1
|
Asensio-Grau A, Ferriz-Jordán M, Hervás D, Heredia A, García-Hernández J, Garriga M, Masip E, Carmen Collado M, Andrés A, Ribes-Koninckx C, Calvo-Lerma J. Faecal lipid profile as a new marker of fat maldigestion, malabsorption and microbiota. Pediatr Res 2024; 96:1794-1802. [PMID: 38778229 DOI: 10.1038/s41390-024-03209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Fat malabsorption in children with cystic fibrosis (CF) leads to poor nutritional status and altered colonic microbiota. This study aimed at establishing the faecal lipid profile in children with CF, and exploring associations between the faecal lipidome and microbiota. METHODS Cross-sectional observational study with children with CF and an age-matched control group. Faecal lipidome was analysed by UHLC-HRMS and microbiota profiling by 16S rRNA amplicon sequencing. RESULTS Among 234 identified lipid species, five lipidome clusters (LC) were obtained with significant differences in triacylglycerols (TG), diacylglycerols (DG), monoacylglycerols (MG) and fatty-acids (FA): LC1 subjects with good digestion and absorption: low TG and low MG and FA; LC2 good digestion and poor absorption: low TG and high MG and FA; LC3 Mild digestion and poor absorption: intermediate TG and high MG and FA; LC4 poor digestion and absorption: high TG and high MG and FA; LC5 outliers. Bacteroidota and Verrucomicrobiota decreased over LC1-LC4, while Proteobacteria increased. Nutritional status indicators were significantly higher in LC1 and decreased over LC2-LC4. CONCLUSION Assessing faecal lipidome may be relevant to determine how dietary lipids are digested and absorbed. This new evidence might be a method to support targeted nutritional interventions towards reverting fat maldigestion or malabsorption. IMPACT Lipidomic analysis enabled the identification of the lipid species related to maldigestion (triglycerides) or malabsorption (monoglycerides and fatty acids). Children with cystic fibrosis can be grouped depending on the faecal lipidome profile related to dietary fat maldigestion or malabsorption. The lipidome profile in faeces is related to the composition of microbiota and nutritional status indicators.
Collapse
Affiliation(s)
- Andrea Asensio-Grau
- Institute of Food Engineering (FoodUPV). Polytechnic University of València, 46022, València, Spain
- Joint Research Unit NutriCuraPDig, València, Spain
| | - Miguel Ferriz-Jordán
- Institute of Food Engineering (FoodUPV). Polytechnic University of València, 46022, València, Spain
| | - David Hervás
- Department of Statistics (EIO). Polytechnic University of València, 46022, València, Spain
| | - Ana Heredia
- Institute of Food Engineering (FoodUPV). Polytechnic University of València, 46022, València, Spain
- Joint Research Unit NutriCuraPDig, València, Spain
| | - Jorge García-Hernández
- Centre for Advanced Microbiology (CAMA). Polytechnic University of València, 46022, València, Spain
| | - María Garriga
- University Hospital Ramón y Cajal, 28034, Madrid, Spain
| | - Etna Masip
- Health Research Institute La Fe, 46026, València, Spain
| | - M Carmen Collado
- Institute of Agrochemistry and Food Technology. Spanish National Research Council (IATA-CSIC), 46980, València, Spain
| | - Ana Andrés
- Institute of Food Engineering (FoodUPV). Polytechnic University of València, 46022, València, Spain
- Joint Research Unit NutriCuraPDig, València, Spain
| | - Carmen Ribes-Koninckx
- Joint Research Unit NutriCuraPDig, València, Spain
- Health Research Institute La Fe, 46026, València, Spain
| | - Joaquim Calvo-Lerma
- Institute of Food Engineering (FoodUPV). Polytechnic University of València, 46022, València, Spain.
- Joint Research Unit NutriCuraPDig, València, Spain.
- Faculty of Pharmacy and Food Science. University of Valencia, Avda. Vicent Andrés Estellés s/n, Burjassot, 46100, València, Spain.
| |
Collapse
|
2
|
Woode RA, Strubberg AM, Liu J, Walker NM, Clarke LL. Increased activity of epithelial Cdc42 Rho GTPase and tight junction permeability in the Cftr knockout intestine. Am J Physiol Gastrointest Liver Physiol 2024; 327:G545-G557. [PMID: 39104325 DOI: 10.1152/ajpgi.00211.2022] [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: 08/26/2022] [Revised: 05/23/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024]
Abstract
Increased intestinal permeability is a manifestation of cystic fibrosis (CF) in people with CF (pwCF) and in CF mouse models. CF transmembrane conductance regulator knockout (Cftr KO) mouse intestine exhibits increased proliferation and Wnt/β-catenin signaling relative to wild-type mice (WT). Since the Rho GTPase Cdc42 plays a central role in intestinal epithelial proliferation and tight junction remodeling, we hypothesized that Cdc42 may be altered in the Cftr KO crypts. Immunofluorescence showed distinct tight junction localization of Cdc42 in Cftr KO fresh crypts and enteroids, the latter indicating an epithelial-autonomous feature. Quantitative PCR and immunoblots revealed similar expression of Cdc42 in the Cftr KO crypts/enteroids relative to WT, whereas pulldown assays showed increased GTP-bound (active) Cdc42 in proportion to total Cdc42 in Cftr KO enteroids. Cdc42 activity in the Cftr KO and WT enteroids could be reduced by inhibition of the Wnt transducer Disheveled. With the use of a dye permeability assay, Cftr KO enteroids exhibited increased paracellular permeability to 3 kDa dextran relative to WT. Leak permeability and Cdc42 tight junction localization were reduced to a greater extent by inhibition of Wnt/β-catenin signaling with endo-IWR1 in Cftr KO relative to WT enteroids. Increased proliferation or inhibition of Cdc42 activity with ML141 in WT enteroids had no effect on permeability. In contrast, inhibition of Cdc42 with ML141 increased permeability to both 3 kDa dextran and tight junction impermeant 500 kDa dextran in Cftr KO enteroids. These data suggest that increased constitutive Cdc42 activity may alter the stability of paracellular permeability in Cftr KO crypt epithelium.NEW & NOTEWORTHY Increased tight junction localization and GTP-bound activity of the Rho GTPase Cdc42 was identified in small intestinal crypts and enteroids of cystic fibrosis (CF) transmembrane conductance regulator knockout (Cftr KO) mice. The increase in epithelial Cdc42 activity was associated with increased Wnt signaling. Paracellular flux of an uncharged solute (3 kDa dextran) in Cftr KO enteroids indicated a moderate leak permeability under basal conditions that was strongly exacerbated by Cdc42 inhibition. These findings suggest increased activity of Cdc42 in the Cftr KO intestine underlies alterations in intestinal permeability.
Collapse
Affiliation(s)
- Rowena A Woode
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Ashlee M Strubberg
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Jinghua Liu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Nancy M Walker
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Lane L Clarke
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
3
|
Roca M, Masip E, Colombo C, Boon M, Hulst JM, Garriga M, de Koning BAE, Bulfamante A, de Boeck K, Ribes-Koninckx C, Calvo-Lerma J. Long-term evaluation of faecal calprotectin levels in a European cohort of children with cystic fibrosis. Arch Dis Child 2024; 109:552-556. [PMID: 38123920 DOI: 10.1136/archdischild-2023-326221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Intestinal inflammation with contradictory data on faecal calprotectin (fCP) levels is documented in patients with cystic fibrosis (CF). The aim of this study was to longitudinally evaluate fCP in a cohort of children with CF and their relationship with clinical variables. DESIGN Prospective observational study to assess evolution of fCP levels, primary aimed at improving fat absorption. Along 1.5 years of follow-up (November 2016-May 2018) with four study visits pertaining to a pilot study (two of four) and to a clinical trial (two of four), the study outcomes were measured. SETTING Six European CF centres in the context of MyCyFAPP Project. SUBJECTS Children with CF and pancreatic insufficiency (2-18 years old). MAIN OUTCOME MEASUREMENTS fCP levels, pulmonary function (percentage of forced expiratory volume in 1 s (FEV1%)) and coefficient of fat absorption (CFA). Additionally, in the last two visits, gastrointestinal (GI) symptoms were evaluated through the PedsQL-GI Questionnaire. Linear mixed regression models were applied to assess association between fCP and FEV1, CFA and GI symptoms. RESULTS Twenty-nine children with CF and pancreatic insufficiency were included. fCP levels were inversely associated with total modified specific PedsQL-GI score (p=0.04) and positively associated with diarrhoea (p=0.03), but not with CFA. Along the four study visits, fCP significantly increased (from 62 to 256 µg/g) and pulmonary function decreased (from 97% to 87%), with a significant inverse association between the two study outcomes (p<0.001). CONCLUSIONS In children with CF, fCP levels are inversely associated with pulmonary function and thus the specificity of fCP as a marker of intestinal inflammation in paediatric patients with CF warrants further investigation.
Collapse
Affiliation(s)
- Maria Roca
- Health Research Institute La Fe, Valencia, Spain
| | - Etna Masip
- Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Carla Colombo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mieke Boon
- Department of Pediatrics, KU Leuven Hospital, Leuven, Belgium
| | - Jessie M Hulst
- Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, Ontario, Canada
- Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Anna Bulfamante
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Kris de Boeck
- Department of Pediatrics, KU Leuven Hospital, Leuven, Belgium
| | | | - Joaquim Calvo-Lerma
- Health Research Institute La Fe, Valencia, Spain
- Universitat de València, Valencia, Spain
| |
Collapse
|
4
|
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
|
5
|
Chadwick C, Lehman H, Luebbert S, Abdul-Aziz R, Borowitz D. Autoimmunity in people with cystic fibrosis. J Cyst Fibros 2023; 22:969-979. [PMID: 36966037 DOI: 10.1016/j.jcf.2023.03.007] [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: 01/02/2023] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/27/2023]
Abstract
Cystic fibrosis (CF) clinicians may see patients who have difficult-to-manage symptoms that do not have a clear CF-related etiology, such as unusual gastrointestinal (GI) complaints, vasculitis, or arthritis. Alterations in immunity, inflammation and intraluminal dysbiosis create a milieu that may lead to autoimmunity, and the CF transmembrane regulator protein may have a direct role as well. While autoantibodies and other autoimmune markers may develop, these may or may not lead to organ involvement, therefore they are helpful but not sufficient to establish an autoimmune diagnosis. Autoimmune involvement of the GI tract is the best-established association. Next steps to understand autoimmunity in CF should include a more in-depth assessment of the community perspective on its impact. In addition, bringing together specialists in various fields including, but not limited to, pulmonology, gastroenterology, immunology, and rheumatology, would lead to cross-dissemination and help define the path forward in basic science and clinical practice.
Collapse
Affiliation(s)
| | - Heather Lehman
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | | | - Rabheh Abdul-Aziz
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Drucy Borowitz
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
6
|
Gaschignard M, Beaufils F, Gallet P, Clouzeau H, Menard J, Costanzo A, Nouard L, Delhaes L, Tetard C, Lamireau T, Fayon M, Bui S, Enaud R. Dietary intake assessment in children with cystic fibrosis using 3-day food diaries: a single-centre study. Front Pediatr 2023; 11:1130792. [PMID: 37228437 PMCID: PMC10203471 DOI: 10.3389/fped.2023.1130792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Background Malnutrition is both a feature and major cause of morbidity in cystic fibrosis (CF). Therefore, nutritional management is an essential element of patient care. In 2016, an international guideline for nutritional management in patients with CF was published. In light of these recommendations, the aim of this study was to investigate the dietary intake of children with CF at the University Hospital of Bordeaux. Methods We conducted a retrospective study at the Paediatric CF Centre of the University Hospital of Bordeaux. Patients aged 2-18 years with CF who completed a 3-day food diary at home between January 2015 and December 2020 were included. Results A total of 130 patients, with a median age of 11.8 [interquartile range (IQR): 8.3; 13.4] years, were included. The median Z-score for BMI was -0.35 (IQR: -0.9; 0.2) and 20% of the patients had a Z-score for BMI < -1. Recommended total energy intakes were achieved in 53% of the patients, particularly those with nutritional support. Recommended protein intake was met in 28% of the cases, while fat and carbohydrate intakes were met in 54%. Vitamin and micronutrient levels were normal in 80% of the patients, with the exception of vitamin K, which was within the therapeutic range in only 42% of the cases. Conclusion Recommended nutritional targets are difficult to achieve in patients with CF, and providing nutritional support during follow-up remains a challenge.
