels. CONCLUSIONS This study demonstrates that excessive PR induces gut microbiota and metabolic disruption in normal mice, with the overgrowth of Gram-negative bacteria releasing LPS that impair smooth muscle contraction, leading to adverse effects such as chronic intestinal pseudo-obstruction.
Collapse
Affiliation(s)
- Shasha Han
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zichen Luo
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shihang Bao
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zihan Xiao
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weichen Xu
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Xie
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Shi
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jin Wang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; College of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jinjun Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
2
|
Mareș CR, Săsăran MO, Mărginean CO. Small Intestinal Bacterial Overgrowth and Childhood Malnutrition: A Comprehensive Review of Available Evidence. Nutrients 2024; 16:4319. [PMID: 39770940 PMCID: PMC11679674 DOI: 10.3390/nu16244319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome is essential for children's normal growth and development, with its formation aligning closely with key stages of growth. Factors like birth method, feeding practices, and antibiotic exposure significantly shape the composition and functionality of the infant gut microbiome. Small intestinal bacterial overgrowth (SIBO) involves an abnormal increase in bacteria within the small intestine. This overgrowth can interfere with digestion, impair nutrient absorption, and lead to both local and systemic inflammation, potentially contributing to malnutrition. In this review, we provide a comprehensive overview of the current understanding of the relationship between SIBO and malnutrition, with a particular focus on the pediatric population. SIBO seems to play an important role in nutrient malabsorption through the gut microbiome imbalance, local inflammation, and disruption of the mucosal intestinal barrier. Additionally, SIBO is more prevalent in digestive disorders linked to malabsorption and malnutrition. Different therapeutic strategies for addressing malnutrition-related SIBO have been proposed. While antibiotics are the primary treatment for SIBO, their effectiveness in promoting weight gain among malnourished children remains uncertain. Hence, future research directed at the impact of microbiome imbalance on nutrient intake and absorption could bring to light new strategies for the effective prevention and treatment of malnutrition.
Collapse
Affiliation(s)
- Cristina Roxana Mareș
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania; (C.R.M.); (C.O.M.)
| | - Maria Oana Săsăran
- Department of Pediatrics 3, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania; (C.R.M.); (C.O.M.)
| |
Collapse
|
3
|
Green N, Chan C, Ooi CY. The gastrointestinal microbiome, small bowel bacterial overgrowth, and microbiome modulators in cystic fibrosis. Pediatr Pulmonol 2024; 59 Suppl 1:S70-S80. [PMID: 39105345 DOI: 10.1002/ppul.26913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 08/07/2024]
Abstract
People with cystic fibrosis (pwCF) have an altered gastrointestinal microbiome. These individuals also demonstrate propensity toward developing small intestinal bacterial overgrowth (SIBO). The dysbiosis present has intestinal and extraintestinal implications, including potential links with the higher rates of gastrointestinal malignancies described in CF. Given these implications, there is growing interest in therapeutic options for microbiome modulation. Alternative therapies, including probiotics and prebiotics, and current CF transmembrane conductance regulator gene modulators are promising interventions for ameliorating gut microbiome dysfunction in pwCF. This article will characterize and discuss the current state of knowledge and expert opinions on gut dysbiosis and SIBO in the context of CF, before reviewing the current evidence supporting gut microbial modulating therapies in CF.
Collapse
Affiliation(s)
- Nicole Green
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Christopher Chan
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Chee Y Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
4
|
Teng L, Dedousis N, Adeshirlarijaney A, Kanshana JS, Liu M, Hodges CA, Kohan AB. Impaired intestinal free fatty acid transport followed by chylomicron malformation, not pancreatic insufficiency, cause metabolic defects in cystic fibrosis. J Lipid Res 2024; 65:100551. [PMID: 39002195 PMCID: PMC11301217 DOI: 10.1016/j.jlr.2024.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/03/2024] [Accepted: 04/29/2024] [Indexed: 07/15/2024] Open
Abstract
Intestinal disease is one of the earliest manifestations of cystic fibrosis (CF) in children and is closely tied to deficits in growth and nutrition, both of which are directly linked to future mortality. Patients are treated aggressively with pancreatic enzyme replacement therapy and a high-fat diet to circumvent fat malabsorption, but this does not reverse growth and nutritional defects. We hypothesized that defects in chylomicron production could explain why CF body weights and nutrition are so resistant to clinical treatments. We used gold standard intestinal lipid absorption and metabolism approaches, including mouse mesenteric lymph cannulation, in vivo chylomicron secretion kinetics, transmission electron microscopy, small intestinal organoids, and chylomicron metabolism assays to test this hypothesis. In mice expressing the G542X mutation in cystic fibrosis transmembrane conductance regulator (CFTR-/- mice), we find that defective FFA trafficking across the epithelium into enterocytes drives a chylomicron formation defect. Furthermore, G542X mice secrete small, triglyceride-poor chylomicrons into the lymph and blood. These defective chylomicrons are cleared into extraintestinal tissues at ∼10-fold faster than WT chylomicrons. This defect in FFA absorption resulting in dysfunctional chylomicrons cannot be explained by steatorrhea or pancreatic insufficiency and is maintained in primary small intestinal organoids treated with micellar lipids. These studies suggest that the ultrahigh-fat diet that most people with CF are counselled to follow may instead make steatorrhea and malabsorption defects worse by overloading the absorptive capacity of the CF small intestine.
Collapse
Affiliation(s)
- Lihong Teng
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nikolaos Dedousis
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aneseh Adeshirlarijaney
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jitendra S Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Craig A Hodges
- Department of Genetics and Genome Sciences and Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Alison B Kohan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Han A, Hudson-Paz C, Robinson BG, Becker L, Jacobson A, Kaltschmidt JA, Garrison JL, Bhatt AS, Monack DM. Temperature-dependent differences in mouse gut motility are mediated by stress. Lab Anim (NY) 2024; 53:148-159. [PMID: 38806681 PMCID: PMC11147774 DOI: 10.1038/s41684-024-01376-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 04/19/2024] [Indexed: 05/30/2024]
Abstract
Researchers have advocated elevating mouse housing temperatures from the conventional ~22 °C to the mouse thermoneutral point of 30 °C to enhance translational research. However, the impact of environmental temperature on mouse gastrointestinal physiology remains largely unexplored. Here we show that mice raised at 22 °C exhibit whole gut transit speed nearly twice as fast as those raised at 30 °C, primarily driven by a threefold increase in colon transit speed. Furthermore, gut microbiota composition differs between the two temperatures but does not dictate temperature-dependent differences in gut motility. Notably, increased stress signals from the hypothalamic-pituitary-adrenal axis at 22 °C have a pivotal role in mediating temperature-dependent differences in gut motility. Pharmacological and genetic depletion of the stress hormone corticotropin-releasing hormone slows gut motility in stressed 22 °C mice but has no comparable effect in relatively unstressed 30 °C mice. In conclusion, our findings highlight that colder mouse facility temperatures significantly increase gut motility through hormonal stress pathways.
Collapse
Affiliation(s)
- Alvin Han
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | | | - Beatriz G Robinson
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Laren Becker
- Department of Medicine (Gastroenterology and Hepatology), Stanford University, Stanford, CA, USA
| | - Amanda Jacobson
- Genentech Inc., Research and Early Development, Immunology Discovery, South San Francisco, CA, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L Garrison
- Buck Institute for Research on Aging, Novato, CA, USA
- Global Consortium for Reproductive Longevity & Equality, Novato, CA, USA
| | - Ami S Bhatt
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Denise M Monack
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Oliveira ICCS, Marinsek GP, Correia LVB, da Silva RCB, Castro IB, Mari RB. Tributyltin (TBT) toxicity: Effects on enteric neuronal plasticity and intestinal barrier of rats' duodenum. Auton Neurosci 2024; 253:103176. [PMID: 38669866 DOI: 10.1016/j.autneu.2024.103176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024]
Abstract
Tributyltin (TBT) is a biocide used in the formulation of antifouling paints and it is highly harmful. Despite the ban, the compound persists in the environment, contaminating marine foodstuffs and household products. Therefore, considering the route of exposure to the contaminant, the gastrointestinal tract (GIT) acts as an important barrier against harmful substances and is a potential biomarker for understanding the consequences of these agents. This work aimed to evaluate histological and neuronal alterations in the duodenum of male Wistar rats that received 20 ng/g TBT and 600 ng/g via gavage for 30 consecutive days. After the experimental period, the animals were euthanized, and the duodenum was intended for neuronal histochemistry (total and metabolically active populations) and histological routine (morphometry and histopathology). The results showed more severe changes in neuronal density and intestinal morphometry in rats exposed to 20 ng/g, such as total neuronal density decrease and reduction of intestinal layers. In rats exposed to 600 ng/g of TBT, it was possible to observe only an increase in intraepithelial lymphocytes. We conclude that TBT can be more harmful to intestinal homeostasis when consumed in lower concentrations.
Collapse
Affiliation(s)
- I C C S Oliveira
- UNESP- São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil.
| | - G P Marinsek
- UNESP- São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil.
| | - L V B Correia
- UNIFESP- Federal University of São Paulo, Institute of Health and Society, Baixada Santista Campus, Santos, SP, Brazil
| | - R C B da Silva
- UNIFESP- Federal University of São Paulo, Institute of Health and Society, Baixada Santista Campus, Santos, SP, Brazil
| | - I B Castro
- UNIFESP- Federal University of São Paulo, Institute of Marine Science, Baixada Santista Campus, Santos, SP, Brazil.
| | - R B Mari
- UNESP- São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil.
| |
Collapse
|
7
|
Roszkowska P, Klimczak E, Ostrycharz E, Rączka A, Wojciechowska-Koszko I, Dybus A, Cheng YH, Yu YH, Mazgaj S, Hukowska-Szematowicz B. Small Intestinal Bacterial Overgrowth (SIBO) and Twelve Groups of Related Diseases-Current State of Knowledge. Biomedicines 2024; 12:1030. [PMID: 38790992 PMCID: PMC11117733 DOI: 10.3390/biomedicines12051030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The human gut microbiota creates a complex microbial ecosystem, characterized by its high population density, wide diversity, and complex interactions. Any imbalance of the intestinal microbiome, whether qualitative or quantitative, may have serious consequences for human health, including small intestinal bacterial overgrowth (SIBO). SIBO is defined as an increase in the number of bacteria (103-105 CFU/mL), an alteration in the bacterial composition, or both in the small intestine. The PubMed, Science Direct, Web of Science, EMBASE, and Medline databases were searched for studies on SIBO and related diseases. These diseases were divided into 12 groups: (1) gastrointestinal disorders; (2) autoimmune disease; (3) cardiovascular system disease; (4) metabolic disease; (5) endocrine disorders; (6) nephrological disorders; (7) dermatological diseases; (8) neurological diseases (9); developmental disorders; (10) mental disorders; (11) genetic diseases; and (12) gastrointestinal cancer. The purpose of this comprehensive review is to present the current state of knowledge on the relationships between SIBO and these 12 disease groups, taking into account risk factors and the causal context. This review fills the evidence gap on SIBO and presents a biological-medical approach to the problem, clearly showing the groups and diseases having a proven relationship with SIBO, as well as indicating groups within which research should continue to be expanded.
