1
|
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
|
2
|
Tauber M, Basso L, Martin J, Bostan L, Pinto MM, Thierry GR, Houmadi R, Serhan N, Loste A, Blériot C, Kamphuis JB, Grujic M, Kjellén L, Pejler G, Paul C, Dong X, Galli SJ, Reber LL, Ginhoux F, Bajenoff M, Gentek R, Gaudenzio N. Landscape of mast cell populations across organs in mice and humans. J Exp Med 2023; 220:e20230570. [PMID: 37462672 PMCID: PMC10354537 DOI: 10.1084/jem.20230570] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
Mast cells (MCs) are tissue-resident immune cells that exhibit homeostatic and neuron-associated functions. Here, we combined whole-tissue imaging and single-cell RNA sequencing datasets to generate a pan-organ analysis of MCs in mice and humans at steady state. In mice, we identify two mutually exclusive MC populations, MrgprB2+ connective tissue-type MCs and MrgprB2neg mucosal-type MCs, with specific transcriptomic core signatures. While MrgprB2+ MCs develop in utero independently of the bone marrow, MrgprB2neg MCs develop after birth and are renewed by bone marrow progenitors. In humans, we unbiasedly identify seven MC subsets (MC1-7) distributed across 12 organs with different transcriptomic core signatures. MC1 are preferentially enriched in the bladder, MC2 in the lungs, and MC4, MC6, and MC7 in the skin. Conversely, MC3 and MC5 are shared by most organs but not skin. This comprehensive analysis offers valuable insights into the natural diversity of MC subtypes in both mice and humans.
Collapse
Affiliation(s)
- Marie Tauber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Lilian Basso
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Jeremy Martin
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Luciana Bostan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Marlene Magalhaes Pinto
- Centre for Inflammation Research and Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Guilhem R. Thierry
- Aix Marseille University, CNRS, INSERM, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Raïssa Houmadi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Nadine Serhan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Alexia Loste
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Camille Blériot
- Institut Necker des Enfants Malades, CNRS UMR8253, Paris, France
| | - Jasper B.J. Kamphuis
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Mirjana Grujic
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lena Kjellén
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carle Paul
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
- Toulouse University and Centre Hospitalier Universitaire, Toulouse, France
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Center for Sensory Biology, Johns Hopkins University, Baltimore, MD, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen J. Galli
- Departments of Pathology and Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Laurent L. Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1015, Gustave Roussy, Villejuif, France
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Marc Bajenoff
- Aix Marseille University, CNRS, INSERM, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Rebecca Gentek
- Centre for Inflammation Research and Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERMUMR1291—CNRS UMR5051—University Toulouse III, Toulouse, France
- Genoskin SAS, Toulouse, France
| |
Collapse
|
3
|
Wang Y, Huang B, Jin T, Ocansey DKW, Jiang J, Mao F. Intestinal Fibrosis in Inflammatory Bowel Disease and the Prospects of Mesenchymal Stem Cell Therapy. Front Immunol 2022; 13:835005. [PMID: 35370998 PMCID: PMC8971815 DOI: 10.3389/fimmu.2022.835005] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal fibrosis is an important complication of inflammatory bowel disease (IBD). In the course of the development of fibrosis, certain parts of the intestine become narrowed, significantly destroying the structure and function of the intestine and affecting the quality of life of patients. Chronic inflammation is an important initiating factor of fibrosis. Unfortunately, the existing anti-inflammatory drugs cannot effectively prevent and alleviate fibrosis, and there is no effective anti-fibrotic drug, which makes surgical treatment the mainstream treatment for intestinal fibrosis and stenosis. Mesenchymal stem cells (MSCs) are capable of tissue regeneration and repair through their self-differentiation, secretion of cytokines, and secretion of extracellular vesicles. MSCs have been shown to play an important therapeutic role in the fibrosis of many organs. However, the role of MSC in intestinal fibrosis largely remained unexplored. This review summarizes the mechanism of intestinal fibrosis, including the role of immune cells, TGF-β, and the gut microbiome and metabolites. Available treatment options for fibrosis, particularly, MSCs are also discussed.