Collapse
Affiliation(s)
- Margaux Gaschignard
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Fabien Beaufils
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux University, INSERM U1045, U1045, Bordeaux, France
| | - Pauline Gallet
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Haude Clouzeau
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Joris Menard
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Aurélie Costanzo
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Lucie Nouard
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Laurence Delhaes
- Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux University, INSERM U1045, U1045, Bordeaux, France
| | - Candice Tetard
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Thierry Lamireau
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Michael Fayon
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux University, INSERM U1045, U1045, Bordeaux, France
| | - Stéphanie Bui
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux University, INSERM U1045, U1045, Bordeaux, France
| | - Raphaël Enaud
- Pediatric Cystic Fibrosis Reference Center (CRCM), Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux University, INSERM U1045, U1045, Bordeaux, France
| |
Collapse
|
7
|
Schwarzenberg SJ, Vu PT, Skalland M, Hoffman LR, Pope C, Gelfond D, Narkewicz MR, Nichols DP, Heltshe SL, Donaldson SH, Frederick CA, Kelly A, Pittman JE, Ratjen F, Rosenfeld M, Sagel SD, Solomon GM, Stalvey MS, Clancy JP, Rowe SM, Freedman SD. Elexacaftor/tezacaftor/ivacaftor and gastrointestinal outcomes in cystic fibrosis: Report of promise-GI. J Cyst Fibros 2023; 22:282-289. [PMID: 36280527 PMCID: PMC10144072 DOI: 10.1016/j.jcf.2022.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/12/2022] [Accepted: 10/10/2022] [Indexed: 02/19/2023]
Abstract
BACKGROUND Elexacaftor/tezacaftor/ivacaftor (ETI) improves pulmonary disease in people with cystic fibrosis (PwCF), but its effect on gastrointestinal symptoms, which also affect quality of life, is not clear. METHODS PROMISE is a 56-center prospective, observational study of ETI in PwCF >12 years and at least one F508del allele. Gastrointestinal symptoms, evaluated by validated questionnaires: Patient Assessment of Upper Gastrointestinal Disorders-Symptom (PAGI-SYM), Patient Assessment of Constipation-Symptom (PAC-SYM), Patient Assessment of Constipation-Quality of Life (PAC-QOL)), fecal calprotectin, steatocrit and elastase-1 were measured before and 6 months after ETI initiation. Mean difference and 95% confidence intervals were obtained from linear regression with adjustment for age and sex. RESULTS 438 participants fully completed at least 1 questionnaire. Mean (SD) for baseline PAGI-SYM, PAC-SYM, and PAC-QOL total scores were 0.56 (0.59), 0.47 (0.45), and 0.69 (0.53) out of maximum 5, 4, and 5, respectively (higher score indicates greater severity). Corresponding age- and sex-adjusted 6 months mean changes (95% CI) in total scores were -0.15 (-0.21, -0.09) for PAGI-SYM, -0.14 (-0.19, -0.09) for PAC-SYM, and -0.15 (-0.21, -0.10) for PAC-QOL. While statistically significant, changes were small and unlikely to be of clinical importance. Fecal calprotectin showed a change (95% CI) from baseline of -66.2 µg/g (-86.1, -46.2) at 6 months, while fecal elastase and steatocrit did not meaningfully change. CONCLUSIONS After 6 months of ETI, fecal markers of inflammation decreased. Gastrointestinal symptoms improved, but the effect size was small. Pancreatic insufficiency did not improve.
Collapse
Affiliation(s)
- Sarah Jane Schwarzenberg
- Department of Pediatrics, University of Minnesota Masonic Children's Hospital, Academic Office Building, 2450 Riverside Ave S AO-201, Minneapolis, MN 55454, USA.
| | - Phuong T Vu
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, WA, USA
| | - Michelle Skalland
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucas R Hoffman
- Department of Pediatrics and Department of Microbiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Christopher Pope
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Michael R Narkewicz
- Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, USA
| | - David P Nichols
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Sonya L Heltshe
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Scott H Donaldson
- Department of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Carla A Frederick
- Jacobs School of Medicine and Biomedical Sciences of the University of Buffalo, NY, USA
| | - Andrea Kelly
- Division of Endocrinology & Diabetes, Children's Hospital of Philadelphia; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica E Pittman
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Felix Ratjen
- Division of Respiratory Medicine, Department of Pediatrics, Translational Medicine, Hospital for Sick Children, University of Toronto, Canada
| | - Margaret Rosenfeld
- Department of Epidemiology and Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Scott D Sagel
- Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, USA
| | - George M Solomon
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael S Stalvey
- Department of Pediatrics and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Steven M Rowe
- Department of Medicine and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Steven D Freedman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Mariotti Zani E, Grandinetti R, Cunico D, Torelli L, Fainardi V, Pisi G, Esposito S. Nutritional Care in Children with Cystic Fibrosis. Nutrients 2023; 15:nu15030479. [PMID: 36771186 PMCID: PMC9921127 DOI: 10.3390/nu15030479] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
Patients with cystic fibrosis (CF) are prone to malnutrition and growth failure, mostly due to malabsorption caused by the derangement in the chloride transport across epithelial surfaces. Thus, optimal nutritional care and support should be an integral part of the management of the disease, with the aim of ameliorating clinical outcomes and life expectancy. In this report, we analyzed the nutrition support across the different ages, in patients with CF, with a focus on the relationships with growth, nutritional status, disease outcomes and the use of the CF transmembrane conductance regulator (CFTR) modulators. The nutrition support goal in CF care should begin as early as possible after diagnosis and include the achievement of an optimal nutritional status to support the growth stages and puberty development in children, that will further support the maintenance of an optimal nutritional status in adult life. The cornerstone of nutrition in patients with CF is a high calorie, high-fat diet, in conjunction with a better control of malabsorption due to pancreatic enzyme replacement therapy, and attention to the adequate supplementation of fat-soluble vitamins. When the oral caloric intake is not enough for reaching the anthropometric nutritional goals, supplemental enteral feeding should be initiated to improve growth and the nutritional status. In the last decade, the therapeutic possibilities towards CF have grown in a consistent way. The positive effects of CFTR modulators on nutritional status mainly consist in the improvement in weight gain and BMI, both in children and adults, and in an amelioration in terms of the pulmonary function and reduction of exacerbations. Several challenges need to be overcome with the development of new drugs, to transform CF from a fatal disease to a treatable chronic disease with specialized multidisciplinary care.
Collapse
Affiliation(s)
- Elena Mariotti Zani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Roberto Grandinetti
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Daniela Cunico
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Lisa Torelli
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Valentina Fainardi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giovanna Pisi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Correspondence: ; Tel.: +39-0521-903524
| |
Collapse
|
9
|
Freedman SD, Zaworski K, Pierzynowska K, Pierzynowski S, Gallotto R, Sathe M, Borowitz DS. Validation of an omega-3 substrate challenge absorption test as an indicator of global fat lipolysis. PLoS One 2023; 18:e0284651. [PMID: 37155649 PMCID: PMC10166528 DOI: 10.1371/journal.pone.0284651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
INTRODUCTION The coefficient of fat absorption (CFA) quantifies fat that remains in stool after digestion and is not a direct measure of lipolysis. CFA has been used to assess treatment of pancreatic insufficiency but does not correlate with pancreatic enzyme replacement therapy dose. We explored use of an omega-3 substrate absorption challenge test as a sensitive test of lipolysis and absorption. METHODS We studied a novel microbially-derived lipase (SNSP003) employing an established surgical model commonly used to study the uptake of macronutrients, the exocrine pancreatic insufficient pig. Pigs were fed a high-fat diet and given a standardized omega-3 substrate challenge to test the effect of lipolysis on its absorption. Blood was drawn at 0, 1, 2, 4, 6, 8, 12, and 24 hours following the substrate challenge and was analyzed for omega-3 and total fat levels (c14:c24). SNSP003 was also compard to porcine pancrelipase. RESULTS The absorption of omega-3 fats was significantly increased following administration of 40, 80 and 120 mg SNSP003 lipase by 51% (p = 0.02), 89%, (p = 0.001) and 64% (p = 0.01), respectively, compared to that observed when no lipase was administered to the pigs, with Tmax at 4 hours. The two highest SNSP003 doses were compared to porcine pancrelipase and no significant differences were observed. Both doses increased plasma total fatty acids (141% for the 80 mg dose (p = 0.001) and 133% for the 120 mg dose (p = 0.006), compared to no lipase) and no significant differences were observed between the SNSP003 lipase doses and porcine pancrelipase. CONCLUSION The omega-3 substrate absorption challenge test differentiates among different doses of a novel microbially-derived lipase and correlates with global fat lipolysis and absorption in exocrine pancreatic insufficient pigs. No significant differences were observed between the two highest novel lipase doses and porcine pancrelipase. Studies in humans should be designed to support the evidence presented here that suggests the omega-3 substrate absorption challenge test has advantages over the coefficient of fat absorption test to study lipase activity.
Collapse
Affiliation(s)
- Steven D Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Kamil Zaworski
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Kateryna Pierzynowska
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
- Department of Biology, Lund University, Lund, Sweden
- Synspira Therapeutics, Inc., Framingham, MA, United States of America
| | - Stefan Pierzynowski
- Department of Biology, Lund University, Lund, Sweden
- Anara AB, Trelleborg, Sweden
- Department of Medical Biology, Institute of Rural Health, Lublin, Poland
| | - Robert Gallotto
- Synspira Therapeutics, Inc., Framingham, MA, United States of America
| | - Meghana Sathe
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Texas Southwestern/Children's Health, Dallas, TX, United States of America
| | - Drucy S Borowitz
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States of America
| |
Collapse
|
10
|
Gaschignard M, Beaufils F, Lussac-Sorton F, Gallet P, Clouzeau H, Menard J, Costanzo A, Nouard L, Delhaes L, Tetard C, Lamireau T, Fayon M, Bui S, Enaud R. Nutritional impact of CFTR modulators in children with cystic fibrosis. Front Pediatr 2023; 11:1130790. [PMID: 37063653 PMCID: PMC10091219 DOI: 10.3389/fped.2023.1130790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/06/2023] [Indexed: 04/18/2023] Open
Abstract
Background Nutritional status is a major prognostic factor for breathing and the survival of patients with cystic fibrosis (CF). Since 2012, the development of CFTR modulators has considerably transformed the outcome of this disease. Indeed, both lung function and body mass index are improved by CFTR modulators, such as Lumacaftor/Ivacaftor. However, few data exist regarding the outcome of nutritional intakes under Lumacaftor/Ivacaftor. Methods We conducted a prospective single-center study in children with CF treated with Lumacaftor/Ivacaftor to evaluate their nutritional intake before and after treatment. Results Thirty-four children were included in this study, with a median age of 12.4 years [11.9; 14.7]. There was no significant improvement in weight, height or BMI. Patients' total energy intake was not significantly changed with Lumacaftor/Ivacaftor, while carbohydrate intakes decreased significantly. We found that blood levels of vitamin E and Selenium were significantly increased under Lumacaftor/Ivacaftor, without a significant increase in supplementation. In patients with a BMI Z-score < 0 at treatment initiation, there was a significant improvement in weight and BMI Z-score, while TEI and carbohydrate intakes were significantly lower. Conclusion We showed that treatment with Lumacaftor/Ivacaftor improved the nutritional status of patients without necessarily being associated with an increase in nutritional intake. Although these data need to be confirmed in larger cohorts, they support the hypothesis that weight gain under modulators is multifactorial, and may be related to a decrease in energy expenditure or an improvement in absorption.
Collapse
Affiliation(s)
- Margaux Gaschignard
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Fabien Beaufils
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, Bordeaux, France
| | - Florian Lussac-Sorton
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, Bordeaux, France
| | - Pauline Gallet
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Haude Clouzeau
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Joris Menard
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Aurélie Costanzo
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Lucie Nouard
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Laurence Delhaes
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, Bordeaux, France
| | - Candice Tetard
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Thierry Lamireau
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
| | - Michael Fayon
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, Bordeaux, France
| | - Stéphanie Bui
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, Bordeaux, France
| | - Raphaël Enaud
- Bordeaux University Hospital, Hôpital Pellegrin-Enfants, Paediatric Cystic Fibrosis Reference Center (CRCM), Centre d'Investigation Clinique (CIC 1401), Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, Bordeaux, France
- Correspondence: Raphaël Enaud
| |
Collapse
|
11
|
Wilson A, Altman K, Schindler T, Schwarzenberg SJ. Updates in Nutrition Management of Cystic Fibrosis in the Highly Effective Modulator Era. Clin Chest Med 2022; 43:727-742. [PMID: 36344077 DOI: 10.1016/j.ccm.2022.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Attainment and maintenance of good nutrition has been an important aspect of management in cystic fibrosis (CF) for decades. In the era of highly effective modulator therapy for CF, the quality of the nutrients we recommend is increasingly important. Our therapy must support our patients' health for many years beyond what we previously thought. Preventing cardiovascular disease, reducing hyperlipidemia, and optimizing lean body mass for active, longer lives now join the long-standing goal of promoting lung function through nutrition. This chapter summarizes recent developments in nutrition in people with CF, with an eye to the evolution of our practice.