Collapse
Affiliation(s)
- Paulina Roszkowska
- Department of Diagnostic Immunology, Pomeranian Medical University, st. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (P.R.); (I.W.-K.)
| | - Emilia Klimczak
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
| | - Ewa Ostrycharz
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
- Doctoral School, University of Szczecin, st. A. Mickiewicz 16, 71-412 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, st. Wąska 13, 71-412 Szczecin, Poland
| | - Aleksandra Rączka
- Department of Genetics, West Pomeranian University of Technology, st. Aleja Piastów 45, 70-311 Szczecin, Poland; (A.R.); (A.D.)
| | - Iwona Wojciechowska-Koszko
- Department of Diagnostic Immunology, Pomeranian Medical University, st. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (P.R.); (I.W.-K.)
| | - Andrzej Dybus
- Department of Genetics, West Pomeranian University of Technology, st. Aleja Piastów 45, 70-311 Szczecin, Poland; (A.R.); (A.D.)
| | - Yeong-Hsiang Cheng
- Department of Biotechnology and Animal Science, National Ilan University, Yilan 26047, Taiwan; (Y.-H.C.); (Y.-H.Y.)
| | - Yu-Hsiang Yu
- Department of Biotechnology and Animal Science, National Ilan University, Yilan 26047, Taiwan; (Y.-H.C.); (Y.-H.Y.)
| | - Szymon Mazgaj
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
| | - Beata Hukowska-Szematowicz
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
- Molecular Biology and Biotechnology Center, University of Szczecin, st. Wąska 13, 71-412 Szczecin, Poland
| |
Collapse
|
8
|
Robertson K, Hahn O, Robinson BG, Faruk AT, Janakiraman M, Namkoong H, Kim K, Ye J, Bishop ES, Hall RA, Wyss-Coray T, Becker LS, Kaltschmidt JA. Gpr37 modulates the severity of inflammation-induced GI dysmotility by regulating enteric reactive gliosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588619. [PMID: 38645163 PMCID: PMC11030428 DOI: 10.1101/2024.04.09.588619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The enteric nervous system (ENS) is contained within two layers of the gut wall and is made up of neurons, immune cells, and enteric glia cells (EGCs) that regulate gastrointestinal (GI) function. EGCs in both inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) change in response to inflammation, referred to as reactive gliosis. Whether EGCs restricted to a specific layer or region within the GI tract alone can influence intestinal immune response is unknown. Using bulk RNA-sequencing and in situ hybridization, we identify G-protein coupled receptor Gpr37 , as a gene expressed only in EGCs of the myenteric plexus, one of the two layers of the ENS. We show that Gpr37 contributes to key components of LPS-induced reactive gliosis including activation of NF-kB and IFN-y signaling and response genes, lymphocyte recruitment, and inflammation-induced GI dysmotility. Targeting Gpr37 in EGCs presents a potential avenue for modifying inflammatory processes in the ENS.
Collapse
|
9
|
Young SM, Woode RA, Williams EC, Ericsson AC, Clarke LL. Fecal dysbiosis and inflammation in intestinal-specific Cftr knockout mice on regimens preventing intestinal obstruction. Physiol Genomics 2024; 56:247-264. [PMID: 38073491 PMCID: PMC11283905 DOI: 10.1152/physiolgenomics.00077.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/08/2023] [Accepted: 12/03/2023] [Indexed: 12/26/2023] Open
Abstract
Chronic intestinal inflammation is a poorly understood manifestation of cystic fibrosis (CF), which may be refractory to ion channel CF transmembrane conductance regulator (CFTR) modulator therapy. People with CF exhibit intestinal dysbiosis, which has the potential for stimulating intestinal and systemic inflammation. CFTR is expressed in organ epithelia, leukocytes, and other tissues. Here, we investigate the contribution of intestinal epithelium-specific loss of Cftr [iCftr knockout (KO)] to dysbiosis and inflammation in mice treated with either of two antiobstructive dietary regimens necessary to maintain CF mouse models [polyethylene glycol (PEG) laxative or a liquid diet (LiqD)]. Feces collected from iCftr KO mice and their wild-type (WT) sex-matched littermates were used to measure fecal calprotectin to evaluate inflammation and to perform 16S rRNA sequencing to characterize the gut microbiome. Fecal calprotectin was elevated in iCftr KO relative to WT mice that consumed either PEG or LiqD. PEG iCftr KO mice did not show a change in α diversity versus WT mice but demonstrated a significant difference in microbial composition (β diversity) with included increases in the phylum Proteobacteria, the family Peptostreptococcaceae, four genera of Clostridia including C. innocuum, and the mucolytic genus Akkermansia. Fecal microbiome analysis of LiqD-fed iCftr KO mice showed both decreased α diversity and differences in microbial composition with increases in the Proteobacteria family Enterobacteriaceae, Firmicutes families Clostridiaceae and Peptostreptococcaceae, and enrichment of Clostridium perfringens, C. innocuum, C. difficile, mucolytic Ruminococcus gnavus, and reduction of Akkermansia. It was concluded that epithelium-specific loss of Cftr is a major driver of CF intestinal dysbiosis and inflammation with significant similarities to previous studies of pan Cftr KO mice.NEW & NOTEWORTHY Chronic intestinal inflammation is a manifestation of cystic fibrosis (CF), a disease caused by loss of the anion channel CF transmembrane conductance regulator (CFTR) that is expressed in many tissues. This study shows that intestinal epithelial cell-specific loss of CFTR [inducible Cftr knockout (KO)] in mice is sufficient to induce intestinal dysbiosis and inflammation. Experiments were performed on mice consuming two dietary regimens routinely used to prevent obstruction in CF mice.
Collapse
Affiliation(s)
- Sarah M Young
- College of Veterinary Medicine, University of Missouri Comparative Medicine Program, Columbia, Missouri, United States
| | - Rowena A Woode
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Estela C Williams
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Aaron C Ericsson
- College of Veterinary Medicine, University of Missouri Comparative Medicine Program, Columbia, Missouri, United States
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- College of Veterinary Medicine, University of Missouri Metagenomics Center, Columbia, Missouri, United States
| | - Lane L Clarke
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
10
|
Robinson BG, Oster BA, Robertson K, Kaltschmidt JA. Loss of ASD-related molecule Cntnap2 affects colonic motility in mice. Front Neurosci 2023; 17:1287057. [PMID: 38027494 PMCID: PMC10665486 DOI: 10.3389/fnins.2023.1287057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Gastrointestinal (GI) symptoms are highly prevalent among individuals with autism spectrum disorder (ASD), but the molecular link between ASD and GI dysfunction remains poorly understood. The enteric nervous system (ENS) is critical for normal GI motility and has been shown to be altered in mouse models of ASD and other neurological disorders. Contactin-associated protein-like 2 (Cntnap2) is an ASD-related synaptic cell-adhesion molecule important for sensory processing. In this study, we examine the role of Cntnap2 in GI motility by characterizing Cntnap2's expression in the ENS and assessing GI function in Cntnap2 mutant mice. We find Cntnap2 expression predominately in enteric sensory neurons. We further assess in vivo and ex vivo GI motility in Cntnap2 mutants and show altered transit time and colonic motility patterns. The overall organization of the ENS appears undisturbed. Our results suggest that Cntnap2 plays a role in GI function and may provide a molecular link between ASD and GI dysfunction.
Collapse
Affiliation(s)
- Beatriz G. Robinson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Beau A. Oster
- Nevada ENDURE Program, University of Nevada, Reno, Reno, NV, United States
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
11
|
Robinson BG, Oster BA, Robertson K, Kaltschmidt JA. Loss of ASD-Related Molecule Cntnap2 Affects Colonic Motility in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537221. [PMID: 37131706 PMCID: PMC10153124 DOI: 10.1101/2023.04.17.537221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gastrointestinal (GI) symptoms are highly prevalent among individuals with autism spectrum disorder (ASD), but the molecular link between ASD and GI dysfunction remains poorly understood. The enteric nervous system (ENS) is critical for normal GI motility and has been shown to be altered in mouse models of ASD and other neurological disorders. Contactin-associated protein-like 2 (Cntnap2) is an ASD-related synaptic cell-adhesion molecule important for sensory processing. In this study, we examine the role of Cntnap2 in GI motility by characterizing Cntnap2's expression in the ENS and assessing GI function in Cntnap2 mutant mice. We find Cntnap2 expression predominately in enteric sensory neurons. We further assess in-vivo and ex-vivo GI motility in Cntnap2 mutants and show altered transit time and colonic motility patterns. The overall organization of the ENS appears undisturbed. Our results suggest that Cntnap2 plays a role in GI function and may provide a molecular link between ASD and GI dysfunction.
Collapse
Affiliation(s)
- Beatriz G. Robinson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Beau A. Oster
- Nevada ENDURE Program, University of Nevada, Reno, Reno, NV 89557, USA
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Sroka N, Rydzewska-Rosołowska A, Kakareko K, Rosołowski M, Głowińska I, Hryszko T. Show Me What You Have Inside-The Complex Interplay between SIBO and Multiple Medical Conditions-A Systematic Review. Nutrients 2022; 15:nu15010090. [PMID: 36615748 PMCID: PMC9824151 DOI: 10.3390/nu15010090] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The microbiota, as a complex of microorganisms in a particular ecosystem, is part of the wider term-microbiome, which is defined as the set of all genetic content in the microbial community. Imbalanced gut microbiota has a great impact on the homeostasis of the organism. Dysbiosis, as a disturbance in bacterial balance, might trigger or exacerbate the course of different pathologies. Small intestinal bacterial overgrowth (SIBO) is a disorder characterized by differences in quantity, quality, and location of the small intestine microbiota. SIBO underlies symptoms associated with functional gastrointestinal disorders (FGD) as well as may alter the presentation of chronic diseases such as heart failure, diabetes, etc. In recent years there has been growing interest in the influence of SIBO and its impact on the whole human body as well as individual systems. Therefore, we aimed to investigate the co-existence of SIBO with different medical conditions. The PubMed database was searched up to July 2022 and we found 580 original studies; inclusion and exclusion criteria let us identify 112 eligible articles, which are quoted in this paper. The present SIBO diagnostic methods could be divided into two groups-invasive, the gold standard-small intestine aspirate culture, and non-invasive, breath tests (BT). Over the years scientists have explored SIBO and its associations with other diseases. Its role has been confirmed not only in gastroenterology but also in cardiology, endocrinology, neurology, rheumatology, and nephrology. Antibiotic therapy could reduce SIBO occurrence resulting not only in the relief of FGD symptoms but also manifestations of comorbid diseases. Although more research is needed, the link between SIBO and other diseases is an important pathway for scientists to follow.