Collapse
Affiliation(s)
- Yifei Wang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bin Huang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- General Surgery Department, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| | - Fei Mao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| |
Collapse
|
4
|
Madla CM, Gavins FKH, Trenfield SJ, Basit AW. Special Populations. BIOPHARMACEUTICS 2022:205-237. [DOI: 10.1002/9781119678366.ch13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Tam RY, van Dorst JM, McKay I, Coffey M, Ooi CY. Intestinal Inflammation and Alterations in the Gut Microbiota in Cystic Fibrosis: A Review of the Current Evidence, Pathophysiology and Future Directions. J Clin Med 2022; 11:649. [PMID: 35160099 PMCID: PMC8836727 DOI: 10.3390/jcm11030649] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-limiting autosomal recessive multisystem disease. While its burden of morbidity and mortality is classically associated with pulmonary disease, CF also profoundly affects the gastrointestinal (GI) tract. Chronic low-grade inflammation and alterations to the gut microbiota are hallmarks of the CF intestine. The etiology of these manifestations is likely multifactorial, resulting from cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction, a high-fat CF diet, and the use of antibiotics. There may also be a bidirectional pathophysiological link between intestinal inflammation and changes to the gut microbiome. Additionally, a growing body of evidence suggests that these GI manifestations may have significant clinical associations with growth and nutrition, quality of life, and respiratory function in CF. As such, the potential utility of GI therapies and long-term GI outcomes are areas of interest in CF. Further research involving microbial modulation and multi-omics techniques may reveal novel insights. This article provides an overview of the current evidence, pathophysiology, and future research and therapeutic considerations pertaining to intestinal inflammation and alterations in the gut microbiota in CF.
Collapse
Affiliation(s)
- Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Isabelle McKay
- Wagga Wagga Base Hospital, Wagga Wagga, NSW 2650, Australia;
| | - Michael Coffey
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, 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, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| |
Collapse
|
6
|
Lin S, Kühn F, Schiergens TS, Zamyatnin AA, Isayev O, Gasimov E, Werner J, Li Y, Bazhin AV. Experimental postoperative ileus: is Th2 immune response involved? Int J Med Sci 2021; 18:3014-3025. [PMID: 34220330 PMCID: PMC8241774 DOI: 10.7150/ijms.59354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Rationale: Postoperative ileus (POI) is a frequent complication arising after gastrointestinal surgery but pathogenesis of POI is still not fully understood. While Th1 immune cells are implicated in POI, the involvement of Th2 cells has not yet been clarified. Given the impact of reactive oxygen species (ROS) in the regulation of Th1 and Th2 balance, we hypothesized that not only Th1 but also Th2 immune response can be involved in the development of experimental POI. Methods: The intestinal transit test was performed using carbon gum arabic. Electron microscopy was employed to assess tissue morphology and the presence of immune cells. Cytokines, IgE and ROS were measured. Immune cells from Peyer's patches were analyzed by Flow Cytometry and toluidine blue staining was used for detection of mast cells. Transcriptional factors were analyzed by Western blot. Results: POI is associated with an increase in both Th2 cytokines and Th2 cells. We have further demonstrated that POI induces a Th2-dependent activation of memory and non-memory B cells. This was accompanied by an increase in a number of mast cells in the colon of POI mice as well by an increased IgE and histamine plasma levels. We found that POI-induced accumulation of ROS was associated with an increased expression of the transcriptional factors HMBGI, NF-κB, and p38. This increased expression seemed to be associated with a Th2 response. Conclusion: Th2 immune response can be involved in the activation of mast cells in POI, which was associated with ROS mediated activation of NF-κB and p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Sisi Lin
- Department of Pathophysiology, Institute of Digestive Disease, Tongji University School of Medicine, 200092, Shanghai, China.,Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, 81377, Munich, Germany
| | - Florian Kühn
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, 81377, Munich, Germany
| | - Tobias S Schiergens
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.,Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Orkhan Isayev
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Eldar Gasimov
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Jens Werner
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Yongyu Li
- Department of Pathophysiology, Institute of Digestive Disease, Tongji University School of Medicine, 200092, Shanghai, China
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| |
Collapse
|
7
|
Rao MC. Physiology of Electrolyte Transport in the Gut: Implications for Disease. Compr Physiol 2019; 9:947-1023. [PMID: 31187895 DOI: 10.1002/cphy.c180011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We now have an increased understanding of the genetics, cell biology, and physiology of electrolyte transport processes in the mammalian intestine, due to the availability of sophisticated methodologies ranging from genome wide association studies to CRISPR-CAS technology, stem cell-derived organoids, 3D microscopy, electron cryomicroscopy, single cell RNA sequencing, transgenic methodologies, and tools to manipulate cellular processes at a molecular level. This knowledge has simultaneously underscored the complexity of biological systems and the interdependence of multiple regulatory systems. In addition to the plethora of mammalian neurohumoral factors and their cross talk, advances in pyrosequencing and metagenomic analyses have highlighted the relevance of the microbiome to intestinal regulation. This article provides an overview of our current understanding of electrolyte transport processes in the small and large intestine, their regulation in health and how dysregulation at multiple levels can result in disease. Intestinal electrolyte transport is a balance of ion secretory and ion absorptive processes, all exquisitely dependent on the basolateral Na+ /K+ ATPase; when this balance goes awry, it can result in diarrhea or in constipation. The key transporters involved in secretion are the apical membrane Cl- channels and the basolateral Na+ -K+ -2Cl- cotransporter, NKCC1 and K+ channels. Absorption chiefly involves apical membrane Na+ /H+ exchangers and Cl- /HCO3 - exchangers in the small intestine and proximal colon and Na+ channels in the distal colon. Key examples of our current understanding of infectious, inflammatory, and genetic diarrheal diseases and of constipation are provided. © 2019 American Physiological Society. Compr Physiol 9:947-1023, 2019.