Collapse
Affiliation(s)
- Alexandra Wilson
- Cystic Fibrosis Clinical Research, Clinical Research Services, National Jewish Health, 1400 Jackson Street, K333, Denver, CO 80206, USA
| | - Kimberly Altman
- Gunnar Esiason Adult Cystic Fibrosis and Lung Center, Columbia University Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - Terri Schindler
- Pediatric Pulmonology, University Hospitals Cleveland Medical Center, Rainbow Babies and Children's Hospital
| | - Sarah Jane Schwarzenberg
- Department of Pediatrics; University of Minnesota Masonic Children's Hospital, Academic Office Building, 2450 Riverside Avenue South AO-201, Minneapolis, MN 55454, USA.
| |
Collapse
|
12
|
Larriba R, Roca M, Masip E, Cañada-Martínez A, Ribes-Koninckx C, Calvo-Lerma J. How macronutrients and pancreatic enzyme supplements dose variability affect fat, protein and starch absorption in children with cystic fibrosis. Dig Liver Dis 2022; 55:513-518. [PMID: 36435715 DOI: 10.1016/j.dld.2022.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND low evidence on the dose of enzymatic supplements used in pancreatic enzyme replacement therapy (PERT) is available. AIM assessing if fat, protein and starch absorption could be related to the dose of the enzymatic supplement, the intra-patient variability in the dose and macronutrient intake. METHODS Four-day food records and 3-day faecal samples were prospectively collected in 69 children with cystic fibrosis. Pearson correlations between enzyme dose and macronutrient absorption, and beta regression models were applied to explain the results. RESULTS the supply of protease units per protein intake (PU/g protein) in relation to lipase units per fat intake (LU/g fat) was low and the intra-patient variability in the dose of enzymes was ±1331 LU/g fat. Fat and starch absorption was >90% while for protein it was 81.5%. The coefficient of fat absorption was associated with an interaction between the dose of LU/g fat and its variability among different days. Lipid and protein intake were also determinants of the coefficient of fat absorption. CONCLUSION the dose of PERT should be re-adjusted to the amount of dietary fat of every meal (constant LU/g fat) to minimize variability and increase fat absorption. Also, the supply of protease should be increased to prevent from protein malabsorption.
Collapse
Affiliation(s)
- Raúl Larriba
- Celiac Disease and Digestive Immunopathology Research Group, Instituto de Investigación Sanitaria La Fe, 46026, València, Spain
| | - María Roca
- Celiac Disease and Digestive Immunopathology Research Group, Instituto de Investigación Sanitaria La Fe, 46026, València, Spain
| | - Etna Masip
- Celiac Disease and Digestive Immunopathology Research Group, Instituto de Investigación Sanitaria La Fe, 46026, València, Spain
| | - Antonio Cañada-Martínez
- Data Science and Biostatistics Unit, Instituto de Investigación Sanitaria La Fe, 46026, València, Spain
| | - Carmen Ribes-Koninckx
- Celiac Disease and Digestive Immunopathology Research Group, Instituto de Investigación Sanitaria La Fe, 46026, València, Spain
| | - Joaquim Calvo-Lerma
- Celiac Disease and Digestive Immunopathology Research Group, Instituto de Investigación Sanitaria La Fe, 46026, València, Spain; Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
13
|
O’Malley Y, Coleman MC, Sun X, Lei J, Yao J, Pulliam CF, Kluz P, McCormick ML, Yi Y, Imai Y, Engelhardt JF, Norris AW, Spitz DR, Uc A. Oxidative stress and impaired insulin secretion in cystic fibrosis pig pancreas. ADVANCES IN REDOX RESEARCH 2022; 5:100040. [PMID: 35903252 PMCID: PMC9328447 DOI: 10.1016/j.arres.2022.100040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cystic fibrosis-related diabetes (CFRD) is one the most common comorbidities in cystic fibrosis (CF). Pancreatic oxidative stress has been postulated in the pathogenesis of CFRD, but no studies have been done to show an association. The main obstacle is the lack of suitable animal models and no immediate availability of pancreas tissue in humans. In the CF porcine model, we found increased pancreatic total glutathione (GSH), glutathione disulfide (GSSG), 3-nitrotyrosine- and 4-hydroxynonenal-modified proteins, and decreased copper zinc superoxide dismutase (CuZnSOD) activity, all indicative of oxidative stress. CF pig pancreas demonstrated increased DHE oxidation (as a surrogate marker of superoxide) in situ compared to non-CF and this was inhibited by a SOD-mimetic (GC4401). Catalase and glutathione peroxidase activities were not different between CF and non-CF pancreas. Isolated CF pig islets had significantly increased DHE oxidation, peroxide production, reduced insulin secretion in response to high glucose and diminished secretory index compared to non-CF islets. Acute treatment with apocynin or an SOD mimetic failed to restore insulin secretion. These results are consistent with the hypothesis that CF pig pancreas is under significant oxidative stress as a result of increased O2 ●- and peroxides combined with reduced antioxidant defenses against reactive oxygen species (ROS). We speculate that insulin secretory defects in CF may be due to oxidative stress.
Collapse
Affiliation(s)
- Yunxia O’Malley
- Stead Family Department of Pediatrics, The University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
| | - Mitchell C. Coleman
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Junying Lei
- Stead Family Department of Pediatrics, The University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
| | - Jianrong Yao
- Stead Family Department of Pediatrics, The University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
| | - Casey F. Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Paige Kluz
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, Iowa 52242, USA
| | - Michael L. McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Yaling Yi
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Yumi Imai
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Andrew W. Norris
- Stead Family Department of Pediatrics, The University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Aliye Uc
- Stead Family Department of Pediatrics, The University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
14
|
Borowitz D, Aronoff N, Cummings LC, Maqbool A, Mulberg AE. Coefficient of Fat Absorption to Measure the Efficacy of Pancreatic Enzyme Replacement Therapy in People With Cystic Fibrosis: Gold Standard or Coal Standard? Pancreas 2022; 51:310-318. [PMID: 35695742 PMCID: PMC9257055 DOI: 10.1097/mpa.0000000000002016] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/02/2022] [Indexed: 12/10/2022]
Abstract
OBJECTIVES We sought data on the validity, reliability, responsiveness, and feasibility of the coefficient of fat absorption (CFA) as a measure of pancreatic enzyme replacement therapy (PERT) efficacy in people with cystic fibrosis (pwCF) and reviewed the literature for alternative measures. METHODS We searched PubMed for the Medical Subject Heading cystic fibrosis and the key words cystic fibrosis, fat absorption, CFA, and fecal fat imbalance; historical articles; and citations in bibliographies. RESULTS The lower the CFA, the greater its variability; thus, it is less variable in healthy individuals who have higher CFA than pwCF. In addition, the test-retest values for CFA are more variable in pwCF than the general population. There is no correlation between CFA and body mass index or PERT dose but CFA is related to gastrointestinal signs and symptoms. Research-quality CFA studies are expensive, time consuming, and odious to pwCF and research staff. Sparse stool tests, breath tests, and blood tests of fat absorption have been studied as potential alternatives to CFA to measure PERT efficacy. CONCLUSIONS Based on the evidence, we conclude that CFA as a measure of the efficacy of PERT is more of a "coal standard" than a gold standard; developing suitable alternatives should be a priority.
Collapse
Affiliation(s)
- Drucy Borowitz
- From the Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences
| | - Nell Aronoff
- University Libraries, University at Buffalo, Buffalo, NY
| | - Linda C. Cummings
- Department of Internal Medicine, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Asim Maqbool
- Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
15
|
Neri LDCL, Simon MISDS, Ambrósio VLS, Barbosa E, Garcia MF, Mauri JF, Guirau RR, Neves MA, Cunha CDAP, Nogueira MC, Alves ACDC, Gurmini J, Servidoni MDF, Epifanio M, Athanazio R. Brazilian Guidelines for Nutrition in Cystic Fibrosis. EINSTEIN-SAO PAULO 2022; 20:eRW5686. [PMID: 35384985 PMCID: PMC8967313 DOI: 10.31744/einstein_journal/2022rw5686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 01/12/2021] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To develop a scientific consensus on nutrition in cystic fibrosis. METHODS Sixteen coordinators elaborated relevant questions on nutritional therapy in cystic fibrosis, which were divided into six sections: nutritional assessment, nutritional recommendations, nutritional intervention, dietary counseling, special situations and enzyme replacement, and gastrointestinal manifestations. Two to three specialists in the field were responsible for each section and obtaining answers formulated based on standardized bibliographic searches. The available literature was searched in the PubMed®/MEDLINE database, after training and standardization of search strategies, to write the best level of evidence for the questions elaborated. Issues related to disagreement were discussed until a consensus was reached among specialists, based on the current scientific literature. RESULTS Forty-two questions were prepared and objectively answered, resulting in a consensus of nutritional therapy in cystic fibrosis. CONCLUSION This work enabled establishing a scientific consensus for nutritional treatment of cystic fibrosis patients.
Collapse
Affiliation(s)
- Lenycia de Cassya Lopes Neri
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrazil Instituto da Criança, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | | | - Valéria Laguna Salomão Ambrósio
- Hospital das ClínicasFaculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSPBrazilHospital das Clínicas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Eliana Barbosa
- Hospital Infantil Joana de GusmãoFlorianópolisSCBrazilHospital Infantil Joana de Gusmão, Florianópolis, SC, Brazil.
| | - Monique Ferreira Garcia
- Hospital Infantil Joana de GusmãoFlorianópolisSCBrazilHospital Infantil Joana de Gusmão, Florianópolis, SC, Brazil.
| | - Juliana Ferreira Mauri
- Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloSPBrazilEscola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Renata Rodrigues Guirau
- Universidade Estadual de CampinasCampinasSPBrazilUniversidade Estadual de Campinas, Campinas, SP, Brazil.
| | | | | | - Marcelo Coelho Nogueira
- Hospital Infantil João Paulo IIBelo HorizonteMGBrazilHospital Infantil João Paulo II, Belo Horizonte, MG, Brazil.
| | - Anna Carolina Di Creddo Alves
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrazil Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Jocemara Gurmini
- Hospital Pequeno PríncipeCuritibaPRBrazilHospital Pequeno Príncipe, Curitiba, PR, Brazil.
| | - Maria de Fatima Servidoni
- Universidade Estadual de CampinasCampinasSPBrazilUniversidade Estadual de Campinas, Campinas, SP, Brazil.
| | - Matias Epifanio
- Pontifícia Universidade Católica do Rio Grande do SulPorto AlegreRSBrazilPontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Rodrigo Athanazio
- Hospital das ClínicasFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrazil Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
16
|
Freswick PN, Reid EK, Mascarenhas MR. Pancreatic Enzyme Replacement Therapy in Cystic Fibrosis. Nutrients 2022; 14:nu14071341. [PMID: 35405954 PMCID: PMC9003370 DOI: 10.3390/nu14071341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
While typically considered a pulmonary disease, cystic fibrosis patients develop significant nutritional complications and comorbidities, especially those who are pancreatic insufficient. Clinicians must have a high suspicion for cystic fibrosis among patients with clinical symptoms of pancreatic insufficiency, and pancreatic enzymatic replacement therapy (PERT) must be urgently initiated. PERT presents a myriad of considerations for patients and their supporting dieticians and clinicians, including types of administration, therapy failures, and complications.
Collapse
Affiliation(s)
- Peter N. Freswick
- Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA
- Correspondence:
| | - Elizabeth K. Reid
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (E.K.R.); (M.R.M.)
| | - Maria R. Mascarenhas
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (E.K.R.); (M.R.M.)
| |
Collapse
|
17
|
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
|
18
|
Drzymała-Czyż S, Dziedzic K, Szwengiel A, Krzyżanowska-Jankowska P, Nowak JK, Nowicka A, Aringazina R, Drzymała S, Kashirskaya N, Walkowiak J. Serum bile acids in cystic fibrosis patients - glycodeoxycholic acid as a potential marker of liver disease. Dig Liver Dis 2022; 54:111-117. [PMID: 34305015 DOI: 10.1016/j.dld.2021.06.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) and CF-related liver disease can lead to disturbances in bile acid metabolism. AIM This study determined serum bile acid concentrations in CF to define their usefulness in liver disease assessment. METHODS Primary, secondary and conjugated bile acid levels were measured in three CF groups (25 patients each) exhibiting: liver cirrhosis, other liver disease, no liver disease, and in 25 healthy subjects (HS). RESULTS Bile acid levels were higher in CF patients than in HS, except for glycodeoxycholic acid (GDCA). However, bile acid concentrations did not differ between patients with cirrhosis and other liver involvement. GDCA and deoxycholic acid (DCA) differentiated CF patients with non-cirrhotic liver disease from those without liver disease (GDCA-AUC: 0.924, 95%CI 0.822-1.000, p<0.001; DCA-AUC: 0.867, 95%CI: 0.731-1.000, p<0.001). Principal component analysis revealed that in CF liver disease was related to GDCA, GGTP activity, severe genotype and pancreatic insufficiency. CONCLUSIONS A CF-specific bile acid profile was defined and shown to relate to liver disease. GDCA differentiates patients with non-cirrhotic liver involvement from those with no detectable liver disease. Hence, GDCA is a candidate for validation as a biomarker of non-cirrhotic progression of liver disease in CF.