Collapse
Affiliation(s)
- Natalia Sroka
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
- Correspondence:
| | - Alicja Rydzewska-Rosołowska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
| | - Katarzyna Kakareko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
| | - Mariusz Rosołowski
- Department of Internal Medicine and Hypertension, Medical University of Białystok, 15-540 Białystok, Poland
| | - Irena Głowińska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
| |
Collapse
|
13
|
Scull CE, Luo M, Jennings S, Taylor CM, Wang G. Cftr deletion in mouse epithelial and immune cells differentially influence the intestinal microbiota. Commun Biol 2022; 5:1130. [PMID: 36289287 PMCID: PMC9605958 DOI: 10.1038/s42003-022-04101-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022] Open
Abstract
Cystic fibrosis (CF) is a life-threatening genetic disorder, caused by mutations in the CF transmembrane-conductance regulator gene (cftr) that encodes CFTR, a cAMP-activated chloride and bicarbonate channel. Clinically, CF lung disease dominates the adult patient population. However, its gastrointestinal illness claims the early morbidity and mortality, manifesting as intestinal dysbiosis, inflammation and obstruction. As CF is widely accepted as a disease of epithelial dysfunction, it is unknown whether CFTR loss-of-function in immune cells contributes to these clinical outcomes. Using cftr genetic knockout and bone marrow transplantation mouse models, we performed 16S rRNA gene sequencing of the intestinal microbes. Here we show that cftr deletion in both epithelial and immune cells collectively influence the intestinal microbiota. However, the immune defect is a major factor determining the dysbiosis in the small intestine, while the epithelial defect largely influences that in the large intestine. This finding revises the current concept by suggesting that CF epithelial defect and immune defect play differential roles in CF intestinal disease.
Collapse
Affiliation(s)
- Callie E Scull
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Scott Jennings
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Christopher M Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Guoshun Wang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
14
|
Abstract
The intestinal tract is protected by epithelium-covering mucus, which is constantly renewed by goblet cells, a specialized type of epithelial cell. Mucus is largely composed of MUC2 mucin, an enormous molecule that poses a high demand on the endoplasmic reticulum (ER) for proper folding and protein assembly, creating a challenge for the secretory machinery in goblet cells. In this issue of the JCI, Grey et al. reveal that the ER resident protein and folding sensor ERN2 (also known as IRE1β) was instrumental for goblet cells to produce sufficient amounts of mucus to form a protective mucus layer. In the absence of ERN2, mucus production was reduced, impairing the mucus barrier, which allowed bacteria to penetrate and cause an epithelial cell stress response. This study emphasizes the importance of a controlled unfolded protein response (UPR) for goblet cell secretion.
Collapse
|
15
|
Tan X, Kini A, Römermann D, Seidler U. The NHE3 Inhibitor Tenapanor Prevents Intestinal Obstructions in CFTR-Deleted Mice. Int J Mol Sci 2022; 23:ijms23179993. [PMID: 36077390 PMCID: PMC9456459 DOI: 10.3390/ijms23179993] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022] Open
Abstract
Mutations in the CFTR chloride channel result in intestinal obstructive episodes in cystic fibrosis (CF) patients and in CF animal models. In this study, we explored the possibility of reducing the frequency of obstructive episodes in cftr−/− mice through the oral application of a gut-selective NHE3 inhibitor tenapanor and searched for the underlying mechanisms involved. Sex- and age-matched cftr+/+ and cftr−/− mice were orally gavaged twice daily with 30 mg kg−1 tenapanor or vehicle for a period of 21 days. Body weight and stool water content was assessed daily and gastrointestinal transit time (GTT) once weekly. The mice were sacrificed when an intestinal obstruction was suspected or after 21 days, and stool and tissues were collected for further analysis. Twenty-one day tenapanor application resulted in a significant increase in stool water content and stool alkalinity and a significant decrease in GTT in cftr+/+ and cftr−/− mice. Tenapanor significantly reduced obstructive episodes to 8% compared to 46% in vehicle-treated cftr−/− mice and prevented mucosal inflammation. A decrease in cryptal hyperproliferation, mucus accumulation, and mucosal mast cell number was also observed in tenapanor- compared to vehicle-treated, unobstructed cftr−/− mice. Overall, oral tenapanor application prevented obstructive episodes in CFTR-deficient mice and was safe in cftr+/+ and cftr−/− mice. These results suggest that tenapanor may be a safe and affordable adjunctive therapy in cystic fibrosis patients to alleviate constipation and prevent recurrent DIOS.
Collapse
Affiliation(s)
| | | | | | - Ursula Seidler
- Correspondence: ; Tel.: +49-5115-329-427; Fax: +49-5115-328-428
| |
Collapse
|
16
|
Changes in Glucose Breath Test in Cystic Fibrosis Patients Treated With 1 Month of Lumacaftor/Ivacaftor. J Pediatr Gastroenterol Nutr 2022; 75:42-47. [PMID: 35442228 DOI: 10.1097/mpg.0000000000003459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Alteration of the airway microbiota is a hallmark of cystic fibrosis (CF) pulmonary disease. Dysfunction of cystic fibrosis transmembrane regulator (CFTR) in the intestine also promotes changes in local microbiota such as small intestinal bacterial overgrowth (SIBO), which is common in CF. We evaluated whether therapy with the CFTR modulator combination lumacaftor/ivacaftor (luma/iva) has a beneficial impact on SIBO as measured by breath testing (BT). METHODS A multicenter longitudinal study of CFTR-dependent disease profiling (NCT02477319) included a prospective evaluation for SIBO by BT. Tidal breath samples were collected after fasting and 15, 30, 45, 60, 90, and 120 minutes after ingestion of glucose, before and 1 month after subjects initiated luma + iva. RESULTS Forty-two subjects enrolled in the sub-study (mean age = 23.3 years; 51% female; 9.5% Latinx); 38 completed a hydrogen BT at both time points, of which 73.7% had a positive BT before luma/iva (baseline) and 65.8% had a positive test after luma/iva ( P = 0.44); shifts from negative to positive were also seen. Use of azithromycin (63.1%) and inhaled antibiotics (60.5%) were not associated with positive BT. Acid-blocking medications were taken by 73% of those with a negative BT at baseline and by 35% with a positive baseline BT ( P = 0.04). CONCLUSION We found a high rate of positive hydrogen breath tests in individuals with CF, confirming that SIBO is common. One month of luma/iva did not significantly change the proportion of those with positive breath hydrogen measurements.
Collapse
|
17
|
Roda J, Maia C, Almeida S, Oliveira RC, Ferreira R, Oliveira G. Faecal calprotectin and rectal histological inflammatory markers in cystic fibrosis: a single-centre study. BMJ Paediatr Open 2022; 6:e001422. [PMID: 36053631 PMCID: PMC9058793 DOI: 10.1136/bmjpo-2022-001422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/12/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE To analyse the association of faecal calprotectin with the genetic and clinical characteristics of paediatric patients with cystic fibrosis (PwCF). In a subset of these patients, we aimed to associate histological inflammatory features of rectal mucosa to faecal calprotectin levels. METHODS In a prospective study, faecal calprotectin levels were collected in all 23 PwCF attending our paediatric centre, together with demographic and clinical data. Associations between faecal calprotectin and clinical features were determined. In 11 of these patients, endoscopic rectal biopsies were obtained and the association between faecal calprotectin and histological inflammatory markers was analysed. Statistical analyses included Spearman's correlation coefficient, Mann-Whitney U test and Fisher's exact test. Sensitivity and specificity was calculated. RESULTS Median age of PwCF was 12 years, 19 had pancreatic insufficiency (PI) (19/23). Seventeen (17/23) had elevated faecal calprotectin, and the median value was 88 µg/g (IQR=178 µg/g). Higher faecal calprotectin levels were observed in the PI group (101 vs 30 µg/g, p=0.027). No significant correlation between elevated faecal calprotectin level and body mass index z-score was found. Five patients (22%) reported abdominal pain, three (13%) complained of diarrhoea and three (13%) had constipation, but these symptoms were not associated with elevated faecal calprotectin.Unspecific focal rectal inflammation was found in four patients (4/11). An association between rectal mucosa inflammation and elevated faecal calprotectin was found (p=0.015). Sensitivity was 100% and specificity was 86%. CONCLUSIONS In our PwCF, elevated faecal calprotectin was frequent, particularly if PI, and it was not related to gastrointestinal symptoms or malnutrition. Elevated faecal calprotectin was present in patients with histological evidence of rectal inflammation. Faecal calprotectin may be an indicator of asymptomatic rectal inflammation in PwCF.
Collapse
Affiliation(s)
- Juliana Roda
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitario de Coimbra EPE Hospital Pediátrico de Coimbra, Coimbra, Portugal
- Clínica Universitária de Pediatria, Universidade de Coimbra Faculdade de Medicina, Coimbra, Portugal
| | - Carla Maia
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitario de Coimbra EPE Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Susana Almeida
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitario de Coimbra EPE Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Rui Caetano Oliveira
- Pathology Department, Centro Hospitalar e Universitário de Coimbra EPE, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Universidade de Coimbra Faculdade de Medicina, Coimbra, Portugal
| | - Ricardo Ferreira
- Pediatric Gastroenterology and Nutrition Unit, Centro Hospitalar e Universitario de Coimbra EPE Hospital Pediátrico de Coimbra, Coimbra, Portugal
| | - Guiomar Oliveira
- Clínica Universitária de Pediatria, Universidade de Coimbra Faculdade de Medicina, Coimbra, Portugal
- Centro de Desenvolvimento da Criança e Centro de Investigação e Formação Clínica, Centro Hospitalar e Universitario de Coimbra EPE Hospital Pediátrico de Coimbra, Coimbra, Portugal
| |
Collapse
|
18
|
Walker NM, Liu J, Young SM, Woode RA, Clarke LL. Goblet cell hyperplasia is not epithelial-autonomous in the Cftr knockout intestine. Am J Physiol Gastrointest Liver Physiol 2022; 322:G282-G293. [PMID: 34878935 PMCID: PMC8793866 DOI: 10.1152/ajpgi.00290.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 02/03/2023]
Abstract
Goblet cell hyperplasia is an important manifestation of cystic fibrosis (CF) disease in epithelial-lined organs. Explants of CF airway epithelium show normalization of goblet cell numbers; therefore, we hypothesized that small intestinal enteroids from Cftr knockout (KO) mice would not exhibit goblet cell hyperplasia. Toll-like receptors 2 and 4 (Tlr2 and Tlr4) were investigated as markers of inflammation and influence on goblet cell differentiation. Ex vivo studies found goblet cell hyperplasia in Cftr KO jejunum compared with wild-type (WT) mice. IL-13, SAM pointed domain-containing ETS transcription factor (Spdef), Tlr2, and Tlr4 protein expression were increased in Cftr KO intestine relative to WT. In contrast, WT and Cftr KO enteroids did not exhibit differences in basal or IL-13-stimulated goblet cell numbers, or differences in expression of Tlr2, Tlr4, and Spdef. Ileal goblet cell numbers in Cftr KO/Tlr4 KO and Cftr KO/Tlr2 KO mice were not different from Cftr KO mice, but enumeration was confounded by altered mucosal morphology. Treatment with Tlr4 agonist LPS did not affect goblet cell numbers in WT or Cftr KO enteroids, whereas the Tlr2 agonist Pam3Csk4 stimulated goblet cell hyperplasia in both genotypes. Pam3Csk4 stimulation of goblet cell numbers was associated with suppression of Notch1 and Neurog3 expression and upregulated determinants of goblet cell differentiation. We conclude that goblet cell hyperplasia and inflammation of the Cftr KO small intestine are not exhibited by enteroids, indicating that this manifestation of CF intestinal disease is not epithelial-automatous but secondary to the altered CF intestinal environment.NEW & NOTEWORTHY Studies of small intestinal organoids from cystic fibrosis (CF) mice show that goblet cell hyperplasia and increased Toll-like receptor 2/4 expression are not primary manifestations of the CF intestine. Intestinal goblet cell hyperplasia in the CF mice was not strongly altered by genetic ablation of Tlr2 and Tlr 4, but could be induced in both wild-type and CF intestinal organoids by a Tlr2-dependent suppression of Notch signaling.