Collapse
Affiliation(s)
- Mrinalini C Rao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
8
|
Hatton GB, Madla CM, Rabbie SC, Basit AW. Gut reaction: impact of systemic diseases on gastrointestinal physiology and drug absorption. Drug Discov Today 2018; 24:417-427. [PMID: 30453059 DOI: 10.1016/j.drudis.2018.11.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/25/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022]
Abstract
It was in 400 BC that Hippocrates reportedly stated that "death sits in the colon". The growth in our knowledge of the intestinal microbiome, the gut-brain axis and their function and imbalance has distinctly uncovered the complex relationship between the gut to disease predisposition and development, heralding the problem and the solution to disease pathology. Human studies of new drug molecules are typically performed in healthy volunteers and their specific disease indication. Approved drugs, however, are used by patients with diverse disease backgrounds. Here, we review the current literature of the gastrointestinal tract reacting to systemic disease pathology that elicits physiological and functional changes that consequently affect oral drug product performance.
Collapse
Affiliation(s)
- Grace B Hatton
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Christine M Madla
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Sarit C Rabbie
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
9
|
Wang X, Hao GL, Gao CC, Wang YX, Liu YH, Qiu ZQ, Li LS, Xu JD. Intestinal mast cells and their function. Shijie Huaren Xiaohua Zazhi 2018; 26:601-608. [DOI: 10.11569/wcjd.v26.i10.601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mast cells develop from the CD34+ precursor cells in bone marrow, are activated in the gut, and can release a variety of bioactive mediators, including histamine, 5-hydroxytryptamine, and tryptase. They play a crucial role in intestinal innate and adaptive immunity because of their diverse secretory granules and unique mature characteristics. Many studies have shown that a variety of intestinal diseases have close relationship with mast cells, especially inflammatory bowel disease, irritable bowel syndrome, and intestinal allergic diseases, which has attracted extensive attention. In this paper, we review the function and mechanism of intestinal mast cells and their role in the treatment of related clinical diseases.
Collapse
Affiliation(s)
- Xue Wang
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Gui-Liang Hao
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | | | | | - Yue-Hong Liu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | | | | | | |
Collapse
|
10
|
Sathe MN, Freeman AJ. Gastrointestinal, Pancreatic, and Hepatobiliary Manifestations of Cystic Fibrosis. Pediatr Clin North Am 2016; 63:679-98. [PMID: 27469182 DOI: 10.1016/j.pcl.2016.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pulmonary disease is the primary cause of morbidity and mortality in people with cystic fibrosis (CF), but significant involvement within gastrointestinal, pancreatic, and hepatobiliary systems occurs as well. As in the airways, defects in CFTR alter epithelial surface fluid, mucus viscosity, and pH, increasing risk of stasis through the various hollow epithelial-lined structures of the gastrointestinal tract. This exerts secondary influences that are responsible for most gastrointestinal, pancreatic, and hepatobiliary manifestations of CF. Understanding these gastrointestinal morbidities of CF is essential in understanding and treating CF as a multisystem disease process and improving overall patient care.