Collapse
Affiliation(s)
- Sławomira Drzymała-Czyż
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland; Department of Bromatology, Poznan University of Medical Sciences, Poznań, Poland
| | - Krzysztof Dziedzic
- Department of Food Science and Nutrition, Institute of Food Technology of Plant Origin, Poznań University of Life Sciences, Poznań, Poland
| | - Artur Szwengiel
- Department of Food Science and Nutrition, Institute of Food Technology of Plant Origin, Poznań University of Life Sciences, Poznań, Poland
| | | | - Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Agata Nowicka
- Department of Pulmonology, Allergology and Respiratory Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Raisa Aringazina
- Department of Internal Diseases No1, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Sylwia Drzymała
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland
| | - Nataliya Kashirskaya
- Department of Genetic Epidemiology (Cystic Fibrosis Group), Research Centre for Medical Genetics, Moscow, Russia
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznań, Poland.
| |
Collapse
|
19
|
Imrei M, Németh D, Szakács Z, Hegyi P, Kiss S, Alizadeh H, Dembrovszky F, Pázmány P, Bajor J, Párniczky A. Increased Prevalence of Celiac Disease in Patients with Cystic Fibrosis: A Systematic Review and Meta-Analysis. J Pers Med 2021; 11:859. [PMID: 34575636 PMCID: PMC8470465 DOI: 10.3390/jpm11090859] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Immune regulation seems to be altered in cystic fibrosis (CF), thus potentially predisposing patients to developing autoimmune diseases (AID). In this meta-analysis, we aimed to evaluate the prevalence of celiac disease (CeD) among CF patients as by far the most commonly reported autoimmune disease in this population and, secondly, to review the observations on other, less frequently studied autoimmune diseases. METHODS We conducted a systematic literature search for studies that discussed AIDs among CF patients. Following standard selection and data collection, we calculated pooled raw prevalence with 95% confidence intervals (CI) for biopsy-verified CeD and seropositivity. RESULTS Out of the 21 eligible studies, 15 reported on CeD. Pooled prevalence of biopsy-verified CeD was 1.8% (CI 1.1-2.7%) according to a homogeneous dataset from six prospective, consecutive screening studies, while it proved to be 2.3% (CI 1.1-4.7%) according to a heterogeneous dataset from the other studies. Tissue transglutaminase IgA positivity was detected in 4.5% of CF cases (CI 2.8-6.9%), while tissue transglutaminase IgA-endomysial antibody IgA double positivity was found in 2.4% of them (CI 1.5-3.9%). Findings on other AIDs were strongly limited. CONCLUSIONS The pooled prevalence of CeD in CF seemed to be more than twice as high compared to the global prevalence; therefore, routine screening of CeD could be considered in CF.
Collapse
Affiliation(s)
- Marcell Imrei
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Dávid Németh
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Zsolt Szakács
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
- Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary;
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
- Centre for Translational Medicine, Department of Medicine, University of Szeged, Tisza Lajos krt. 109., H-6725 Szeged, Hungary
| | - Szabolcs Kiss
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Tisza Lajos krt. 109., H-6725 Szeged, Hungary
| | - Hussain Alizadeh
- Division of Hematology, First Department of Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary;
| | - Fanni Dembrovszky
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Piroska Pázmány
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
| | - Judit Bajor
- Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary;
| | - Andrea Párniczky
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary; (M.I.); (D.N.); (Z.S.); (P.H.); (S.K.); (F.D.); (P.P.)
- János Szentágothai Research Centre, University of Pécs, Szigeti út 12., H-7624 Pécs, Hungary
- Heim Pál National Pediatric Institute, H-1089 Budapest, Hungary
| |
Collapse
|
20
|
Calvo-Lerma J, Boon M, Colombo C, de Koning B, Asseiceira I, Garriga M, Roca M, Claes I, Bulfamante A, Walet S, Pereira L, Ruperto M, Masip E, Asensio-Grau A, Giana A, Affourtit P, Heredia A, Vicente S, Andrés A, de Boeck K, Hulst J, Ribes-Koninckx C. Clinical evaluation of an evidence-based method based on food characteristics to adjust pancreatic enzyme supplements dose in cystic fibrosis. J Cyst Fibros 2020; 20:e33-e39. [PMID: 33279468 DOI: 10.1016/j.jcf.2020.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Patients with cystic fibrosis (CF) and pancreatic insufficiency need pancreatic enzyme replacement therapy (PERT) for dietary lipids digestion. There is limited evidence for recommending the adequate PERT dose for every meal, and controlling steatorrhea remains a challenge. This study aimed to evaluate a new PERT dosing method supported by a self-management mobile-app. METHODS Children with CF recruited from 6 European centres were instructed to use the app, including an algorithm for optimal PERT dosing based on in vitro digestion studies for every type of food. At baseline, a 24h self-selected diet was registered in the app, and usual PERT doses were taken by the patient. After 1 month, the same diet was followed, but PERT doses were indicated by the app. Change in faecal fat and coefficient of fat absorption (CFA) were determined. RESULTS 58 patients (median age 8.1 years) participated. Baseline fat absorption was high: median CFA 96.9%, median 2.4g faecal fat). After intervention CFA did not significantly change, but range of PERT doses was reduced: interquartile ranges narrowing from 1447-3070 at baseline to 1783-2495 LU/g fat when using the app. Patients with a low baseline fat absorption (CFA<90%, n=12) experienced significant improvement in CFA after adhering to the recommended PERT dose (from 86.3 to 94.0%, p=0.031). CONCLUSION the use of a novel evidence-based PERT dosing method, based on in vitro fat digestion studies incorporating food characteristics, was effective in increasing CFA in patients with poor baseline fat absorption and could safely be implemented in clinical practice.
Collapse
Affiliation(s)
- Joaquim Calvo-Lerma
- Instituto de Investigación Sanitaria La Fe de Valencia. Cystic Fibrosis Unit. 46026 Valencia, Spain; Universitat Politècnica de València, Research Institute of Food Engineering for Development. 46022 Valencia, Spain.
| | - Mieke Boon
- Department of pediatrics, Center for Cystic Fibrosis, University Hospital Leuven. Leuven, Belgium
| | - Carla Colombo
- CF Center, Università degli Studi di Milano. Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico. 20122 Milan, Italy
| | - Barbara de Koning
- Department of Pediatrics, div of Gastro-Enterology and div of Respiratory Medicine and Allergology, Erasmus MC- Sophia Children's Hospital, University Hospital Rotterdam. Rotterdam, the Netherlands
| | - Inês Asseiceira
- Centro de Fibrose Quística, Hospital de Santa Maria, Lisboa. Lisbon, Portugal
| | - Maria Garriga
- Hospital Universitario Ramón y Cajal. Cystic Fibrosis Unit. 28034 Madrid, Spain
| | - Maria Roca
- Instituto de Investigación Sanitaria La Fe de Valencia. Cystic Fibrosis Unit. 46026 Valencia, Spain
| | - Ine Claes
- Department of pediatrics, Center for Cystic Fibrosis, University Hospital Leuven. Leuven, Belgium
| | - Anna Bulfamante
- CF Center, Università degli Studi di Milano. Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico. 20122 Milan, Italy
| | - Sylvia Walet
- Department of Pediatrics, div of Gastro-Enterology and div of Respiratory Medicine and Allergology, Erasmus MC- Sophia Children's Hospital, University Hospital Rotterdam. Rotterdam, the Netherlands
| | - Luisa Pereira
- Centro de Fibrose Quística, Hospital de Santa Maria, Lisboa. Lisbon, Portugal
| | - Mar Ruperto
- Hospital Universitario Ramón y Cajal. Cystic Fibrosis Unit. 28034 Madrid, Spain
| | - Etna Masip
- Instituto de Investigación Sanitaria La Fe de Valencia. Cystic Fibrosis Unit. 46026 Valencia, Spain
| | - Andrea Asensio-Grau
- Universitat Politècnica de València, Research Institute of Food Engineering for Development. 46022 Valencia, Spain
| | - Arianna Giana
- CF Center, Università degli Studi di Milano. Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico. 20122 Milan, Italy
| | - Philine Affourtit
- Department of Pediatrics, div of Gastro-Enterology and div of Respiratory Medicine and Allergology, Erasmus MC- Sophia Children's Hospital, University Hospital Rotterdam. Rotterdam, the Netherlands
| | - Ana Heredia
- Universitat Politècnica de València, Research Institute of Food Engineering for Development. 46022 Valencia, Spain
| | - Saioa Vicente
- Hospital Universitario Ramón y Cajal. Cystic Fibrosis Unit. 28034 Madrid, Spain
| | - Ana Andrés
- Universitat Politècnica de València, Research Institute of Food Engineering for Development. 46022 Valencia, Spain
| | - Kris de Boeck
- Department of pediatrics, Center for Cystic Fibrosis, University Hospital Leuven. Leuven, Belgium
| | - Jessie Hulst
- Department of Pediatrics, div of Gastro-Enterology and div of Respiratory Medicine and Allergology, Erasmus MC- Sophia Children's Hospital, University Hospital Rotterdam. Rotterdam, the Netherlands; Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Canada
| | - Carmen Ribes-Koninckx
- Instituto de Investigación Sanitaria La Fe de Valencia. Cystic Fibrosis Unit. 46026 Valencia, Spain
| |
Collapse
|
21
|
Oral Glutathione and Growth in Cystic Fibrosis: A Multicenter, Randomized, Placebo-controlled, Double-blind Trial. J Pediatr Gastroenterol Nutr 2020; 71:771-777. [PMID: 32960827 PMCID: PMC8220910 DOI: 10.1097/mpg.0000000000002948] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The nutritional status of children with cystic fibrosis (CF) is associated with mortality and morbidity. Intestinal inflammation may contribute to impaired digestion, absorption, and nutrient utilization in patients with CF and oral glutathione may reduce inflammation, promoting improved nutritional status in patients with CF. METHODS The GROW study was a prospective, multicenter, randomized, placebo-controlled, double-blind, phase II clinical trial in pancreatic insufficient patients with CF between the ages of 2 and 10 years. Patients received reduced glutathione or placebo orally daily for 24 weeks. The primary endpoint was the difference in change in weight-for-age z-scores from baseline through week 24 between treatment groups. Secondary endpoints included other anthropometrics, serum, and fecal inflammatory markers in addition to other clinical outcomes. RESULTS Fifty-eight participants completed the study. No significant differences were seen between glutathione (n = 30) and placebo (n = 28) groups in the 6-month change in weight-for-age z-score (-0.08; 95% CI: -0.22 to 0.06; P = 0.25); absolute change in weight (kg) (-0.18; 95% CI: -0.55 to 0.20; P = 0.35); or absolute change in BMI kg/m (-0.06; 95% CI: -0.37 to 0.25; P = 0.69). There were no significant differences in other secondary endpoints. Overall, glutathione was safe and well tolerated. CONCLUSIONS Oral glutathione supplementation did not impact growth or change serum or fecal inflammatory markers in pancreatic insufficient children with CF when compared with placebo.
Collapse
|
22
|
Measures of Dietary Fat and Energy Absorption in Healthy Adults. Pancreas 2020; 49:845-854. [PMID: 32590620 PMCID: PMC8335406 DOI: 10.1097/mpa.0000000000001587] [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: 12/24/2022]
Abstract
OBJECTIVES Existing reference ranges for stool fat and energy absorption were developed using subjects in controlled environments on precise diets. This study measured energy and fat absorption in healthy, community-dwelling adults eating a moderate-to-high fat American diet via stool- and serum-based methods. METHODS This was a secondary analysis of healthy subjects recruited as the comparison group in a chronic pancreatitis study. Subjects recorded dietary intake and collected stool over 3-day periods. Stool was analyzed for fat content using the coefficient of fat absorption and for energy content using bomb calorimetry. The malabsorption blood test (MBT) was used to measure dietary fat absorption. RESULTS Nineteen subjects had mean daily stool measures of 143 g wet weight, 4.1 g of fat, and 178 kcal. The mean coefficients of fat and energy absorption were 96% and 93%, respectively. The mean MBT area under the curve cut-point was greater than 8 mg·h/dL. CONCLUSIONS This study confirms the historical reference range for the coefficient of fat absorption in contemporary healthy, community-dwelling adults on a moderate-to-high fat diet. The study contributes to the development of reference range values for multiple bomb calorimetry-based outcomes of stool energy losses and to the serum-based MBT as a promising method for measuring fat absorption.