Collapse
Affiliation(s)
- Nancy M Walker
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Jinghua Liu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Sarah M Young
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Pathobiology, University of Missouri, Columbia, Missouri
| | - Rowena A Woode
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Lane L Clarke
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
Rodriguez DA, Ryan PM, Quigley EMM. Small Intestinal Bacterial Overgrowth. TEXTBOOK OF PEDIATRIC GASTROENTEROLOGY, HEPATOLOGY AND NUTRITION 2022:567-584. [DOI: 10.1007/978-3-030-80068-0_42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Intestinal function and transit associate with gut microbiota dysbiosis in cystic fibrosis. J Cyst Fibros 2021; 21:506-513. [PMID: 34895838 DOI: 10.1016/j.jcf.2021.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/04/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Most people with cystic fibrosis (pwCF) suffer from gastrointestinal symptoms and are at risk of gut complications. Gut microbiota dysbiosis is apparent within the CF population across all age groups, with evidence linking dysbiosis to intestinal inflammation and other markers of health. This pilot study aimed to investigate the potential relationships between the gut microbiota and gastrointestinal physiology, transit, and health. STUDY DESIGN Faecal samples from 10 pwCF and matched controls were subject to 16S rRNA sequencing. Results were combined with clinical metadata and MRI metrics of gut function to investigate relationships. RESULTS pwCF had significantly reduced microbiota diversity compared to controls. Microbiota compositions were significantly different, suggesting remodelling of core and rarer satellite taxa in CF. Dissimilarity between groups was driven by a variety of taxa, including Escherichia coli, Bacteroides spp., Clostridium spp., and Faecalibacterium prausnitzii. The core taxa were explained primarily by CF disease, whilst the satellite taxa were associated with pulmonary antibiotic usage, CF disease, and gut function metrics. Species-specific ordination biplots revealed relationships between taxa and the clinical or MRI-based variables observed. CONCLUSIONS Alterations in gut function and transit resultant of CF disease are associated with the gut microbiota composition, notably the satellite taxa. Delayed transit in the small intestine might allow for the expansion of satellite taxa resulting in potential downstream consequences for core community function in the colon.
Collapse
|
21
|
Mishra V, Bose A, Kiran S, Banerjee S, Shah IA, Chaukimath P, Reshi MM, Srinivas S, Barman A, Visweswariah SS. Gut-associated cGMP mediates colitis and dysbiosis in a mouse model of an activating mutation in GUCY2C. J Exp Med 2021; 218:212653. [PMID: 34546338 PMCID: PMC8480670 DOI: 10.1084/jem.20210479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
Activating mutations in receptor guanylyl cyclase C (GC-C), the target of gastrointestinal peptide hormones guanylin and uroguanylin, and bacterial heat-stable enterotoxins cause early-onset diarrhea and chronic inflammatory bowel disease (IBD). GC-C regulates ion and fluid secretion in the gut via cGMP production and activation of cGMP-dependent protein kinase II. We characterize a novel mouse model harboring an activating mutation in Gucy2c equivalent to that seen in an affected Norwegian family. Mutant mice demonstrated elevated intestinal cGMP levels and enhanced fecal water and sodium content. Basal and linaclotide-mediated small intestinal transit was higher in mutant mice, and they were more susceptible to DSS-induced colitis. Fecal microbiome and gene expression analyses of colonic tissue revealed dysbiosis, up-regulation of IFN-stimulated genes, and misregulation of genes associated with human IBD and animal models of colitis. This novel mouse model thus provides molecular insights into the multiple roles of intestinal epithelial cell cGMP, which culminate in dysbiosis and the induction of inflammation in the gut.
Collapse
Affiliation(s)
- Vishwas Mishra
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Avipsa Bose
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Shashi Kiran
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Sanghita Banerjee
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Idrees A Shah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Pooja Chaukimath
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Mudasir M Reshi
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Swarna Srinivas
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Anaxee Barman
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
22
|
Henen S, Denton C, Teckman J, Borowitz D, Patel D. Review of Gastrointestinal Motility in Cystic Fibrosis. J Cyst Fibros 2021; 20:578-585. [PMID: 34147362 DOI: 10.1016/j.jcf.2021.05.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Gastrointestinal manifestations in patients with cystic fibrosis (CF) are extremely common and have recently become a research focus. Gastrointestinal (GI) dysfunction is poorly understood in the CF population, despite many speculations including the role of luminal pH, bacterial overgrowth, and abnormal microbiome. Nevertheless, dysmotility is emerging as a possible key player in CF intestinal symptoms. Our review article aims to explore the sequelae of defective cystic fibrosis transmembrane conductance regulator (CFTR) genes on the GI tract as studied in both animals and humans, describe various presentations of intestinal dysmotility in CF, review newer diagnostic motility techniques including intraluminal manometry, and review the current literature regarding the potential role of dysmotility in CF-related intestinal pathologies.
Collapse
Affiliation(s)
- Sara Henen
- Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104.
| | - Christine Denton
- Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104
| | - Jeff Teckman
- Interim Chair, Department of Pediatrics, Professor of Pediatrics and Biochemistry, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand BLVD, St. Louis, MO 63104.
| | - Drucy Borowitz
- Emeritus Professor of Clinical Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, 1001 Main Street, Buffalo, NY, 14203.
| | - Dhiren Patel
- Associate Professor and Medical Director, Neurogastroenterology and Motility Program, Department of Pediatrics, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104.
| |
Collapse
|
23
|
Impaired Intestinal Sodium Transport in Inflammatory Bowel Disease: From the Passenger to the Driver's Seat. Cell Mol Gastroenterol Hepatol 2021; 12:277-292. [PMID: 33744482 PMCID: PMC8165433 DOI: 10.1016/j.jcmgh.2021.03.005] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/22/2022]
Abstract
Although impaired intestinal sodium transport has been described for decades as a ubiquitous feature of inflammatory bowel disease (IBD), whether and how it plays a pivotal role in the ailment has remained uncertain. Our identification of dominant mutations in receptor guanylyl cyclase 2C as a cause of IBD-associated familial diarrhea syndrome brought a shift in the way we envision impaired sodium transport. Is this just a passive collateral effect resulting from intestinal inflammation, or is it a crucial regulator of IBD pathogenesis? This review summarizes the mutational spectrum and underlying mechanisms of monogenic IBD associated with congenital sodium diarrhea. We constructed a model proposing that impaired sodium transport is an upstream pathogenic factor in IBD. The review also synthesized emerging insights from microbiome and animal studies to suggest how sodium malabsorption can serve as a unifying mediator of downstream pathophysiology. Further investigations into the mechanisms underlying salt and water transport in the intestine will provide newer approaches for understanding the ion-microbiome-immune cross-talk that serves as a driver of IBD. Model systems, such as patient-derived enteroids or induced pluripotent stem cell models, are warranted to unravel the role of individual genes regulating sodium transport and to develop more effective epithelial rescue and repair therapies.
Collapse
|
24
|
Abstract
Generating the barriers that protect our inner surfaces from bacteria and other challenges requires large glycoproteins called mucins. These come in two types, gel-forming and transmembrane, all characterized by large, highly O-glycosylated mucin domains that are diversely decorated by Golgi glycosyltransferases to become extended rodlike structures. The general functions of mucins on internal epithelial surfaces are to wash away microorganisms and, even more importantly, to build protective barriers. The latter function is most evident in the large intestine, where the inner mucus layer separates the numerous commensal bacteria from the epithelial cells. The host's conversion of MUC2 to the outer mucus layer allows bacteria to degrade the mucin glycans and recover the energy content that is then shared with the host. The molecular nature of the mucins is complex, and how they construct the extracellular complex glycocalyx and mucus is poorly understood and a future biochemical challenge.
Collapse
Affiliation(s)
- Gunnar C Hansson
- Department of Medical Biochemistry, University of Gothenburg, SE 405 30 Gothenburg, Sweden;
| |
Collapse
|
25
|
Yeh KM, Johansson O, Le H, Rao K, Markus I, Perera DS, Lubowski DZ, King DW, Zhang L, Chen H, Liu L. Cystic fibrosis transmembrane conductance regulator modulates enteric cholinergic activities and is abnormally expressed in the enteric ganglia of patients with slow transit constipation. J Gastroenterol 2019; 54:994-1006. [PMID: 31392489 DOI: 10.1007/s00535-019-01610-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/31/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cystic fibrosis transmembrane conductance regulator (CFTR) was recently found in the enteric nervous system, where its role is unclear. We aimed to identify which enteric neuronal structures express CFTR, whether CFTR modulates enteric neurotransmission and if altered CFTR expression is associated with slow transit constipation (STC). METHODS Immunofluorescence double labeling was performed to localize CFTR with various neuronal and glial cell markers in the human colon. The immunoreactivity (IR) of CFTR and choline acetyltransferase (ChAT) on myenteric plexus of control and STC colon was quantitatively analyzed. In control colonic muscle strips, electrical field stimulation (EFS) evoked contractile responses and the release of acetylcholine (ACh) was measured in the presence of the CFTR channel inhibitor, CFTR(inh)-172. RESULTS CFTR-IR was densely localized to myenteric ganglia, where it was co-localized with neuronal markers HuC/D and β-tubulin, and glial marker S-100 but little with glial fibrillary acidic protein. Vesicular ACh transport was almost exclusively co-localized with CFTR, but neurons expressing nitric oxide synthase were CFTR negative. Significant reductions of CFTR-IR (P < 0.01) and ChAT-IR (P < 0.05) were observed on myenteric ganglia of STC compared to control. Pre-treatment of colonic muscle strips with CFTR(inh)-172 (10 µM) significantly inhibited EFS-evoked contractile responses (P < 0.01) and ACh release (P < 0.05). CONCLUSIONS Co-localization of CFTR-IR with cholinergic markers, inhibition of EFS-induced colonic muscle contractility and ACh release by CFTR(inh)-172 suggest that CFTR modulates enteric cholinergic neurotransmission. The downregulation of CFTR and ChAT in myenteric ganglia of STC correlated with the impaired contractile responses to EFS.