Collapse
Affiliation(s)
- Meghana Nitin Sathe
- Division of Pediatric Gastroenterology and Nutrition, Children's Health, University of Texas Southwestern, F4.06, 1935 Medical District Drive, Dallas, TX 75235, USA
| | - Alvin Jay Freeman
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Healthcare of Atlanta, Emory University, 2015 Uppergate Drive, Northeast, Atlanta, GA 30322, USA.
| |
Collapse
|
11
|
Haller W, Ledder O, Lewindon PJ, Couper R, Gaskin KJ, Oliver M. Cystic fibrosis: An update for clinicians. Part 1: Nutrition and gastrointestinal complications. J Gastroenterol Hepatol 2014; 29:1344-55. [PMID: 25587613 DOI: 10.1111/jgh.12546] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Com G, Cetin N, O'Brien CE. Complicated Clostridium difficile colitis in children with cystic fibrosis: association with gastric acid suppression? J Cyst Fibros 2013; 13:37-42. [PMID: 23993432 DOI: 10.1016/j.jcf.2013.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/02/2013] [Accepted: 08/07/2013] [Indexed: 01/06/2023]
Abstract
Patients with cystic fibrosis (CF) have several risk factors for Clostridium difficile colonization such as frequent hospitalization and exposure to a broad array of antibiotics utilized for the control, eradication, and prophylaxis of respiratory pathogens. However, despite this high rate of colonization, the occurrence of C. difficile infection (CDI) in CF is rare. We report three children with CF who presented with severe community-associated CDI. All three children had complicated courses and one died. These children were in good health without significant morbidities, and were not frequently hospitalized nor did they receive frequent antibiotic courses. The occurrence of 3 severe cases within a 15-month period prompted us to report these cases and review the literature in regard to CDI. We reviewed the CF GI tract as possible risk factors for a high rate of C. difficile colonization in individuals with CF. Since a high percentage of individuals with CF are on gastric acid blocking agents, we also focused on gastric acid suppression as a potential risk factor for CDI.
Collapse
Affiliation(s)
- G Com
- University of Arkansas Medical Sciences, Department of Pediatrics, Pediatric Pulmonology, Arkansas Children's Hospital, United States.
| | - N Cetin
- University of Arkansas Medical Sciences, Department of Pathology, United States
| | - C E O'Brien
- University of Arkansas Medical Sciences, College of Pharmacy, Department of Pharmacy Practice, United States; College of Medicine, Department of Pediatrics, Division of Pharmacology/Toxicology, United States
| |
Collapse
|
13
|
Lv XX, Wang XX, Li K, Wang ZY, Li Z, Lv Q, Fu XM, Hu ZW. Rupatadine protects against pulmonary fibrosis by attenuating PAF-mediated senescence in rodents. PLoS One 2013; 8:e68631. [PMID: 23869224 PMCID: PMC3711902 DOI: 10.1371/journal.pone.0068631] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/30/2013] [Indexed: 12/11/2022] Open
Abstract
A similar immune response is implicated in the pathogenesis of pulmonary fibrosis and allergic disorders. We investigated the potential therapeutic efficacy and mechanism of rupatadine, a dual antagonist of histamine and platelet-activation factor (PAF), in bleomycin- (BLM-) and silica-induced pulmonary fibrosis. The indicated dosages of rupatadine were administered in rodents with bleomycin or silica-induced pulmonary fibrosis. The tissue injury, fibrosis, inflammatory cells and cytokines, and lung function were examined to evaluate the therapeutic efficacy of rupatadine. The anti-fibrosis effect of rupatadine was compared with an H1 or PAF receptor antagonist, and efforts were made to reveal rupatadine’s anti-fibrotic mechanism. Rupatadine promoted the resolution of pulmonary inflammation and fibrosis in a dose-dependent manner, as indicated by the reductions in inflammation score, collagen deposition and epithelial-mesenchymal transformation, and infiltration or expression of inflammatory cells or cytokines in the fibrotic lung tissue. Thus, rupatadine treatment improved the declined lung function and significantly decreased animal death. Moreover, rupatadine was able not only to attenuate silica-induced silicosis but also to produce a superior therapeutic efficacy compared to pirfenidone, histamine H1 antagonist loratadine, or PAF antagonist CV-3988. The anti-fibrotic action of rupatadine might relate to its attenuation of BLM- or PAF-induced premature senescence because rupatadine treatment protected against the in vivo and in vitro activation of the p53/p21-dependent senescence pathway. Our studies indicate that rupatadine promotes the resolution of pulmonary inflammation and fibrosis by attenuating the PAF-mediated senescence response. Rupatadine holds promise as a novel drug to treat the devastating disease of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiao-xi Lv