Collapse
|
23
|
Pancreatic Function in Chronic Pancreatitis: A Cohort Study Comparing 3 Methods of Detecting Fat Malabsorption and the Impact of Short-term Pancreatic Enzyme Replacement Therapy. Pancreas 2019; 48:1068-1078. [PMID: 31404029 PMCID: PMC7243202 DOI: 10.1097/mpa.0000000000001381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Reliable pancreatic function tests in patients with chronic pancreatitis (CP) are needed. This cohort study identified malabsorption in people with CP compared with healthy people and then investigated short-term pancreatic enzyme replacement therapy (PERT) and fat malabsorption, nutritional status, and quality of life (QOL). METHODS Subjects with CP were evaluated before and after PERT and compared with the healthy cohort using coefficient of fat absorption (CFA), stool bomb calorimetry, and the malabsorption blood test (MBT). Anthropometrics, micronutrients, and QOL data were collected. Group means at baseline and after PERT were analyzed. RESULTS The 24 subjects with CP had greater stool energy loss (5668 cal/g [standard deviation {SD}, 753] vs 5152 cal/g [SD, 418], P < 0.01), reduced triglyceride absorption (MBT, 8.3 mg·h/dL [SD, 4.3] vs 17.7 mg·h/dL [SD, 10.3], P < 0.001), lower fat intake, and poorer QOL. Differences in CFA were not significant (90.9% [SD, 12.8] vs 95.4% [SD, 9.3]). After PERT, triglyceride absorption (Δ = 1.7 [SD, 3], P < 0.05) and QOL increased. CONCLUSIONS The MBT detected changes in triglyceride absorption in the absence of CFA changes. The MBT may be helpful in guiding PERT initiation in patients with CP before significant morbidity.
Collapse
|
24
|
Gawenis LR, Hodges CA, McHugh DR, Valerio DM, Miron A, Cotton CU, Liu J, Walker NM, Strubberg AM, Gillen AE, Mutolo MJ, Kotzamanis G, Bosch J, Harris A, Drumm ML, Clarke LL. A BAC Transgene Expressing Human CFTR under Control of Its Regulatory Elements Rescues Cftr Knockout Mice. Sci Rep 2019; 9:11828. [PMID: 31413336 PMCID: PMC6694137 DOI: 10.1038/s41598-019-48105-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/30/2019] [Indexed: 01/25/2023] Open
Abstract
Small-molecule modulators of cystic fibrosis transmembrane conductance regulator (CFTR) biology show promise in the treatment of cystic fibrosis (CF). A Cftr knockout (Cftr KO) mouse expressing mutants of human CFTR would advance in vivo testing of new modulators. A bacterial artificial chromosome (BAC) carrying the complete hCFTR gene including regulatory elements within 40.1 kb of DNA 5' and 25 kb of DNA 3' to the gene was used to generate founder mice expressing hCFTR. Whole genome sequencing indicated a single integration site on mouse chromosome 8 (8qB2) with ~6 gene copies. hCFTR+ offspring were bred to murine Cftr KO mice, producing hCFTR+/mCftr- (H+/m-) mice, which had normal survival, growth and goblet cell function as compared to wild-type (WT) mice. Expression studies showed hCFTR protein and transcripts in tissues typically expressing mCftr. Functionally, nasal potential difference and large intestinal short-circuit (Isc) responses to cAMP stimulation were similar in magnitude to WT mice, whereas small intestinal cAMP ΔIsc responses were reduced. A BAC transgenic mouse with functional hCFTR under control of its regulatory elements has been developed to enable the generation of mouse models of hCFTR mutations by gene editing for in vivo testing of new CF therapies.
Collapse
Affiliation(s)
- Lara R Gawenis
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Dr, Columbia, Missouri, 65211-3300, USA
- Department of Biomedical Sciences, University of Missouri, E102 Veterinary Medicine Bldg., Columbia, Missouri, 65211, USA
| | - Craig A Hodges
- Departments of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Daniel R McHugh
- Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Dana M Valerio
- Departments of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Alexander Miron
- Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Calvin U Cotton
- Departments of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Departments of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jinghua Liu
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Dr, Columbia, Missouri, 65211-3300, USA
| | - Nancy M Walker
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Dr, Columbia, Missouri, 65211-3300, USA
| | - Ashlee M Strubberg
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Dr, Columbia, Missouri, 65211-3300, USA
- Department of Biomedical Sciences, University of Missouri, E102 Veterinary Medicine Bldg., Columbia, Missouri, 65211, USA
| | - Austin E Gillen
- Human Molecular Genetics Program, Lurie Children's Research Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60614, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael J Mutolo
- Human Molecular Genetics Program, Lurie Children's Research Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60614, USA
| | - George Kotzamanis
- Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Jürgen Bosch
- Departments of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- InterRayBio, LLC, Baltimore, MD, USA
| | - Ann Harris
- Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Mitchell L Drumm
- Departments of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Lane L Clarke
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Dr, Columbia, Missouri, 65211-3300, USA.
- Department of Biomedical Sciences, University of Missouri, E102 Veterinary Medicine Bldg., Columbia, Missouri, 65211, USA.
| |
Collapse
|
25
|
Abstract
Malnutrition occurs when nutrient intake does not meet the needs for normal body functions and as a consequence leads to alterations of growth and development in children. Chronic illness puts children at risk for developing malnutrition. Because of children's rapid periods of growth and development, early diagnosis, prevention, and management of malnutrition are paramount. The reasons for malnutrition in children with chronic disease are multifactorial and are related to the underlying disease and non-illness-associated factors. This review addresses the causes, evaluation, and management of malnutrition in pediatric congenital heart disease, chronic kidney disease, liver disease, and cystic fibrosis.
Collapse
Affiliation(s)
- Catherine Larson-Nath
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Praveen Goday
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
26
|
Calvo-Lerma J, Hulst J, Boon M, Colombo C, Masip E, Ruperto M, Fornés-Ferrer V, van der Wiel E, Claes I, Garriga M, Roca M, Crespo-Escobar P, Bulfamante A, Woodcock S, Martínez-Barona S, Andrés A, de Boeck K, Ribes-Koninckx C, on behalf of MyCyFAPP project. Clinical validation of an evidence-based method to adjust Pancreatic Enzyme Replacement Therapy through a prospective interventional study in paediatric patients with Cystic Fibrosis. PLoS One 2019; 14:e0213216. [PMID: 30861039 PMCID: PMC6413926 DOI: 10.1371/journal.pone.0213216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A method to adjust Pancreatic Enzyme Replacement Therapy in Cystic Fibrosis is not currently available. OBJECTIVES To assess the in vivo efficacy of a method to adjust the dose of enzymatic supplement in CF extrapolated from previous in vitro digestion studies (theoretical optimal dose, TOD). Secondly, to assess how individual patient characteristics influence the expected coefficient of fat absorption (CFA) and thus to identify an individual correction factor to improve TOD. METHODS A prospective interventional study in 43 paediatric patients with CF from 5 European centres. They followed a 24h fixed diet with the theoretical optimal dose for each meal. Faecal collection was carried out between colorimetric markers in order to include all the faeces corresponding to the fixed diet. Beta regression models were applied to assess the associations of individual patient characteristics with the CFA. RESULTS Median CFA was 90% (84, 94% 1st, 3rd Q.) with no significant differences among centres. Intestinal transit time was positively associated with CFA (p = 0.007), but no statistical associations were found with and age, gender, phenotype or BMI. Regression model showed no improvement of the in vitro predicted theoretical optimal dose when taking individual patient characteristics into account. CONCLUSION Strict adherence to the theoretical optimal dose of enzymatic supplement for a prescribed meal, led to median CFA levels at the clinical target of 90% with a low variability between patients. The proposed method can be considered as a first approach for an evidence-based method in PERT dosing based on food characteristics. Results have to be confirmed in free dietary settings.
Collapse
Affiliation(s)
- Joaquim Calvo-Lerma
- Instituto de Investigación Sanitaria La Fe de Valencia, Valencia, Spain
- Universitat Politècnica de València, Research Institute of Food Engineering for Development, Valencia, Spain
| | - Jessie Hulst
- Erasmus Medical Center, Sophia Children’s Hospital, GD Rotterdam, the Netherlands
| | - Mieke Boon
- Pediatric Pulmonology and Cystic Fibrosis Unit, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Carla Colombo
- CF Center, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Via Commenda 9, Milan, Italy
| | - Etna Masip
- Instituto de Investigación Sanitaria La Fe de Valencia, Valencia, Spain
| | - Mar Ruperto
- Hospital Universitario Ramón y Cajal, Carretera Colmenar Viejo km 9, Madrid, Spain
| | | | - Els van der Wiel
- Erasmus Medical Center, Sophia Children’s Hospital, GD Rotterdam, the Netherlands
| | - Ine Claes
- Pediatric Pulmonology and Cystic Fibrosis Unit, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Maria Garriga
- Hospital Universitario Ramón y Cajal, Carretera Colmenar Viejo km 9, Madrid, Spain
| | - Maria Roca
- Instituto de Investigación Sanitaria La Fe de Valencia, Valencia, Spain
| | | | - Anna Bulfamante
- CF Center, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Via Commenda 9, Milan, Italy
| | - Sandra Woodcock
- Erasmus Medical Center, Sophia Children’s Hospital, GD Rotterdam, the Netherlands
| | | | - Ana Andrés
- Universitat Politècnica de València, Research Institute of Food Engineering for Development, Valencia, Spain
| | - Kris de Boeck
- Pediatric Pulmonology and Cystic Fibrosis Unit, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | | | | |
Collapse
|
27
|
Bertolini A, van de Peppel IP, Doktorova-Demmin M, Bodewes FAJA, de Jonge H, Bijvelds M, Verkade HJ, Jonker JW. Defective FXR-FGF15 signaling and bile acid homeostasis in cystic fibrosis mice can be restored by the laxative polyethylene glycol. Am J Physiol Gastrointest Liver Physiol 2019; 316:G404-G411. [PMID: 30653340 DOI: 10.1152/ajpgi.00188.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The gastrointestinal phenotype of cystic fibrosis (CF) features intestinal bile acid (BA) malabsorption, impaired intestinal farnesoid X receptor (FXR) activation, and consequently reduced fibroblast growth factor 19 (FGF19, FGF15 in mice) production. The osmotic laxative polyethylene glycol (PEG) has been shown to decrease intestinal mucus accumulation in CF mice and could, by doing so, improve BA reabsorption. Here we determined the effect of PEG on BA excretion and FXR-FGF15 signaling in CF mice. Male Cftr-/-tm1Unc (CF) and wild-type (WT) littermates were administered PEG 4000 in drinking water and fed either chow or a semisynthetic diet. PEG was withdrawn for 3 days before termination. Fecal BA excretion was measured at PEG dosages of 37 g/l (100%) and 0 g/l (0%). Ileal FXR activation was assessed by gene expression of its downstream targets Fgf15 and small heterodimer partner ( Shp). In CF mice, PEG withdrawal increased fecal BA excretion on either diet compared with full PEG dosage (chow, 2-fold, P = 0.06; semisynthetic, 4.4-fold, P = 0.007). PEG withdrawal did not affect fecal BA excretion in WT mice on either diet. After PEG withdrawal, gene expression levels of intestinal FXR target genes Fgf15 and Shp were decreased in CF mice but unaffected in WT littermates. PEG did not affect the gene expression of the main intestinal BA transporter apical sodium-dependent bile acid transporter (ASBT). PEG treatment ameliorates intestinal BA malabsorption in CF mice and restores intestinal FXR-FGF15 signaling, independent from Asbt gene expression. These findings highlight the potential of PEG in the prevention and treatment of the gastrointestinal phenotype of CF. NEW & NOTEWORTHY A gastrointestinal feature of cystic fibrosis is bile acid malabsorption and consequent impairment of farnesoid X receptor (FXR)-fibroblast growth factor 15 (FGF15) signaling. FXR-FGF15 signaling regulates various metabolic processes and could be implicated in metabolic and gastrointestinal complications of cystic fibrosis, such as diabetes and liver disease. In cystic fibrosis mice, treatment with the osmotic laxative polyethylene glycol is associated with decreased fecal bile acid loss and restoration of FXR-FGF15 signaling.