Collapse
Affiliation(s)
- Ka Ming Yeh
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Olle Johansson
- Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Huy Le
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Karan Rao
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Irit Markus
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | | | | | | | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Hongzhuan Chen
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Liu
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
26
|
Altman K, McDonald CM, Michel SH, Maguiness K. Nutrition in cystic fibrosis: From the past to the present and into the future. Pediatr Pulmonol 2019; 54 Suppl 3:S56-S73. [PMID: 31715089 DOI: 10.1002/ppul.24521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Abstract
Nutritional management is an integral part of multidisciplinary care for persons with cystic fibrosis. This review will look at how nutrition care has evolved over time. In addition, we will look at how some newer therapies impact nutrition care.
Collapse
Affiliation(s)
- Kimberly Altman
- Gunnar Esiason Adult Cystic Fibrosis and Lung Center, New York Presbyterian/Columbia University Medical Center, New York, New York, United States
| | | | - Suzanne H Michel
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Karen Maguiness
- Section of Pediatric Pulmonology, Allergy, and Sleep Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, United States
| |
Collapse
|
27
|
Avelar Rodriguez D, Ryan PM, Toro Monjaraz EM, Ramirez Mayans JA, Quigley EM. Small Intestinal Bacterial Overgrowth in Children: A State-Of-The-Art Review. Front Pediatr 2019; 7:363. [PMID: 31552207 PMCID: PMC6737284 DOI: 10.3389/fped.2019.00363] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is a heterogenous and poorly understood entity characterised by an excessive growth of select microorganisms within the small intestine. This excessive bacterial biomass, in turn, disrupts host physiology in a myriad of ways, leading to gastrointestinal and non-gastrointestinal symptoms and complications. SIBO is a common cause of non-specific gastrointestinal symptoms in children, such as chronic abdominal pain, abdominal distention, diarrhoea, and flatulence, amongst others. In addition, it has recently been implicated in the pathophysiology of stunting, a disease that affects millions of children worldwide. Risk factors such as acid-suppressive therapies, alterations in gastrointestinal motility and anatomy, as well as impoverished conditions, have been shown to predispose children to SIBO. SIBO can be diagnosed via culture-dependant or culture-independent approaches. SIBO's epidemiology is limited due to the lack of uniformity and consensus of its diagnostic criteria, as well as the paucity of literature available. Antibiotics remain the first-line treatment option for SIBO, although emerging modalities such as probiotics and diet manipulation could also have a role. Herein, we present a state-of-the-art-review which aims to comprehensively outline the most current information on SIBO in children, with particular emphasis on the gut microbiota.
Collapse
Affiliation(s)
- David Avelar Rodriguez
- Pediatric Gastroenterology and Nutrition Unit, National Institute of Pediatrics, Mexico City, Mexico
| | | | | | | | - Eamonn Martin Quigley
- Lynda K. and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
28
|
Enaud R, Hooks KB, Barre A, Barnetche T, Hubert C, Massot M, Bazin T, Clouzeau H, Bui S, Fayon M, Berger P, Lehours P, Bébéar C, Nikolski M, Lamireau T, Delhaes L, Schaeverbeke T. Intestinal Inflammation in Children with Cystic Fibrosis Is Associated with Crohn's-Like Microbiota Disturbances. J Clin Med 2019; 8:jcm8050645. [PMID: 31083321 PMCID: PMC6572243 DOI: 10.3390/jcm8050645] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is a systemic genetic disease that leads to pulmonary and digestive disorders. In the majority of CF patients, the intestine is the site of chronic inflammation and microbiota disturbances. The link between gut inflammation and microbiota dysbiosis is still poorly understood. The main objective of this study was to assess gut microbiota composition in CF children depending on their intestinal inflammation. We collected fecal samples from 20 children with CF. Fecal calprotectin levels were measured and fecal microbiota was analyzed by 16S rRNA sequencing. We observed intestinal inflammation was associated with microbiota disturbances characterized mainly by increased abundances of Staphylococcus, Streptococcus, and Veillonella dispar, along with decreased abundances of Bacteroides, Bifidobacterium adolescentis, and Faecalibacterium prausnitzii. Those changes exhibited similarities with that of Crohn's disease (CD), as evidenced by the elevated CD Microbial-Dysbiosis index that we applied for the first time in CF. Furthermore, the significant over-representation of Streptococcus in children with intestinal inflammation appears to be specific to CF and raises the issue of gut-lung axis involvement. Taken together, our results provide new arguments to link gut microbiota and intestinal inflammation in CF and suggest the key role of the gut-lung axis in the CF evolution.
Collapse
Affiliation(s)
- Raphaël Enaud
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Katarzyna B Hooks
- Bordeaux Bioinformatics Center, University Bordeaux, F-33000 Bordeaux, France.
- Laboratoire Bordelais de Recherche en Informatique, CNRS, University Bordeaux, UMR 5800, F-33400 Talence, France.
| | - Aurélien Barre
- Bordeaux Bioinformatics Center, University Bordeaux, F-33000 Bordeaux, France.
- Laboratoire Bordelais de Recherche en Informatique, CNRS, University Bordeaux, UMR 5800, F-33400 Talence, France.
| | - Thomas Barnetche
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
- Service de Rhumatologie, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Christophe Hubert
- INSERM, MRGM, University Bordeaux, U1211, F-33000 Bordeaux, France.
- PGTB, University Bordeaux, F-33000 Bordeaux, France.
| | - Marie Massot
- BIOGECO, INRA, University Bordeaux, F-33610 Cestas, France.
| | - Thomas Bazin
- INRA-Bordeaux Aquitaine Centre, University Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Haude Clouzeau
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
| | - Stéphanie Bui
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
| | - Michael Fayon
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Philippe Lehours
- BaRITOn, INSERM, University Bordeaux, UMR1053, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Cécile Bébéar
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
- INRA-Bordeaux Aquitaine Centre, University Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Macha Nikolski
- Bordeaux Bioinformatics Center, University Bordeaux, F-33000 Bordeaux, France.
- Laboratoire Bordelais de Recherche en Informatique, CNRS, University Bordeaux, UMR 5800, F-33400 Talence, France.
| | - Thierry Lamireau
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Laurence Delhaes
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Thierry Schaeverbeke
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
- Service de Rhumatologie, CHU Bordeaux, F-33000 Bordeaux, France.
- INRA-Bordeaux Aquitaine Centre, University Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, CHU Bordeaux, F-33000 Bordeaux, France.
| |
Collapse
|
29
|
Hwang SJ, Pardo DM, Zheng H, Bayguinov Y, Blair PJ, Fortune‐Grant R, Cook RS, Hennig GW, Shonnard MC, Grainger N, Peri LE, Verma SD, Rock J, Sanders KM, Ward SM. Differential sensitivity of gastric and small intestinal muscles to inducible knockdown of anoctamin 1 and the effects on gastrointestinal motility. J Physiol 2019; 597:2337-2360. [PMID: 30843201 PMCID: PMC6487927 DOI: 10.1113/jp277335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/22/2019] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Electrical pacemaking in gastrointestinal muscles is generated by specialized interstitial cells of Cajal that produce the patterns of contractions required for peristalsis and segmentation in the gut. The calcium-activated chloride conductance anoctamin-1 (Ano1) has been shown to be responsible for the generation of pacemaker activity in GI muscles, but this conclusion is established from studies of juvenile animals in which effects of reduced Ano1 on gastric emptying and motor patterns could not be evaluated. Knocking down Ano1 expression using Cre/LoxP technology caused dramatic changes in in gastric motor activity, with disrupted slow waves, abnormal phasic contractions and delayed gastric emptying; modest changes were noted in the small intestine. Comparison of the effects of Ano1 antagonists on muscles from juvenile and adult small intestinal muscles suggests that conductances in addition to Ano1 may develop with age and contribute to pacemaker activity. ABSTRACT Interstitial cells of Cajal (ICC) generate slow waves and transduce neurotransmitter signals in the gastrointestinal (GI) tract, facilitating normal motility patterns. ICC express a Ca2+ -activated Cl- conductance (CaCC), and constitutive knockout of the channel protein anoctamin-1 leads to loss of slow waves in gastric and intestinal muscles. These knockout experiments were performed on juvenile mice. However, additional experiments demonstrated significant differences in the sensitivity of gastric and intestinal muscles to antagonists of anoctamin-1 channels. Furthermore, the significance of anoctamin-1 and the electrical and mechanical behaviours facilitated by this conductance have not been evaluated on the motor behaviours of adult animals. Cre/loxP technology was used to generate cell-specific knockdowns of anoctamin-1 in ICC (KitCreERT2/+ ;Ano1tm2jrr/+ ) in GI muscles. The recombination efficiency of KitCreERT was evaluated with an eGFP reporter, molecular techniques and immunohistochemistry. Electrical and contractile experiments were used to examine the consequences of anoctamin-1 knockdown on pacemaker activity, mechanical responses, gastric motility patterns, gastric emptying and GI transit. Reduced anoctamin-1 caused loss of gastric, but not intestinal slow waves. Irregular spike complexes developed in gastric muscles, leading to uncoordinated antral contractions, delayed gastric emptying and increased total GI transit time. Slow waves in intestinal muscles of juvenile mice were more sensitive to anoctamin-1 antagonists than slow waves in adult muscles. The low susceptibility to anoctamin-1 knockdown and weak efficacy of anoctamin-1 antagonists in inhibiting slow waves in adult small intestinal muscles suggest that a conductance in addition to anoctamin-1 may develop in small intestinal ICC with ageing and contribute to pacemaker activity.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - David M. Pardo
- Department of AnatomyUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Haifeng Zheng
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Yulia Bayguinov
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Peter J. Blair
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Rachael Fortune‐Grant
- Faculty of BiologyMedicine and HealthSchool of Biological SciencesUniversity of ManchesterUK
| | - Robert S. Cook
- School of Molecular and Cellular BiologyUniversity of LeedsLeedsLS2 9JTUK
| | - Grant W. Hennig
- Department of PharmacologyThe University of VermontUVM College of MedicineBurlingtonVT05405USA
| | - Matthew C. Shonnard
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Nathan Grainger
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Lauren E. Peri
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Sonali Deep Verma
- Department of AnatomyUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Jason Rock
- Centre for Regenerative MedicineBoston University School of MedicineBostonMA02118USA
| | - Kenton M. Sanders
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Sean M. Ward
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| |
Collapse
|
30
|
McHugh DR, Cotton CU, Moss FJ, Vitko M, Valerio DM, Kelley TJ, Hao S, Jafri A, Drumm ML, Boron WF, Stern RC, McBennett K, Hodges CA. Linaclotide improves gastrointestinal transit in cystic fibrosis mice by inhibiting sodium/hydrogen exchanger 3. Am J Physiol Gastrointest Liver Physiol 2018; 315:G868-G878. [PMID: 30118317 PMCID: PMC9925117 DOI: 10.1152/ajpgi.00261.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastrointestinal dysfunction in cystic fibrosis (CF) is a prominent source of pain among patients with CF. Linaclotide, a guanylate cyclase C (GCC) receptor agonist, is a US Food and Drug Administration-approved drug prescribed for chronic constipation but has not been widely used in CF, as the cystic fibrosis transmembrane conductance regulator (CFTR) is the main mechanism of action. However, anecdotal clinical evidence suggests that linaclotide may be effective for treating some gastrointestinal symptoms in CF. The goal of this study was to determine the effectiveness and mechanism of linaclotide in treating CF gastrointestinal disorders using CF mouse models. Intestinal transit, chloride secretion, and intestinal lumen fluidity were assessed in wild-type and CF mouse models in response to linaclotide. CFTR and sodium/hydrogen exchanger 3 (NHE3) response to linaclotide was also evaluated. Linaclotide treatment improved intestinal transit in mice carrying either F508del or null Cftr mutations but did not induce detectable Cl- secretion. Linaclotide increased fluid retention and fluidity of CF intestinal contents, suggesting inhibition of fluid absorption. Targeted inhibition of sodium absorption by the NHE3 inhibitor tenapanor produced improvements in gastrointestinal transit similar to those produced by linaclotide treatment, suggesting that inhibition of fluid absorption by linaclotide contributes to improved gastrointestinal transit in CF. Our results demonstrate that linaclotide improves gastrointestinal transit in CF mouse models by increasing luminal fluidity through inhibiting NHE3-mediated sodium absorption. Further studies are necessary to assess whether linaclotide could improve CF intestinal pathologies in patients. GCC signaling and NHE3 inhibition may be therapeutic targets for CF intestinal manifestations. NEW & NOTEWORTHY Linaclotide's primary mechanism of action in alleviating chronic constipation is through cystic fibrosis transmembrane conductance regulator (CFTR), negating its use in patients with cystic fibrosis (CF). For the first time, our findings suggest that in the absence of CFTR, linaclotide can improve fluidity of the intestinal lumen through the inhibition of sodium/hydrogen exchanger 3. These findings suggest that linaclotide could improve CF intestinal pathologies in patients.