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Xiao-xing Wang
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Ke Li
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Zi-yan Wang
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Zhe Li
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Qi Lv
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Xiao-ming Fu
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Zhuo-Wei Hu
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
- * E-mail:
| |
Collapse
|
14
|
How the airway smooth muscle in cystic fibrosis reacts in proinflammatory conditions: implications for airway hyper-responsiveness and asthma in cystic fibrosis. THE LANCET RESPIRATORY MEDICINE 2013; 1:137-47. [PMID: 24429094 DOI: 10.1016/s2213-2600(12)70058-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Among patients with cystic fibrosis there is a high prevalence (40-70%) of asthma signs and symptoms such as cough and wheezing and airway hyper-responsiveness to inhaled histamine or methacholine. Whether these abnormal airway responses are due to a primary deficiency in the cystic fibrosis transmembrane conductance regulator (CFTR) or are secondary to the inflammatory environment in the cystic fibrosis lungs is not clear. A role for the CFTR in smooth muscle function is emerging, and alterations in contractile signalling have been reported in CFTR-deficient airway smooth muscle. Persistent bacterial infection, especially with Pseudomonas aeruginosa, stimulates interleukin-8 release from the airway epithelium, resulting in neutrophilic inflammation. Increased neutrophilia and skewing of CFTR-deficient T-helper cells to type 2 helper T cells creates an inflammatory environment characterised by high concentrations of tumour necrosis factor α, interleukin-8, and interleukin-13, which might all contribute to increased contractility of airway smooth muscle in cystic fibrosis. An emerging role of interleukin-17, which is raised in patients with cystic fibrosis, in airway smooth muscle proliferation and hyper-responsiveness is apparent. Increased understanding of the molecular mechanisms responsible for the altered smooth muscle physiology in patients with cystic fibrosis might provide insight into airway dysfunction in this disease.
Collapse
|
15
|
Risse PA, Kachmar L, Matusovsky OS, Novali M, Gil FR, Javeshghani S, Keary R, Haston CK, Michoud MC, Martin JG, Lauzon AM. Ileal smooth muscle dysfunction and remodeling in cystic fibrosis. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1-8. [PMID: 22538405 DOI: 10.1152/ajpgi.00356.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Patients with cystic fibrosis (CF) often suffer from gastrointestinal cramps and intestinal obstruction. The CF transmembrane conductance regulator (CFTR) channel has been shown to be expressed in vascular and airway smooth muscle (SM). We hypothesized that the absence of CFTR expression alters the gastrointestinal SM function and that these alterations may show strain-related differences in the mouse. The aim of this study was to measure the contractile properties of the ileal SM in two CF mouse models. CFTR(-/-) and CFTR(+/+) mice were studied on BALB/cJ and C57BL/6J backgrounds. Responsiveness of ileal strips to electrical field stimulation (EFS), methacholine (MCh), and isoproterenol was measured. The mass and the cell density of SM layers were measured morphometrically. Finally, the maximal velocity of shortening (Vmax) and the expression of the fast (+)insert myosin isoform were measured in the C57BL/6J ileum. Ileal hyperreactivity was observed in response to EFS and MCh in CFTR(-/-) compared with CFTR(+/+) mice in C57BL/6J background. This latter observation was not reproduced by acute inhibition of CFTR with CFTR(inh)172. BALB/cJ CFTR(-/-) mice exhibited a significant increase of SM mass with a lower density of cells compared with CFTR(+/+), whereas no difference was observed in the C57BL/6J background. In addition, in this latter strain, ileal strips from CFTR(-/-) exhibited a significant increase in Vmax compared with control and expressed a greater proportion of the fast (+)insert SM myosin isoform with respect to total myosin. BALB/cJ CFTR(-/-) ilium had a greater relaxation to isoproterenol than the CFTR(+/+) mice when precontracted with EFS, but no difference was observed in response to exogeneous MCh. In vivo, the lack of CFTR expression induces a different SM ileal phenotype in different mouse strains, supporting the importance of modifier genes in determining intestinal SM properties.