Collapse
Affiliation(s)
- Anna Bertolini
- Section of Molecular Metabolism and Nutrition, Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands.,Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center , Groningen , The Netherlands
| | - Ivo P van de Peppel
- Section of Molecular Metabolism and Nutrition, Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands.,Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center , Groningen , The Netherlands
| | - Marcela Doktorova-Demmin
- Section of Molecular Metabolism and Nutrition, Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Frank A J A Bodewes
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center , Groningen , The Netherlands
| | - Hugo de Jonge
- Gastroenterology & Hepatology, Erasmus MC-University Medical Center Rotterdam , The Netherlands
| | - Marcel Bijvelds
- Gastroenterology & Hepatology, Erasmus MC-University Medical Center Rotterdam , The Netherlands
| | - Henkjan J Verkade
- Section of Molecular Metabolism and Nutrition, Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands.,Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center , Groningen , The Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
28
|
Furnari M, De Alessandri A, Cresta F, Haupt M, Bassi M, Calvi A, Haupt R, Bodini G, Ahmed I, Bagnasco F, Giannini EG, Casciaro R. The role of small intestinal bacterial overgrowth in cystic fibrosis: a randomized case-controlled clinical trial with rifaximin. J Gastroenterol 2019; 54:261-270. [PMID: 30232597 DOI: 10.1007/s00535-018-1509-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/08/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Scientific literature shows a high prevalence of Small Intestinal Bacterial Overgrowth (SIBO) in patients with Cystic Fibrosis (CF). The role of SIBO in nutritional status and gastrointestinal symptoms in CF is not known. Our aim was to study epidemiology and clinical impact of SIBO while assessing the efficacy of rifaximin in eradicating SIBO in CF patients. METHODS Symptoms questionnaire and Glucose Breath Test (GBT) were given to 79 CF patients (median age 19.6 years; 9.2-36.9). Subjects with a positive GBT were enrolled in a randomized controlled trial and received rifaximin 1200 mg for 14 days or no treatment. Questionnaire and GBT were repeated 1 month after the end of treatment or 45 days after the first negative GBT. RESULTS Out of 79 patients, 25 were affected by SIBO (31.6%) with a significant correlation with lower BMI, SDS-BMI (p < 0.05) and serum albumin levels (p < 0.05), independently from pancreas insufficiency. Twenty-three patients took part in the randomized trial, 13 patients (56.5%) in rifaximin group and 10 patients (43.5%) in control group. Eradication rate of SIBO was 9/10 (90%) in rifaximin group and 2/6 (33.3%) in control group (p < 0.05). In the rifaximin group, gastrointestinal symptom improvement was observed in 4/5 patients aged ≤ 14 years and in 0/5 patients aged > 14 years (p < 0.05); in 2/6 patients in the control group. CONCLUSIONS CF patients show a high prevalence of SIBO, related to a poorer nutritional status. Rifaximin therapy is well tolerated and the results are promising in terms of efficacy in eradicating small intestinal bacterial overgrowth in CF.
Collapse
Affiliation(s)
- Manuele Furnari
- Department of Internal Medicine, Gastroenterology Unit, Policlinico "San Martino", University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy.
| | | | - Federico Cresta
- Cystic Fibrosis Center, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Maria Haupt
- Cystic Fibrosis Center, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Marta Bassi
- Cystic Fibrosis Center, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Angela Calvi
- Gastroenterology and Pediatric Endoscopy Unit, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Riccardo Haupt
- Epidemiology and Biostatistic Service, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Giorgia Bodini
- Department of Internal Medicine, Gastroenterology Unit, Policlinico "San Martino", University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Iftikhar Ahmed
- Department of Gastroenterology, Aldara Hospital and Medical Centre, Riyadh, Kingdom of Saudi Arabia
| | - Francesca Bagnasco
- Epidemiology and Biostatistic Service, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Edoardo Giovanni Giannini
- Department of Internal Medicine, Gastroenterology Unit, Policlinico "San Martino", University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Rosaria Casciaro
- Cystic Fibrosis Center, IRCCS G. Gaslini Institute, Genoa, Italy
| |
Collapse
|
29
|
Calvo-Lerma J, Fornés-Ferrer V, Peinado I, Heredia A, Ribes-Koninckx C, Andrés A. A first approach for an evidence-based in vitro digestion method to adjust pancreatic enzyme replacement therapy in cystic fibrosis. PLoS One 2019; 14:e0212459. [PMID: 30794618 PMCID: PMC6386532 DOI: 10.1371/journal.pone.0212459] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/01/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Patients with cystic fibrosis have to take enzymatic supplements to allow for food digestion. However, an evidence-based method to adjust Pancreatic Enzyme Replacement Therapy (PERT) is inexistent, and lipid content of meals is used as a rough criterion. OBJECTIVE In this study, an in vitro digestion model was set up to determine the theoretical optimal dose (TOD) of enzymatic supplement for a selection of foods, which is the dose that allows for maximum lipolysis extent. METHODS A static in vitro digestion model was applied to simulate digestion of eight foods covering a wide range of lipid contents. First, the dose of the enzymatic supplement was fixed at 2000 lipase units per gram of fat (LU/g fat) using intestinal pH and bile salt concentration as variables. Second, intestinal pH and bile salt concentrations were fixed and the variable was the dose of the enzymatic supplement. Lipolysis extent was determined by measuring the free fatty acids released from initial triglycerides content of foods after digestion. Results in terms of percentage of lipolysis extent were fitted into a linear-mixed segmented model and the deducted equations were used to predict the TOD to reach 90% of lipolysis in every food. In addition, the effect of intestinal pH and bile salt concentration were investigated. RESULTS The predictive equations obtained for the assessed foods showed that lipolysis was not only dependent on the dose of the enzyme supplement or the lipid content. Moreover, intestinal pH and bile salt concentration had significant effects on lipolysis. Therefore an evidence-based model can be developed taking into account these variables. CONCLUSIONS Depending on food characteristics, a specific TOD should be assigned to achieve an optimal digestion extent. This work represents a first step towards an evidence-based method for PERT dosing, which will be applied in an in vivo setting to validate its efficacy.
Collapse
Affiliation(s)
- Joaquim Calvo-Lerma
- Universitat Politècnica de València, Research Institute of Food Engineering for Development, Valencia, Spain
- Instituto de Investigación Sanitaria La Fe de Valencia, Valencia, Spain
| | | | - Irene Peinado
- Universitat Politècnica de València, Research Institute of Food Engineering for Development, Valencia, Spain
| | - Ana Heredia
- Universitat Politècnica de València, Research Institute of Food Engineering for Development, Valencia, Spain
| | | | - Ana Andrés
- Universitat Politècnica de València, Research Institute of Food Engineering for Development, Valencia, Spain
| |
Collapse
|
30
|
Khalaf RT, Green D, Amankwah EK, Peck J, Carr V, Goldenberg NA, Wilsey M. Percutaneous Endoscopic Gastrostomy Tubes May Be Associated With Preservation of Lung Function in Patients With Cystic Fibrosis. Nutr Clin Pract 2018; 34:290-296. [DOI: 10.1002/ncp.10219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Racha T. Khalaf
- Digestive Health Institute & Section of Pediatric Gastroenterology; Hepatology and Nutrition; Children's Hospital Colorado; University of Colorado School of Medicine; Aurora Colorado USA
- Department of Medical Education; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
| | - Deanna Green
- Department of Pulmonology; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
| | - Ernest K. Amankwah
- Health Informatics; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
| | - Jacquelin Peck
- Department of Anesthesiology; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
| | - Vanessa Carr
- Department of Nutrition Services; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
- Medical Nutrition; Kate Farms, Inc; Santa Barbara California USA
| | - Neil A. Goldenberg
- Department of Hematology; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
| | - Michael Wilsey
- Department of Gastroenterology; Hepatology and Nutrition; Johns Hopkins All Children's Hospital; St. Petersburg Florida USA
| |
Collapse
|
31
|
Calvo‐Lerma J, Fornés‐Ferrer V, Heredia A, Andrés A. In Vitro Digestion of Lipids in Real Foods: Influence of Lipid Organization Within the Food Matrix and Interactions with Nonlipid Components. J Food Sci 2018; 83:2629-2637. [PMID: 30216443 PMCID: PMC6282792 DOI: 10.1111/1750-3841.14343] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/27/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022]
Abstract
In vitro digestion research has scarcely addressed the assessment of the complexity of digestion in real food. The aim of the present study was to evaluate the influence of intestinal conditions, nonlipid components, and lipid organization within the food matrix on lipolysis extent. A selection of 52 foods was studied under different simulated intestinal conditions, including those related to patients with cystic fibrosis (pH6, bile salts 1 mM due to decreased pancreatic and biliary secretions) and to healthy subjects (pH7, bile salts 10 mM). Linear mixed regression models were applied to explain associations of food properties with lipolysis. Normal intestinal conditions allowed for optimal lipolysis in most of the foods in contrast to the altered intestinal scenario (30 compared with 1 food reaching > 90% lipolysis). Lipid-protein and lipid-starch interactions were evidenced to significantly affect lipolysis (P < 0.001) in all the digestion conditions, decreasing in those foods with low fat and high protein or high starch content. In addition, under decreased intestinal pH and bile concentration, lipolysis was lower in foods with complex solid structures and continuous lipid phase than in the oil-in-water continuous aqueous phase (global P < 0.01). However, in the normal conditions lipid organization within the food matrix did not show a significant effect on lipolysis (global P = 0.08). In conclusion, food properties play a crucial role in lipolysis, which should be considered when establishing dietary recommendations. PRACTICAL APPLICATION Food composition, lipid organization within the food matrix, and gastrointestinal conditions are key factors affecting lipolysis. Knowledge on that can be used to modulate lipolysis performance after food ingestion. Different applications are foreseen, as food design and nutritional recommendations for the general populations and specific target groups. The most immediate application is related to the scope of the research project that frames this work (www.mycyfapp.eu). These results have contributed to the development of a mobile app for cystic fibrosis patients, which includes an algorithm for enzyme dose prediction based on food properties. The app is currently being tested in a clinical trial setting.
Collapse
Affiliation(s)
- Joaquim Calvo‐Lerma
- Inst. de Ingeniería de Alimentos para el DesarrolloUniv. Politècnica de ValènciaCamino de Vera s/n.46022 ValenciaSpain
- Inst. de Investigación Sanitaria La FeAvenida Fernando Abril Martorell 10646026ValenciaSpain
| | - Victoria Fornés‐Ferrer
- Inst. de Investigación Sanitaria La FeAvenida Fernando Abril Martorell 10646026ValenciaSpain
| | - Ana Heredia
- Inst. de Ingeniería de Alimentos para el DesarrolloUniv. Politècnica de ValènciaCamino de Vera s/n.46022 ValenciaSpain
| | - Ana Andrés
- Inst. de Ingeniería de Alimentos para el DesarrolloUniv. Politècnica de ValènciaCamino de Vera s/n.46022 ValenciaSpain
| |
Collapse
|
32
|
van de Peppel IP, Bodewes FAJA, Verkade HJ, Jonker JW. Bile acid homeostasis in gastrointestinal and metabolic complications of cystic fibrosis. J Cyst Fibros 2018; 18:313-320. [PMID: 30201330 DOI: 10.1016/j.jcf.2018.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022]
Abstract
With the improved treatment of the pulmonary complications of cystic fibrosis (CF), gastrointestinal problems have become more important in the morbidity in CF. A hallmark of the gastrointestinal phenotype of CF, apart from pancreatic insufficiency, is a disruption of bile acid homeostasis. Bile acid homeostasis is important for many gastrointestinal processes including fat absorption, inflammation, microbial composition, as well as regulation of whole body energy metabolism. This review describes the impairment of bile acid homeostasis in CF, its possible consequences for gastrointestinal and metabolic complications and its potential as a target for therapy.
Collapse
Affiliation(s)
- Ivo P van de Peppel
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, the Netherlands; Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands.
| | - Frank A J A Bodewes
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Henkjan J Verkade
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, the Netherlands; Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands.
| |
Collapse
|
33
|
Ratchford TL, Teckman JH, Patel DR. Gastrointestinal pathophysiology and nutrition in cystic fibrosis. Expert Rev Gastroenterol Hepatol 2018; 12:853-862. [PMID: 30019967 DOI: 10.1080/17474124.2018.1502663] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cystic fibrosis (CF) is a severe, progressive, multisystemic disease that is caused by mutations in the cystic fibrosis transmembrane conductance regulator gene. Optimizing nutrition is critical, as higher growth parameters are associated with better pulmonary function and outcomes, but unfortunately patients with this disease are prone to malnutrition, growth failure, and vitamin deficiencies. The purpose of this review is to provide a timely highlight of the physiologic processes and outcome data to support today's management strategies, as well as review these principles themselves. Areas covered: This review covers the background of the importance of vigilant attention to nutrition and growth in these patients, the underlying physiology leading to an abnormal gastrointestinal tract and its role in CF malnutrition, and current evaluation and management strategies to address nutrition in CF. Analysis of up-to-date relevant literature was performed using PubMed. Expert commentary: Advances in research and clinical developments over the years have improved knowledge of this disease as well as patient outcomes. Of particular importance is optimizing nutrition especially in the early stages of life, as well as accounting for the markedly abnormal CF intestinal milieu when addressing the gastrointestinal and nutritional needs of these patients.