Collapse
Affiliation(s)
- Daniel R. McHugh
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Calvin U. Cotton
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fraser J. Moss
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Megan Vitko
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Dana M. Valerio
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Thomas J. Kelley
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,4Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Shuyu Hao
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Anjum Jafri
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mitchell L. Drumm
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Walter F. Boron
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio,5Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio,6Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Robert C. Stern
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,7Rainbow Babies and Children’s Hospital, Cleveland, Ohio
| | - Kimberly McBennett
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,7Rainbow Babies and Children’s Hospital, Cleveland, Ohio
| | - Craig A. Hodges
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
31
|
Philp AR, Riquelme TT, Millar-Büchner P, González R, Sepúlveda FV, Cid LP, Flores CA. Kcnn4 is a modifier gene of intestinal cystic fibrosis preventing lethality in the Cftr-F508del mouse. Sci Rep 2018; 8:9320. [PMID: 29915289 PMCID: PMC6006244 DOI: 10.1038/s41598-018-27465-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/04/2018] [Indexed: 02/07/2023] Open
Abstract
Nearly 70% of cystic fibrosis (CF) patients bear the phenylalanine-508 deletion but disease severity differs greatly, and is not explained by the existence of different mutations in compound heterozygous. Studies demonstrated that genes other than CFTR relate to intestinal disease in humans and CF-mouse. Kcnn4, the gene encoding the calcium-activated potassium channel KCa3.1, important for intestinal secretion, is present in a locus linked with occurrence of intestinal CF-disease in mice and humans. We reasoned that it might be a CF-modifier gene and bred a CF-mouse with Kcnn4 silencing, finding that lethality was almost abolished. Silencing of Kcnn4 did not improve intestinal secretory functions, but rather corrected increased circulating TNF-α level and reduced intestinal mast cell increase. Given the importance of mast cells in intestinal disease additional double mutant CF-animals were tested, one lacking mast cells (C-kitW-sh/W-sh) and Stat6-/- to block IgE production. While mast cell depletion had no effect, silencing Stat6 significantly reduced lethality. Our results show that Kcnn4 is an intestinal CF modifier gene partially acting through a STAT6-dependent mechanism.
Collapse
Affiliation(s)
- Amber R Philp
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Texia T Riquelme
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
| | - Pamela Millar-Büchner
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Rodrigo González
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
| | | | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
| | - Carlos A Flores
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile.
| |
Collapse
|
32
|
Benezech A, Desmazes-Dufeu N, Baumstarck K, Bouvier M, Coltey B, Reynaud-Gaubert M, Vitton V. Prevalence of Fecal Incontinence in Adults with Cystic Fibrosis. Dig Dis Sci 2018; 63:982-988. [PMID: 29086331 DOI: 10.1007/s10620-017-4825-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 10/23/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Patients with cystic fibrosis (CF) are deemed at risk of developing urinary incontinence (UI) due to repeated coughing and other factors causing increased pressure on the pelvic floor. Fecal incontinence (FI) is probably derived from the same mechanism, but only very few data are available on its frequency. AIMS The aim of this study was to determine the prevalence of FI in an adult population with CF. METHODS This retrospective study was conducted from January 2012 to June 2014. Patients were recruited from Marseille referral center for adult CF. They were asked to fill in a self-completed anonymous questionnaire for symptom assessment of UI and FI. Clinical data and a detailed history of CF were also recorded. RESULTS A total of 155 out of 190 patients (92 females) of mean age 30.5 ± 11 years completed the survey. Seventy-three patients (47%) were lung transplanted. Forty patients (25.8%) reported FI with a mean St Mark's score of 4.9 ± 2. Thirty-five patients (22.6%) reported UI. Eighteen patients (11.6%) reported both FI and UI. FI was significantly more frequent in older patients (34.27 vs. 29.54 years, p = 0.03) and in patients with associated UI (p = 0.001). No relationship was found between respiratory, bacterial, nutritional status, transplantation, pancreatic status, practice of physiotherapy, delivery history, and FI. CONCLUSIONS The high prevalence of FI in CF and its negative impacts need to integrate this symptom in the overall treatment of this pathology. The systematic early detection of FI may allow its rapid management and limit their consequences.
Collapse
Affiliation(s)
- Alban Benezech
- Plateforme d'Interface Clinique, Aix Marseille Université, CNRS, CRN2M-UMR7286, 13344, Marseille Cedex 20, France. .,APHM, Hôpital NORD, Service de Gastroentérologie, CHU Nord, Chemin des Bourrely, 13915, Marseille Cedex 20, France.
| | - Nadine Desmazes-Dufeu
- APHM, Hôpital NORD, Service de Pneumologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM) Adulte, 13915, Marseille Cedex 20, France
| | - Karine Baumstarck
- Unité d'aide méthodologique à la Recherche Clinique, Laboratoire de Santé Publique, Aix-Marseille Université, Marseille Cedex 20, France
| | - Michel Bouvier
- Plateforme d'Interface Clinique, Aix Marseille Université, CNRS, CRN2M-UMR7286, 13344, Marseille Cedex 20, France.,APHM, Hôpital NORD, Service de Gastroentérologie, CHU Nord, Chemin des Bourrely, 13915, Marseille Cedex 20, France
| | - Bérengère Coltey
- APHM, Hôpital NORD, Service de Pneumologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM) Adulte, 13915, Marseille Cedex 20, France
| | - Martine Reynaud-Gaubert
- APHM, Hôpital NORD, Service de Pneumologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM) Adulte, 13915, Marseille Cedex 20, France.,URMITE - CNRS - IRD UMR 6236, Faculté de Médecine de Marseille, Aix Marseille Université, Marseille Cedex 20, France
| | - Véronique Vitton
- Plateforme d'Interface Clinique, Aix Marseille Université, CNRS, CRN2M-UMR7286, 13344, Marseille Cedex 20, France.,APHM, Hôpital NORD, Service de Gastroentérologie, CHU Nord, Chemin des Bourrely, 13915, Marseille Cedex 20, France
| |
Collapse
|
33
|
Amarachintha S, Harmel-Laws E, Steinbrecher KA. Guanylate cyclase C reduces invasion of intestinal epithelial cells by bacterial pathogens. Sci Rep 2018; 8:1521. [PMID: 29367634 PMCID: PMC5784150 DOI: 10.1038/s41598-018-19868-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/20/2017] [Indexed: 12/24/2022] Open
Abstract
The guanylate cyclase C (GC-C) receptor regulates electrolyte and water secretion into the gut following activation by the E. coli enterotoxin STa, or by weaker endogenous agonists guanylin and uroguanylin. Our previous work has demonstrated that GC-C plays an important role in controlling initial infection as well as carrying load of non-invasive bacterial pathogens in the gut. Here, we use Salmonella enterica serovar Typhimurium to determine whether GC-C signaling is important in host defense against pathogens that actively invade enterocytes. In vitro studies indicated that GC-C signaling significantly reduces Salmonella invasion into Caco2-BBE monolayers. Relative to controls, GC-C knockout mice develop severe systemic illness following oral Salmonella infection, characterized by disrupted intestinal mucus layer, elevated cytokines and organ CFUs, and reduced animal survival. In Salmonella-infected wildtype mice, oral gavage of GC-C agonist peptide reduced host/pathogen physical interaction and diminished bacterial translocation to mesenteric lymph nodes. These studies suggest that early life susceptibility to STa-secreting enterotoxigenic E. coli may be counter-balanced by a critical role of GC-C in protecting the mucosa from non-STa producing, invasive bacterial pathogens.
Collapse
Affiliation(s)
- Surya Amarachintha
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Eleana Harmel-Laws
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Kris A Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229, USA.
| |
Collapse
|
34
|
Alginate Oligomers and Their Use as Active Pharmaceutical Drugs. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2018. [DOI: 10.1007/978-981-10-6910-9_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
35
|
Burke DG, Fouhy F, Harrison MJ, Rea MC, Cotter PD, O'Sullivan O, Stanton C, Hill C, Shanahan F, Plant BJ, Ross RP. The altered gut microbiota in adults with cystic fibrosis. BMC Microbiol 2017; 17:58. [PMID: 28279152 PMCID: PMC5345154 DOI: 10.1186/s12866-017-0968-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 03/03/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cystic Fibrosis (CF) is an autosomal recessive disease that affects the function of a number of organs, principally the lungs, but also the gastrointestinal tract. The manifestations of cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction in the gastrointestinal tract, as well as frequent antibiotic exposure, undoubtedly disrupts the gut microbiota. To analyse the effects of CF and its management on the microbiome, we compared the gut microbiota of 43 individuals with CF during a period of stability, to that of 69 non-CF controls using 454-pyrosequencing of the 16S rRNA gene. The impact of clinical parameters, including antibiotic therapy, on the results was also assessed. RESULTS The CF-associated microbiome had reduced microbial diversity, an increase in Firmicutes and a reduction in Bacteroidetes compared to the non-CF controls. While the greatest number of differences in taxonomic abundances of the intestinal microbiota was observed between individuals with CF and the healthy controls, gut microbiota differences were also reported between people with CF when grouped by clinical parameters including % predicted FEV1 (measure of lung dysfunction) and the number of intravenous (IV) antibiotic courses in the previous 12 months. Notably, CF individuals presenting with severe lung dysfunction (% predicted FEV1 ≤ 40%) had significantly (p < 0.05) reduced gut microbiota diversity relative to those presenting with mild or moderate dysfunction. A significant negative correlation (-0.383, Simpson's Diversity Index) was also observed between the number of IV antibiotic courses and gut microbiota diversity. CONCLUSIONS This is one of the largest single-centre studies on gut microbiota in stable adults with CF and demonstrates the significantly altered gut microbiota, including reduced microbial diversity seen in CF patients compared to healthy controls. The data show the impact that CF and it's management have on gut microbiota, presenting the opportunity to develop CF specific probiotics to minimise microbiota alterations.