Collapse
Affiliation(s)
- P-A Risse
- Meakins-Christie Laboratories, McGill University, 3626 St.-Urbain St., Montréal, Québec, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
De Lisle RC, Mueller R, Roach E. Lubiprostone ameliorates the cystic fibrosis mouse intestinal phenotype. BMC Gastroenterol 2010; 10:107. [PMID: 20843337 PMCID: PMC2945989 DOI: 10.1186/1471-230x-10-107] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 09/15/2010] [Indexed: 12/16/2022] Open
Abstract
Background Cystic fibrosis (CF) is caused by mutations in the CFTR gene that impair the function of CFTR, a cAMP-regulated anion channel. In the small intestine loss of CFTR function creates a dehydrated, acidic luminal environment which is believed to cause an accumulation of mucus, a phenotype characteristic of CF. CF mice have small intestinal bacterial overgrowth, an altered innate immune response, and impaired intestinal transit. We investigated whether lubiprostone, which can activate the CLC2 Cl- channel, would improve the intestinal phenotype in CF mice. Methods Cftrtm1UNC (CF) and wildtype (WT) littermate mice on the C57BL/6J background were used. Lubiprostone (10 μg/kg-day) was administered by gavage for two weeks. Mucus accumulation was estimated from crypt lumen widths in periodic acid-Schiff base, Alcian blue stained sections. Luminal bacterial load was measured by qPCR for the bacterial 16S gene. Gastric emptying and small intestinal transit in fasted mice were assessed using gavaged rhodamine dextran. Gene expression was evaluated by Affymetrix Mouse430 2.0 microarray and qRT-PCR. Results Crypt width in control CF mice was 700% that of WT mice (P < 0.001). Lubiprostone did not affect WT crypt width but, unexpectedly, increased CF crypt width 22% (P = 0.001). Lubiprostone increased bacterial load in WT mice to 490% of WT control levels (P = 0.008). Conversely, lubiprostone decreased bacterial overgrowth in CF mice by 60% (P = 0.005). Lubiprostone increased gastric emptying at 20 min postgavage in both WT (P < 0.001) and CF mice (P < 0.001). Lubiprostone enhanced small intestinal transit in WT mice (P = 0.024) but not in CF mice (P = 0.377). Among other innate immune markers, expression of mast cell genes was elevated 4-to 40-fold in the CF intestine as compared to WT, and lubiprostone treatment of CF mice decreased expression to WT control levels. Conclusions These results indicate that lubiprostone has some benefits for the CF intestinal phenotype, especially on bacterial overgrowth and the innate immune response. The unexpected observation of increased mucus accumulation in the crypts of lubiprostone-treated CF mice suggests the possibility that lubiprostone increases mucus secretion.
Collapse
Affiliation(s)
- Robert C De Lisle
- Anatomy & Cell Biology, University of Kansas School of Medicine, Kansas City, KS 66160, USA.
| | | | | |
Collapse
|
17
|
Canale-Zambrano JC, Auger ML, Haston CK. Toll-like receptor-4 genotype influences the survival of cystic fibrosis mice. Am J Physiol Gastrointest Liver Physiol 2010; 299:G381-90. [PMID: 20522639 DOI: 10.1152/ajpgi.00003.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Toll-like receptor (Tlr) 4 is a lipopolysaccharide (LPS) receptor that contributes to the regulation of intestinal cell homeostasis, a condition that is altered in the intestines of cystic fibrosis mice. Herein, we assessed whether Tlr4 genotype influences cystic fibrosis intestinal disease by producing and phenotyping 12-wk (adult)- and 4-day (neonate)-old mice derived from BALB cystic fibrosis transmembrane conductance regulator, Cftr(+/tm1Unc) and C.C3-Tlr4(Lps-d)/J (Tlr4(-/-)), progenitors. Intestinal disease was assayed through mouse survival, crypt-villus axis (CVA) length, cell proliferation, bacterial load, bacterial classification, inflammatory cell infiltrate, and mucus content measures. Of the 77 Cftr(-/-) (CF) mice produced, only one Cftr/Tlr4 double-mutant mouse lived to the age of 12 wk while the majority of the remainder succumbed at approximately 4 days of age. The survival of CF Tlr4(+/-) mice exceeded that of both CF Tlr4(+/+) and Cftr/Tlr4 double-mutant mice. Adult CF mice presented increased Tlr4 expression, CVA length, crypt cell proliferation, and bacterial load relative to non-CF mice, but no differences were detected in Tlr4(+/-) compared with Tlr4(+/+) CF mice. The double-mutant neonates did not differ from Tlr4(+/+) or Tlr4(+/-) CF mice by intestinal CVA length or bacterial load, but fewer Tlr4(+/-) CF neonates presented with luminal mucus obstruction in the distal ileum, and the intestinal mast cell increase of CF mice was not evident in double-mutant neonates. We conclude that Tlr4 deficiency reduces the survival, but does not alter the intestinal phenotypes, of extended CVA or increased bacterial load in BALB CF mice.