Collapse
Affiliation(s)
- Thomas L Ratchford
- a Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition , Saint Louis University School of Medicine , Saint Louis , Missouri , USA
| | - Jeffrey H Teckman
- a Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition , Saint Louis University School of Medicine , Saint Louis , Missouri , USA
| | - Dhiren R Patel
- a Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition , Saint Louis University School of Medicine , Saint Louis , Missouri , USA
| |
Collapse
|
34
|
Becker KA, Riethmüller J, Seitz AP, Gardner A, Boudreau R, Kamler M, Kleuser B, Schuchman E, Caldwell CC, Edwards MJ, Grassmé H, Brodlie M, Gulbins E. Sphingolipids as targets for inhalation treatment of cystic fibrosis. Adv Drug Deliv Rev 2018; 133:66-75. [PMID: 29698625 DOI: 10.1016/j.addr.2018.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 01/19/2023]
Abstract
Studies over the past several years have demonstrated the important role of sphingolipids in cystic fibrosis (CF), chronic obstructive pulmonary disease and acute lung injury. Ceramide is increased in airway epithelial cells and alveolar macrophages of CF mice and humans, while sphingosine is dramatically decreased. This increase in ceramide results in chronic inflammation, increased death of epithelial cells, release of DNA into the bronchial lumen and thereby an impairment of mucociliary clearance; while the lack of sphingosine in airway epithelial cells causes high infection susceptibility in CF mice and possibly patients. The increase in ceramide mediates an ectopic expression of β1-integrins in the luminal membrane of CF epithelial cells, which results, via an unknown mechanism, in a down-regulation of acid ceramidase. It is predominantly this down-regulation of acid ceramidase that results in the imbalance of ceramide and sphingosine in CF cells. Correction of ceramide and sphingosine levels can be achieved by inhalation of functional acid sphingomyelinase inhibitors, recombinant acid ceramidase or by normalization of β1-integrin expression and subsequent re-expression of endogenous acid ceramidase. These treatments correct pulmonary inflammation and prevent or treat, respectively, acute and chronic pulmonary infections in CF mice with Staphylococcus aureus and mucoid or non-mucoid Pseudomonas aeruginosa. Inhalation of sphingosine corrects sphingosine levels only and seems to mainly act against the infection. Many antidepressants are functional inhibitors of the acid sphingomyelinase and were designed for systemic treatment of major depression. These drugs could be repurposed to treat CF by inhalation.
Collapse
|
35
|
O'Malley Y, Rotti PG, Thornell IM, Vanegas Calderón OG, Febres-Aldana C, Durham K, Yao J, Li X, Zhu Z, Norris AW, Zabner J, Engelhardt JF, Uc A. Development of a polarized pancreatic ductular cell epithelium for physiological studies. J Appl Physiol (1985) 2018; 125:97-106. [PMID: 29517421 PMCID: PMC6086968 DOI: 10.1152/japplphysiol.00043.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 01/06/2023] Open
Abstract
Pancreatic ductular epithelial cells comprise the majority of duct cells in pancreas, control cystic fibrosis transmembrane conductance regulator (CFTR)-dependent bicarbonate ([Formula: see text]) secretion, but are difficult to grow as a polarized monolayer. Using NIH-3T3-J2 fibroblast feeder cells and a Rho-associated kinase inhibitor, we produced well-differentiated and polarized porcine pancreatic ductular epithelial cells. Cells grown on semipermeable filters at the air-liquid interface developed typical epithelial cell morphology and stable transepithelial resistance and expressed epithelial cell markers (zona occludens-1 and β-catenin), duct cell markers (SOX-9 and CFTR), but no acinar (amylase) or islet cell (chromogranin) markers. Polarized cells were studied in Ussing chambers bathed in Krebs-Ringer [Formula: see text] solution at 37°C gassed with 5% CO2 to measure short-circuit currents ( Isc). Ratiometric measurement of extracellular pH was performed with fluorescent SNARF-conjugated dextran at 5% CO2. Cells demonstrated a baseline Isc (12.2 ± 3.2 μA/cm2) that increased significantly in response to apical forskolin-IBMX (∆ Isc: 35.4 ± 3.8 μA/cm2, P < 0.001) or basolateral secretin (∆ Isc: 31.4 ± 2.5 μA/cm2, P < 0.001), both of which increase cellular levels of cAMP. Subsequent addition of apical GlyH-101, a CFTR inhibitor, decreased the current (∆ Isc: 20.4 ± 3.8 μA/cm2, P < 0.01). Extracellular pH and [Formula: see text] concentration increased significantly after forskolin-IBMX (pH: 7.18 ± 0.23 vs. 7.53 ± 0.19; [Formula: see text] concentration, 14.5 ± 5.9 vs. 31.8 ± 13.4 mM; P < 0.05 for both). We demonstrate the development of a polarized pancreatic ductular epithelial cell epithelium with CFTR-dependent [Formula: see text] secretion in response to secretin and cAMP. This model is highly relevant, as porcine pancreas physiology is very similar to humans and pancreatic damage in the cystic fibrosis pig model recapitulates that of humans. NEW & NOTEWORTHY Pancreas ductular epithelial cells control cystic fibrosis transmembrane conductance regulator (CFTR)-dependent bicarbonate secretion. Their function is critical because when CFTR is deficient in cystic fibrosis bicarbonate secretion is lost and the pancreas is damaged. Mechanisms that control pancreatic bicarbonate secretion are incompletely understood. We generated well-differentiated and polarized porcine pancreatic ductular epithelial cells and demonstrated feasibility of bicarbonate secretion. This novel method will advance our understanding of pancreas physiology and mechanisms of bicarbonate secretion.
Collapse
Affiliation(s)
- Yunxia O'Malley
- Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
| | - Pavana G Rotti
- Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Ian M Thornell
- Department of Internal Medicine, University of Iowa , Iowa City, Iowa
| | | | - Christopher Febres-Aldana
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center , Miami Beach, Florida
| | - Katelin Durham
- Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
| | - Jianrong Yao
- Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
| | - Xiaopeng Li
- Department of Internal Medicine, University of Iowa , Iowa City, Iowa
| | - Zheng Zhu
- Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
| | - Andrew W Norris
- Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa , Iowa City, Iowa
| | - Joseph Zabner
- Department of Internal Medicine, University of Iowa , Iowa City, Iowa
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Aliye Uc
- Stead Family Department of Pediatrics, University of Iowa , Iowa City, Iowa
| |
Collapse
|
36
|
Bolia R, Ooi CY, Lewindon P, Bishop J, Ranganathan S, Harrison J, Ford K, van der Haak N, Oliver MR. Practical approach to the gastrointestinal manifestations of cystic fibrosis. J Paediatr Child Health 2018; 54:609-619. [PMID: 29768684 DOI: 10.1111/jpc.13921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/07/2018] [Accepted: 03/02/2018] [Indexed: 12/14/2022]
Abstract
Cystic fibrosis (CF) is the most common, life-shortening, genetic illness affecting children in Australia and New Zealand. The genetic abnormality results in abnormal anion transport across the apical membrane of epithelial cells in a number of organs, including the lungs, gastrointestinal tract, liver and genito-urinary tract. Thus, CF is a multi-system disorder that requires a multi-disciplinary approach. Respiratory disease is the predominant cause of both morbidity and mortality in patients with CF. However, there are significant and clinically relevant gastrointestinal, liver, pancreatic and nutritional manifestations that must be detected and managed in a timely and structured manner. The aim of this review is to provide evidence-based information and clinical algorithms to guide the nutritional and gastrointestinal management of patients with CF.
Collapse
Affiliation(s)
- Rishi Bolia
- Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Chee Y Ooi
- School of Women and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,miCF Research Centre and Department of Gastroenterology, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Peter Lewindon
- Department of Gastroenterology, Lady Cilento Children's Hospital and Queensland Liver Transplant Service, Brisbane, Queensland, Australia
| | - Jonathan Bishop
- Department of Paediatric Gastroenterology, Starship Children's Hospital, Auckland, New Zealand
| | - Sarath Ranganathan
- Department of Respiratory Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.,Infection and Immunity Theme, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Jo Harrison
- Department of Respiratory Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.,Infection and Immunity Theme, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Kristyn Ford
- Department of Nutrition and Dietetics, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Natalie van der Haak
- Department of Nutrition and Dietetics, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Mark R Oliver
- Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
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
|
38
|
Strubberg AM, Liu J, Walker NM, Stefanski CD, MacLeod RJ, Magness ST, Clarke LL. Cftr Modulates Wnt/β-Catenin Signaling and Stem Cell Proliferation in Murine Intestine. Cell Mol Gastroenterol Hepatol 2017; 5:253-271. [PMID: 29675451 PMCID: PMC5904038 DOI: 10.1016/j.jcmgh.2017.11.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/18/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Cystic fibrosis (CF) patients and CF mouse models have increased risk for gastrointestinal tumors. CF mice show augmented intestinal proliferation of unknown etiology and an altered intestinal environment. We examined the role of the cystic fibrosis transmembrane conductance regulator (Cftr) in Wnt/β-catenin signaling, stem cell proliferation, and its functional expression in the active intestinal stem cell (ISC) population. Dysregulation of intracellular pH (pHi) in CF ISCs was investigated for facilitation of Wnt/β-catenin signaling. METHODS Crypt epithelia from wild-type (WT) and CF mice were compared ex vivo and in intestinal organoids (enteroids) for proliferation and Wnt/β-catenin signaling by standard assays. Cftr in ISCs was assessed by immunoblot of sorted Sox9 enhanced green fluorescent protein(EGFP) intestinal epithelia and pHi regulation by confocal microfluorimetry of leucine-rich G-protein-coupled receptor 5 ISCs. Plasma membrane association of the Wnt transducer Dishevelled 2 (Dvl2) was assessed by fluorescence imaging of live enteroids from WT and CF mice crossed with Dvl2-EGFP/ACTB-tdTomato,-EGFP)Luo/J (RosamT/mG) mice. RESULTS Relative to WT, CF intestinal crypts showed an ∼30% increase in epithelial and Lgr5+ ISC proliferation and increased Wnt/β-catenin signaling. Cftr was expressed in Sox9EGFPLo ISCs and loss of Cftr induced an alkaline pHi in ISCs. CF crypt-base columnar cells showed a generalized increase in plasma membrane Dvl2-EGFP association as compared with WT. Dvl2-EGFP membrane association was charge- and pH-dependent and increased in WT crypt-base columnar cells by Cftr inhibition. CONCLUSIONS CF intestine shows increased ISC proliferation and Wnt/β-catenin signaling. Loss of Cftr increases pHi in ISCs, which stabilizes the plasma membrane association of the Wnt transducer Dvl, likely facilitating Wnt/β-catenin signaling. Absence of Cftr-dependent suppression of ISC proliferation in the CF intestine may contribute to increased risk for intestinal tumors.
Collapse
Key Words
- CBC, crypt-base columnar cell
- CCH, carbachol
- CF, cystic fibrosis
- Cftr, cystic fibrosis transmembrane conductance regulator
- Cystic Fibrosis
- DEP, Dishevelled, Egl-10, and Pleckstrin
- Dishevelled
- Dvl, Dishevelled
- EGFP, enhanced green fluorescent protein
- EdU, 5-ethynyl-2’-deoxyuridine
- Fz, Frizzled
- GI, gastrointestinal
- ISC, intestinal stem cell
- Intracellular pH
- KO, knockout
- Lgr5, leucine-rich G-protein–coupled receptor 5
- Neoplasia
- Organoids
- PBS, phosphate-buffered saline
- PDZ, Post synaptic density protein, Drosophila disc large tumor suppressor, and Zonula occludens-1 protein
- PH3, phospho-histone H3
- ROI, region of interest
- WT, wild type
- pHi, intracellular pH
Collapse
Affiliation(s)
- Ashlee M. Strubberg
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Jinghua Liu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Nancy M. Walker
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Casey D. Stefanski
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - R. John MacLeod
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Scott T. Magness
- Department of Medicine, Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lane L. Clarke
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,Correspondence Address correspondence to: Lane L. Clarke, DVM, PhD, 324D Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, Missouri 65211-3300. fax: (573) 884–4232.
| |
Collapse
|
39
|
Singh VK, Schwarzenberg SJ. Pancreatic insufficiency in Cystic Fibrosis. J Cyst Fibros 2017; 16 Suppl 2:S70-S78. [DOI: 10.1016/j.jcf.2017.06.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 12/19/2022]
|
40
|
Grassmé H, Henry B, Ziobro R, Becker KA, Riethmüller J, Gardner A, Seitz AP, Steinmann J, Lang S, Ward C, Schuchman EH, Caldwell CC, Kamler M, Edwards MJ, Brodlie M, Gulbins E. β1-Integrin Accumulates in Cystic Fibrosis Luminal Airway Epithelial Membranes and Decreases Sphingosine, Promoting Bacterial Infections. Cell Host Microbe 2017; 21:707-718.e8. [PMID: 28552668 PMCID: PMC5475347 DOI: 10.1016/j.chom.2017.05.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 03/10/2017] [Accepted: 05/02/2017] [Indexed: 11/18/2022]
Abstract
Chronic pulmonary colonization with bacterial pathogens, particularly Pseudomonas aeruginosa, is the primary cause of morbidity and mortality in patients with cystic fibrosis (CF). We observed that β1-integrins accumulate on the luminal membrane of upper-airway epithelial cells from mice and humans with CF. β1-integrin accumulation is due to increased ceramide and the formation of ceramide platforms that trap β1-integrins on the luminal pole of bronchial epithelial cells. β1-integrins downregulate acid ceramidase expression, resulting in further accumulation of ceramide and consequent reduction of surface sphingosine, a lipid that kills bacteria. Interrupting this vicious cycle by triggering surface β1-integrin internalization via anti-β1-integrin antibodies or the RGD peptide ligand-or by genetic or pharmacological correction of ceramide levels-normalizes β1-integrin distribution and sphingosine levels in CF epithelial cells and prevents P. aeruginosa infection in CF mice. These findings suggest a therapeutic avenue to ameliorate CF-associated bacterial infections.