Collapse
Affiliation(s)
- D G Burke
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - F Fouhy
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland
| | - M J Harrison
- HRB Clinical Research Facility, University College Cork, Cork, Ireland.,Cork Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland
| | - M C Rea
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - P D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - O O'Sullivan
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - C Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - C Hill
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - F Shanahan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - B J Plant
- APC Microbiome Institute, University College Cork, Cork, Ireland.,HRB Clinical Research Facility, University College Cork, Cork, Ireland.,Cork Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland.,Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - R P Ross
- APC Microbiome Institute, University College Cork, Cork, Ireland. .,College of Science, Engineering and Food Science (SEFS), University College Cork, Cork, Ireland.
| |
Collapse
|
36
|
Garg M, Ooi CY. The Enigmatic Gut in Cystic Fibrosis: Linking Inflammation, Dysbiosis, and the Increased Risk of Malignancy. Curr Gastroenterol Rep 2017; 19:6. [PMID: 28155088 DOI: 10.1007/s11894-017-0546-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Intestinal inflammation, dysbiosis, and increased gastrointestinal malignancy risks are well-described in patients with cystic fibrosis (CF). However, there is limited understanding of their pathophysiology. This review aims to discuss these issues and assess potential links between them. RECENT FINDINGS Evidence of links between intestinal inflammation and dysbiosis (an imbalance in intestinal microbial populations) exist. Recent studies have demonstrated reduction in intestinal inflammation with probiotic administration. Both bacterial dysbiosis and gut inflammation contribute to the suboptimal nutritional status seen in children with CF. Short-chain fatty acids may be reduced in the gut lumen as a result of bacterial imbalances and may promote inflammation. Inflammation and bacterial dysbiosis in CF may also contribute to emerging adult complications such as gastrointestinal malignancy. An increase in carcinogenic microbes and reduction in microbes protective against cancer have been found in CF, linking bacterial dysbiosis and cancer. Murine studies suggest the CF gene, cystic fibrosis transmembrane conductance regulator (CFTR) gene, itself may be a tumour suppressor gene. The pathophysiology of interactions among intestinal inflammation, dysbiosis, and malignancy in CF is not clearly understood and requires further research.
Collapse
Affiliation(s)
- Millie Garg
- School of Women's and Children's Health, Medicine, University of New South Wales, Randwick, NSW, 2031, Australia
| | - Chee Y Ooi
- School of Women's and Children's Health, Medicine, University of New South Wales, Randwick, NSW, 2031, Australia.
- Department of Paediatric Gastroenterology, Sydney Children's Hospital, Randwick, NSW, 2031, Australia.
| |
Collapse
|
37
|
Long ZZ, Nie QH. Paneth cells, antimicrobial peptides, and intestinal mucosal immunity. Shijie Huaren Xiaohua Zazhi 2017; 25:209-219. [DOI: 10.11569/wcjd.v25.i3.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Paneth cells (PCs) and their antimicrobial peptides (AMPs) are closely related to the ecological unbalance of gut microbiota, intestinal inflammation, and systemic infections. The dysfunction of intestinal mucosal barrier and innate immunity is always accompanied with changes in the levels of AMPs, for example, alpha-defensins and other inflammatory mediators produced by PCs. Studies show that PCs can alter their functional status and release effector molecules under some conditions such as cholinergic agonists, microorganisms and their antigens, enteral nutrition, and genetic susceptibility. Therefore, these conditions can induce inflammatory bowel disease, bacterial translocation, overgrowth of gut microbe, and damage to immune tolerance. The research on PCs can provide new targets and strategies for clinical treatment of the relevant diseases. In this paper, we discuss the relationship of PCs and their AMPs with intestinal mucosal barrier and innate immunity.
Collapse
|
38
|
Vitko M, Valerio DM, Rye PD, Onsøyen E, Myrset AH, Dessen A, Drumm ML, Hodges CA. A novel guluronate oligomer improves intestinal transit and survival in cystic fibrosis mice. J Cyst Fibros 2016; 15:745-751. [DOI: 10.1016/j.jcf.2016.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 12/22/2022]
|
39
|
Rogers GB, Narkewicz MR, Hoffman LR. The CF gastrointestinal microbiome: Structure and clinical impact. Pediatr Pulmonol 2016; 51:S35-S44. [PMID: 27662102 PMCID: PMC5303757 DOI: 10.1002/ppul.23544] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 02/06/2023]
Abstract
The gastrointestinal (GI) microbiome is shaped by host diet, immunity, and other physicochemical characteristics of the GI tract, and perturbations such as antibiotic treatments can lead to persistent changes in microbial constituency and function. These GI microbes also play critical roles in host nutrition and health. A growing body of evidence suggests that the GI microbiome in people with CF is altered, and that these dysbioses contribute to disease manifestations in many organs, both within and beyond the GI tract. Therapies that people with CF receive, even those targeting the respiratory tract, may impact the CF GI microbiome in ways that can influence the outcome of treatment. These new perspectives on the microbial contents of the CF intestine offer new opportunities for preventing a variety of CF-associated disorders. Pediatr Pulmonol. 2016;51:S35-S44. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Geraint B Rogers
- SAHMRI Infection and Immunity Theme, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Michael R Narkewicz
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado.,Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado
| | - Lucas R Hoffman
- Departments of Pediatrics and Microbiology, University of Washington, Seattle, Washington. .,Seattle Children's Hospital, Seattle, Washington.
| |
Collapse
|
40
|
Decreased Expression of Enterocyte Nutrient Assimilation Genes and Proteins in the Small Intestine of Cystic Fibrosis Mouse. J Pediatr Gastroenterol Nutr 2016; 62:627-34. [PMID: 26551319 DOI: 10.1097/mpg.0000000000001030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Cystic fibrosis (CF) has major effects on the intestinal tract with potential consequences on nutrition, but these are not fully understood. I investigated the possibility of altered enterocyte maturation in CF, as suggested by decreased enterocyte nutrient assimilation gene expression in published transcriptome analysis of the small intestine of CF mouse. METHODS In CF and wild-type (WT) mice, enterocyte gene/protein expression was analyzed by quantitative realtime polymerase chain reaction (qRT-PCR), enzyme histochemistry, immunohistochemistry, and Western blot. One group of mice was maintained on a control liquid diet; to manipulate the gut microbiota, a second group was treated with oral antibiotics; to improve hydration of the gut lumen, a third group was given a laxative drinking solution. RESULTS On the control diet in the CF intestine, there were decreased levels (67%-85% reduction of WT levels) of enterocyte genes/proteins. Antibiotics did not normalize the expression of enterocyte markers in the CF mouse. In contrast, the laxative treatment of CF mice significantly increased expression to near WT levels. CONCLUSIONS These studies suggest that the environment of the CF intestinal lumen plays a role in reduced maturation of enterocytes. Because changing the gut lumen environment can affect enterocyte maturation, this is not a cell-autonomous effect of loss of CF transmembrane conductance regulator.
Collapse
|
41
|
Abstract
Cystic fibrosis is a life-limiting, recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Increased survival outcomes and the multisystem nature of the disease, including the involvement of hepatobiliary and gastrointestinal tracts, now require the need for more extensive knowledge and expertise in cystic fibrosis among gastroenterologists. Manifestations are either a direct consequence of the primary defect in cystic fibrosis or a secondary complication of the disease or therapy. Adult patients with cystic fibrosis also have an increased risk of malignancy in the gastrointestinal and pancreatico-biliary tracts compared with the general population. Novel treatments that target the basic defects in the CFTR protein have emerged, but to date not much is known about their effects on the gastrointestinal and hepatobiliary systems. The introduction of such therapies has provided new opportunities for the application of intestinal endpoints in clinical trials and the understanding of underlying disease mechanisms that affect the gut in cystic fibrosis.
Collapse
Affiliation(s)
- Chee Y Ooi
- Sydney Children's Hospital, School of Women's and Children's Health, High Street, Randwick, New South Wales 2031, Sydney, Australia
| | - Peter R Durie
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children 555 University Avenue Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
42
|
|
43
|
Borowitz D. CFTR, bicarbonate, and the pathophysiology of cystic fibrosis. Pediatr Pulmonol 2015; 50 Suppl 40:S24-S30. [PMID: 26335950 DOI: 10.1002/ppul.23247] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/26/2015] [Accepted: 06/03/2015] [Indexed: 12/18/2022]
Abstract
The gene that encodes for the cystic fibrosis transmembrane regulator protein (CFTR) was identified in 1989, yet major pathophysiologic questions remain unanswered. There is emerging evidence that CFTR is a bicarbonate channel, a driver of chloride-bicarbonate exchange and through its action on local pH, a regulator of other ion channels and of proteins that function optimally in a neutral environment. In both the respiratory and gastrointestinal (GI) tracts, bicarbonate drives ionic content and fluid on epithelial surfaces, allows mucins to unfold and become slippery, and contributes to innate immunity. In the GI tract bicarbonate neutralizes gastric acid to support digestion and absorption. When CFTR is dysfunctional, lack of bicarbonate secretion disrupts these normal processes and thus leads directly to the clinical symptoms and signs of CF. This article synthesizes evidence from cell, animal, and human investigations that support these concepts. Bicarbonate secretion does not seem to be the same in all tissues and varies with physiologic demand. Thus, tissue type and whether conditions are baseline or stimulated needs to be taken into account when evaluating the evidence concerning the role of bicarbonate in the pathophysiology of CF as a regulator of local pH. Basic and applied research that focuses on the role of CFTR-mediated bicarbonate secretion helps explain many of the diverse clinical manifestations that are CF.