Collapse
|
18
|
Meyerholz DK, Stoltz DA, Namati E, Ramachandran S, Pezzulo AA, Smith AR, Rector MV, Suter MJ, Kao S, McLennan G, Tearney GJ, Zabner J, McCray PB, Welsh MJ. Loss of cystic fibrosis transmembrane conductance regulator function produces abnormalities in tracheal development in neonatal pigs and young children. Am J Respir Crit Care Med 2010; 182:1251-61. [PMID: 20622026 DOI: 10.1164/rccm.201004-0643oc] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
RATIONALE Although airway abnormalities are common in patients with cystic fibrosis (CF), it is unknown whether they are all secondary to postnatal infection and inflammation, which characterize the disease. OBJECTIVES To learn whether loss of the cystic fibrosis transmembrane conductance regulator (CFTR) might affect major airways early in life, before the onset of inflammation and infection. METHODS We studied newborn CFTR⁻(/)⁻ pig trachea, using computed tomography (CT) scans, pathology, and morphometry. We retrospectively analyzed trachea CT scans in young children with CF and also previously published data of infants with CF. MEASUREMENTS AND MAIN RESULTS We discovered three abnormalities in the porcine CF trachea. First, the trachea and mainstem bronchi had a uniformly small caliber and cross-sections of trachea were less circular than in controls. Second, trachealis smooth muscle had an altered bundle orientation and increased transcripts in a smooth muscle gene set. Third, submucosal gland units occurred with similar frequency in the mucosa of CF and control airways, but CF submucosal glands were hypoplastic and had global reductions in tissue-specific transcripts. To learn whether any of these changes occurred in young patients with CF, we examined CT scans from children 2 years of age and younger, and found that CF tracheas were less circular in cross-section, but lacked differences in lumen area. However, analysis of previously published morphometric data showed reduced tracheal lumen area in neonates with CF. CONCLUSIONS Our findings in newborn CF pigs and young patients with CF suggest that airway changes begin during fetal life and may contribute to CF pathogenesis and clinical disease during postnatal life.
Collapse
|
19
|
Abstract
BACKGROUND Cystic fibrosis (CF) has multiple effects on the gastrointestinal system, including altered motility. The Cftr knockout mouse model of CF has impaired small intestinal transit but the mechanism is unknown. METHODS Behaviour of circular smooth muscle was studied in an organ bath. Expression levels of prostaglandin (PG) degradative genes were measured by quantitative RT-PCR, and PGE(2) levels were measured by enzyme immunoassay. KEY RESULTS Cystic fibrosis circular muscle activity was erratic and had variable frequency of contractions, as compared to WT. The CF tissue was non-responsive to cholinergic stimulation or direct KCl depolarization. PGE(2) and PGF(2alpha) are significantly elevated in the CF mouse small intestine, and we hypothesized these contribute to impaired smooth muscle activity. After inhibition of PG synthesis, the CF circular muscle exhibited greater cholinergic responsiveness, which was reversed by exogenous PGE(2). PGF(2alpha) enhanced activity of CF tissue only after inhibition of PG synthesis. The enteric microbiota was implicated in PGE(2)-mediated dysmotility because broad spectrum antibiotic treated WT mice, which have slowed transit, exhibit impaired circular muscle activity. This was accompanied by decreased expression of PG degradative genes and increased intestinal PGE(2) levels. Furthermore, administration of oral laxative, which eradicates bacterial overgrowth and improves transit in CF mice, increased expression of PG degradative genes, decreased PGE(2) levels, and improved CF muscle activity. CONCLUSIONS & INFERENCES These results suggest that the enteric microbiota modulates PGE(2) levels in a complex manner, which affects enteric smooth muscle activity and contributes to slower small intestinal transit in CF.
Collapse
Affiliation(s)
- R C de Lisle
- Anatomy & Cell Biology, University of Kansas School of Medicine, Kansas City, KS, USA.
| | | | | |
Collapse
|