Collapse
Affiliation(s)
- Drucy Borowitz
- University at Buffalo, State University of New York, Buffalo, New York
| |
Collapse
|
44
|
Bazett M, Honeyman L, Stefanov AN, Pope CE, Hoffman LR, Haston CK. Cystic fibrosis mouse model-dependent intestinal structure and gut microbiome. Mamm Genome 2015; 26:222-34. [PMID: 25721416 DOI: 10.1007/s00335-015-9560-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/13/2015] [Indexed: 01/15/2023]
Abstract
Mice with a null mutation in the cystic fibrosis transmembrane conductance regulator (Cftr) gene show intestinal structure alterations and bacterial overgrowth. To determine whether these changes are model-dependent and whether the intestinal microbiome is altered in cystic fibrosis (CF) mouse models, we characterized the ileal tissue and intestinal microbiome of mice with the clinically common ΔF508 Cftr mutation (FVB/N Cftr(tm1Eur)) and with Cftr null mutations (BALB/c Cftr(tm1UNC) and C57BL/6 Cftr(tm1UNC)). Intestinal disease in 12-week-old CF mice, relative to wild-type strain controls, was measured histologically. The microbiome was characterized by pyrosequencing of the V4-V6 region of the 16S rRNA gene and intestinal load was measured by RT-PCR of the 16S rRNA gene. The CF-associated increases in ileal crypt to villus axis distention, goblet cell hyperplasia, and muscularis externa thickness were more severe in the BALB/c and C57BL/6 Cftr(tm1UNC) mice than in the FVB/N Cftr(tm1Eur) mice. Intestinal bacterial load was significantly increased in all CF models, compared to levels in controls, and positively correlated with circular muscle thickness in CF, but not wild-type, mice. Microbiome profiling identified Bifidobacterium and groups of Lactobacillus to be of altered abundance in the CF mice but overall bacterial frequencies were not common to the three CF strains and were not correlative of major histological changes. In conclusion, intestinal structure alterations, bacterial overgrowth, and dysbiosis were each more severe in BALB/c and C57BL/6 Cftr(tm1UNC) mice than in the FVB/N Cftr(tm1Eur) mice. The intestinal microbiome differed among the three CF mouse models.
Collapse
Affiliation(s)
- Mark Bazett
- Meakins-Christie Laboratories, Departments of Medicine and Human Genetics, McGill University, 3626 St. Urbain, Montreal, QC, H2X 2P2, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Shalem T, Weiss B. Probiotic Supplementation and Pulmonary Exacerbations in Patients with Cystic Fibrosis. DIET AND EXERCISE IN CYSTIC FIBROSIS 2015:243-246. [DOI: 10.1016/b978-0-12-800051-9.00028-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
46
|
Wilhelm CL, Yarovinsky F. Apicomplexan infections in the gut. Parasite Immunol 2014; 36:409-20. [PMID: 25201405 DOI: 10.1111/pim.12115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 03/20/2014] [Indexed: 12/12/2022]
Abstract
Toxoplasma gondii and Cryptosporidium parvum are intracellular protozoan parasites that establish infection through the small intestinal bowel after the ingestion of contaminated food products. These Apicomplexan parasites have emerged as an important cause of chronic and fatal disease in immunodeficient individuals, in addition to being investigated as possible triggers of inflammatory bowel disease. T. gondii disseminates to the brain and other tissues after infection, whereas C. parvum remains localized to the intestine. In the following review, we will discuss the pathogenesis of these parasitic diseases in the small intestine, the site of initial invasion. Themes include the sequence of invasion, the structure of Th1 immunity provoked by these parasites and the contribution of intestinal microbiota to the development of the mucosal immune response.
Collapse
Affiliation(s)
- C L Wilhelm
- Departments of Immunology, University of Texas Southwestern Medical School, Dallas, TX, USA
| | | |
Collapse
|
47
|
Reed DE, Pigrau M, Lu J, Moayyedi P, Collins SM, Bercik P. Bead study: a novel method to measure gastrointestinal transit in mice. Neurogastroenterol Motil 2014; 26:1663-8. [PMID: 25264134 DOI: 10.1111/nmo.12442] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/21/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Intestinal transit assessment in mice using existing methods requires long recording periods or euthanization of animals to localize a tracer. We have developed a novel in vivo method to assess gastrointestinal (GI) transit in mice based on a clinically used 'shapes study'. METHODS Mice (n = 70) were gavaged with 5 steel beads and barium 3 h before, with another dose of barium gavaged 10 min before imaging. Mice were fluoroscoped for 20-60 s, and then most of them were euthanized and the GI tract removed to confirm the localization of the beads fluoroscopically. The in vivo and postmortem recordings were analyzed and each bead was scored depending on its location; a total score was calculated by adding individual bead scores. Total scores obtained from the two methods were compared. A group of mice (n = 10) were examined on three occasions, before and after treatment with loperamide or prucalopride. KEY RESULTS The stomach and cecum were consistently outlined by barium, serving as reference landmarks. There was an excellent overall correlation between in vivo and postmortem transit scores (r = 0.93). Analysis of scores for individual gut segments revealed high agreement for stomach, cecum, and expelled beads, and moderate agreement for the small bowel and colon. Gastrointestinal transit scores were decreased by loperamide and increased by prucalopride compared with baseline. CONCLUSIONS & INFERENCES Metallic beads are reliably localized by videofluoroscopy in vivo within the GI tract. This novel imaging method enables repetitive measurements of GI transit in vivo and detects changes induced by motility-modifying agents.
Collapse
Affiliation(s)
- D E Reed
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Mastroianni JR, Lu W, Selsted ME, Ouellette AJ. Differential Susceptibility of Bacteria to Mouse Paneth Cell α-Defensins under Anaerobic Conditions. Antibiotics (Basel) 2014; 3:493-508. [PMID: 25383215 PMCID: PMC4220453 DOI: 10.3390/antibiotics3040493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Small intestinal Paneth cells secrete α-defensin peptides, termed cryptdins (Crps) in mice, into the intestinal lumen, where they confer immunity to oral infections and define the composition of the ileal microbiota. In these studies, facultative bacteria maintained under aerobic or anaerobic conditions displayed differential sensitivities to mouse α-defensins under in vitro assay conditions. Regardless of oxygenation, Crps 2 and 3 had robust and similar bactericidal activities against S. typhimurium and S. flexneri, but Crp4 activity against S. flexneri was attenuated in the absence of oxygen. Anaerobic bacteria varied in their susceptibility to Crps 2-4, with Crp4 showing less activity than Crps 2 and 3 against Enterococcus faecalis, and Bacteroides fragilis in anaerobic assays, but Fusobacterium necrophorum was killed only by Crp4 and not by Crps 2 and 3. The influence of anaerobiosis in modulating Crp bactericidal activities in vitro suggests that α-defensin effects on the enteric microbiota may be subject to regulation by local oxygen tension.
Collapse
Affiliation(s)
- Jennifer R. Mastroianni
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC Norris Cancer Center, Los Angeles, CA 90089-9601, USA; E-Mails: (J.R.M.); (M.E.S.)
| | - Wuyuan Lu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Institute of Human Virology, Baltimore, MD 21201, USA; E-Mail:
| | - Michael E. Selsted
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC Norris Cancer Center, Los Angeles, CA 90089-9601, USA; E-Mails: (J.R.M.); (M.E.S.)
| | - André J. Ouellette
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC Norris Cancer Center, Los Angeles, CA 90089-9601, USA; E-Mails: (J.R.M.); (M.E.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-323-442-7959
| |
Collapse
|
49
|
Digestive system dysfunction in cystic fibrosis: challenges for nutrition therapy. Dig Liver Dis 2014; 46:865-74. [PMID: 25053610 DOI: 10.1016/j.dld.2014.06.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/10/2014] [Accepted: 06/28/2014] [Indexed: 02/08/2023]
Abstract
Cystic fibrosis can affect food digestion and nutrient absorption. The underlying mutation of the cystic fibrosis trans-membrane regulator gene depletes functional cystic fibrosis trans-membrane regulator on the surface of epithelial cells lining the digestive tract and associated organs, where Cl(-) secretion and subsequently secretion of water and other ions are impaired. This alters pH and dehydrates secretions that precipitate and obstruct the lumen, causing inflammation and the eventual degradation of the pancreas, liver, gallbladder and intestine. Associated conditions include exocrine pancreatic insufficiency, impaired bicarbonate and bile acid secretion and aberrant mucus formation, commonly leading to maldigestion and malabsorption, particularly of fat and fat-soluble vitamins. Pancreatic enzyme replacement therapy is used to address this insufficiency. The susceptibility of pancreatic lipase to acidic and enzymatic inactivation and decreased bile availability often impedes its efficacy. Brush border digestive enzyme activity and intestinal uptake of certain disaccharides and amino acids await clarification. Other complications that may contribute to maldigestion/malabsorption include small intestine bacterial overgrowth, enteric circular muscle dysfunction, abnormal intestinal mucus, and intestinal inflammation. However, there is some evidence that gastric digestive enzymes, colonic microflora, correction of fatty acid abnormalities using dietary n-3 polyunsaturated fatty acid supplementation and emerging intestinal biomarkers can complement nutrition management in cystic fibrosis.
Collapse
|
50
|
Ren W, Chen S, Yin J, Duan J, Li T, Liu G, Feng Z, Tan B, Yin Y, Wu G. Dietary arginine supplementation of mice alters the microbial population and activates intestinal innate immunity. J Nutr 2014; 144:988-95. [PMID: 24670969 DOI: 10.3945/jn.114.192120] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Currently, little is known about the function of arginine in the homeostasis of the intestinal immune system. This study was conducted to test the hypothesis that dietary arginine supplementation may alter intestinal microbiota and innate immunity in mice. Mice were fed a basal diet (containing 0.93% l-arginine; grams per gram) or the basal diet supplemented with 0.5% l-arginine for 14 d. We studied the composition of intestinal microbiota, the activation of innate immunity, and the expression of toll-like receptors (Tlrs), proinflammatory cytokines, and antimicrobials in the jejunum, ileum, or colon of mice. Signal transduction pathway activation in the jejunum and ileum, including TLR4-nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), mitogen-activated protein kinase (MAPK), and phosphoinositide-3 kinase (PI3K)/PI3K-protein kinase B (Akt), was analyzed by Western blotting. Quantitative polymerase chain reaction analysis revealed that arginine supplementation induced (P < 0.05) a shift in the Firmicutes-to-Bacteroidetes ratio to favor Bacteroidetes in the jejunum (0.33 ± 0.04 vs. 1.0 ± 0.22) and ileum (0.20 ± 0.08 vs. 1.0 ± 0.27) compared with the control group. This finding coincided with greater (P < 0.05) activation of the innate immune system, including TLR signaling, as well as expression of proinflammatory cytokines, secretory immunoglobulin A, mucins, and Paneth antimicrobials in the jejunum and ileum. Finally, arginine supplementation reduced (P < 0.05) expression of the proteins for NF-κB, MAPK, and PI3K-Akt signaling pathways but activated (P < 0.05) p38 and c-Jun N-terminal protein kinase in the jejunum and the ileum, respectively. Collectively, dietary arginine supplementation of mice changes the intestinal microbiota, contributing to the activation of intestinal innate immunity through NF-κB, MAPK, and PI3K-phosphorylated Akt signaling pathways.
Collapse
Affiliation(s)
- Wenkai Ren
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Shuai Chen
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Jie Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Jielin Duan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Tiejun Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Gang Liu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Zemeng Feng
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Bie Tan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Yulong Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and
| | - Guoyao Wu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; and Department of Animal Science, Texas A&M University, College Station, TX
| |
Collapse
|