Collapse
Affiliation(s)
- Heike Grassmé
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Brian Henry
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany; Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, Ohio 45229, USA
| | - Regan Ziobro
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Joachim Riethmüller
- Center for Pediatric Clinical Studies, Children's Clinic, University of Tuebingen, Hoppe-Seyler-Strasse 1, 72076 Tübingen, Germany
| | - Aaron Gardner
- Institute of Cellular Medicine, Newcastle University, c/o Level 3, Clinical Resource Building, Great North Children's Hospital, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Aaron P Seitz
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, Ohio 45229, USA
| | - Joerg Steinmann
- Department of Medical Microbiology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Christopher Ward
- Institute of Cellular Medicine, Newcastle University, c/o Level 3, Clinical Resource Building, Great North Children's Hospital, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Edward H Schuchman
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Charles C Caldwell
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, Ohio 45229, USA
| | - Markus Kamler
- West German Heart and Vascular Center Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Michael J Edwards
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, Ohio 45229, USA
| | - Malcolm Brodlie
- Institute of Cellular Medicine, Newcastle University, c/o Level 3, Clinical Resource Building, Great North Children's Hospital, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany; Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, Ohio 45229, USA.
| |
Collapse
|
41
|
Calvo-Lerma J, Martinez-Jimenez CP, Lázaro-Ramos JP, Andrés A, Crespo-Escobar P, Stav E, Schauber C, Pannese L, Hulst JM, Suárez L, Colombo C, Barreto C, de Boeck K, Ribes-Koninckx C. Innovative approach for self-management and social welfare of children with cystic fibrosis in Europe: development, validation and implementation of an mHealth tool (MyCyFAPP). BMJ Open 2017; 7:e014931. [PMID: 28302638 PMCID: PMC5372031 DOI: 10.1136/bmjopen-2016-014931] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION For the optimal management of children with cystic fibrosis, there are currently no efficient tools for the precise adjustment of pancreatic enzyme replacement therapy, either for advice on appropriate dietary intake or for achieving an optimal nutrition status. Therefore, we aim to develop a mobile application that ensures a successful nutritional therapy in children with cystic fibrosis. METHODS AND ANALYSIS A multidisciplinary team of 12 partners coordinate their efforts in 9 work packages that cover the entire so-called 'from laboratory to market' approach by means of an original and innovative co-design process. A cohort of 200 patients with cystic fibrosis aged 1-17 years are enrolled. We will develop an innovative, clinically tested mobile health application for patients and health professionals involved in cystic fibrosis management. The mobile application integrates the research knowledge and innovative tools for maximising self-management with the aim of leading to a better nutritional status, quality of life and disease prognosis. Bringing together different and complementary areas of knowledge is fundamental for tackling complex challenges in disease treatment, such as optimal nutrition and pancreatic enzyme replacement therapy in cystic fibrosis. Patients are expected to benefit the most from the outcomes of this innovative project. ETHICS AND DISSEMINATION The project is approved by the Ethics Committee of the coordinating organisation, Hospital Universitari La Fe (Ref: 2014/0484). Scientific findings will be disseminated via journals and conferences addressed to clinicians, food scientists, information and communications technology experts and patients. The specific dissemination working group within the project will address the wide audience communication through the website (http://www.mycyfapp.eu), the social networks and the newsletter.
Collapse
Affiliation(s)
- Joaquim Calvo-Lerma
- Instituto de Investigación Sanitaria La Fe. Avenida Fernando Abril Martorell, Valencia, Spain
- Instituto de Ingeniería de Alimentos para el Desarrollo, Universitat Politècnica de València, Valencia, Spain
| | - Celia P Martinez-Jimenez
- University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | | | - Ana Andrés
- Instituto de Ingeniería de Alimentos para el Desarrollo, Universitat Politècnica de València, Valencia, Spain
| | - Paula Crespo-Escobar
- Instituto de Investigación Sanitaria La Fe. Avenida Fernando Abril Martorell, Valencia, Spain
| | | | | | | | - Jessie M Hulst
- Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lucrecia Suárez
- Servicio Madrileño de Salud—Hospital Universitario Ramón y Cajal, Colmenar Viejo, Spain
| | - Carla Colombo
- Università degli Studi di Milano. Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Celeste Barreto
- Associação Portuguesa para a Investigação e Desenvolvimento da Faculdade de Medicina, Lisbon, Portugal
| | - Kris de Boeck
- Department of Paediatrics, University Hospital of Leuven, University of Leuven, Leuven, Belgium
| | - Carmen Ribes-Koninckx
- Instituto de Investigación Sanitaria La Fe. Avenida Fernando Abril Martorell, Valencia, Spain
| |
Collapse
|
42
|
Enteral tube feeding for individuals with cystic fibrosis: Cystic Fibrosis Foundation evidence-informed guidelines. J Cyst Fibros 2016; 15:724-735. [PMID: 27599607 DOI: 10.1016/j.jcf.2016.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 01/12/2023]
Abstract
Nutrition is integral to the care of individuals with cystic fibrosis (CF). Better nutritional status is associated with improved pulmonary function. In some individuals with CF, enteral tube feeding can be useful in achieving optimal nutritional status. Current nutrition guidelines do not include detailed recommendations for enteral tube feeding. The Cystic Fibrosis Foundation convened an expert panel to develop enteral tube feeding recommendations based on a systematic review of the evidence and expert opinion. These guidelines address when to consider enteral tube feeding, assessment of confounding causes of poor nutrition in CF, preparation of the patient for placement of the enteral feeding tube, management of the tube after placement and education about enteral feeding. These recommendations are intended to guide the CF care team, individuals with CF, and their families through the enteral tube feeding process.
Collapse
|
43
|
Demeyer S, De Boeck K, Witters P, Cosaert K. Beyond pancreatic insufficiency and liver disease in cystic fibrosis. Eur J Pediatr 2016; 175:881-94. [PMID: 27055450 DOI: 10.1007/s00431-016-2719-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Cystic fibrosis is a life shortening hereditary disease, primarily leading to progressive pulmonary infection and exocrine pancreatic dysfunction. Several gastrointestinal complications other than malabsorption can arise during the disease course and with the progressively increasing life span of patients with CF; new and more rare complications are being recognized. We review the literature on gastrointestinal manifestations in CF, excluding the liver and pancreas. CONCLUSION We describe the clinical presentation and treatment of more common conditions like gastroesophageal reflux, small intestinal bacterial overgrowth, intussusception, meconium ileus, distal intestinal obstruction syndrome, and constipation, and we also discuss what is known on celiac disease, appendicitis, fibrosing colonopathy, inflammation and inflammatory bowel disease and gastrointestinal cancer. WHAT IS KNOWN • Gastrointestinal complications arise early in the course of the disease and have a severe impact on the quality of life of the patients. What is New: • This review is a concise summary of the current literature on gastrointestinal complications of cystic fibrosis. • We focused on clinical presentation and diagnostic investigations and provide a comprehensive resume of the current treatment options.
Collapse
Affiliation(s)
- Stephanie Demeyer
- Universitaire Ziekenhuizen Leuven, Leuven, Vlaams-Brabant, Belgium. .,Department of Pediatrics, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium.
| | - Kris De Boeck
- Department of Pediatrics, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium
| | - Peter Witters
- Department of Pediatrics, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium
| | - Katrien Cosaert
- Department of Pharmocology, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Development of cystic fibrosis transmembrane conductance regulator (CFTR) modulators, small molecule therapies that target the basic defect in cystic fibrosis (CF), represents a new era in CF treatment. This review highlights recent progress in CF therapeutics as an example of precision medicine and personalized approaches to test CFTR modulators using preclinical model systems. RECENT FINDINGS CFTR modulators are now clinically available for approximately 50% of the United States CF population. The CFTR potentiator, ivacaftor, is approved for people with CF ages 2 years and older with at least one gating mutation (G551D, G1244E, G1349D, G178R, G551S, S1251N, S1255P, S549N, or S549R) or the R117H conductance mutation. The recent Food and Drug Administration approval of the corrector/potentiator combination, lumacaftor/ivacaftor, expands modulator therapy to people with CF homozygous for the F508del mutation, ages 12 years and older. Ivacaftor and lumacaftor, however, do not fully restore CFTR activity. Thus, next-generation correctors and potentiators are in development. Read-through agents targeting nonsense mutations and genotype agnostic treatments (gene-editing and gene therapy) are also in various phases of clinical development. SUMMARY CFTR modulators promise to transform the therapeutic landscape in CF in a precision based fashion. Areas of ongoing research include developing drugs for all mutation classes so that all persons with CF can benefit from these therapies, and refining preclinical assays that allow the selection of the most effective treatments on an individual basis.
Collapse
Affiliation(s)
- Stacey L Martiniano
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | | |
Collapse
|
45
|
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
|
46
|
Management of pancreatic, gastrointestinal and liver complications in adult cystic fibrosis. Rev Mal Respir 2015; 32:566-85. [DOI: 10.1016/j.rmr.2014.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 12/09/2014] [Indexed: 01/27/2023]
|
47
|
Abstract
Cystic fibrosis is an autosomal recessive, monogenetic disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The gene defect was first described 25 years ago and much progress has been made since then in our understanding of how CFTR mutations cause disease and how this can be addressed therapeutically. CFTR is a transmembrane protein that transports ions across the surface of epithelial cells. CFTR dysfunction affects many organs; however, lung disease is responsible for the vast majority of morbidity and mortality in patients with cystic fibrosis. Prenatal diagnostics, newborn screening and new treatment algorithms are changing the incidence and the prevalence of the disease. Until recently, the standard of care in cystic fibrosis treatment focused on preventing and treating complications of the disease; now, novel treatment strategies directly targeting the ion channel abnormality are becoming available and it will be important to evaluate how these treatments affect disease progression and the quality of life of patients. In this Primer, we summarize the current knowledge, and provide an outlook on how cystic fibrosis clinical care and research will be affected by new knowledge and therapeutic options in the near future. For an illustrated summary of this Primer, visit: http://go.nature.com/4VrefN.
Collapse
|
48
|
Mascarenhas MR, Mondick J, Barrett JS, Wilson M, Stallings VA, Schall JI. Malabsorption blood test: Assessing fat absorption in patients with cystic fibrosis and pancreatic insufficiency. J Clin Pharmacol 2015; 55:854-65. [PMID: 25689042 DOI: 10.1002/jcph.484] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/12/2015] [Indexed: 12/19/2022]
Abstract
The malabsorption blood test (MBT), consisting of pentadecanoic acid (PA), a free fatty acid, and triheptadecanoic acid (THA), a triglyceride that requires pancreatic lipase for absorption of the heptadecanoic acid (HA), was developed to assess fat malabsorption in patients with cystic fibrosis (CF) and pancreatic insufficiency (PI). The objective was to construct a population pharmacokinetic (PK) model to describe PA and HA disposition in healthy subjects and CF subjects. A model was simultaneously fit to PA and HA concentrations, consisting of 1-compartment disposition and a transit model to describe absorption. PA bioavailability estimates for CF subjects without pancreatic enzyme administration (1.07 [0.827, 1.42]) and with enzymes (0.88 [0.72, 1.09]) indicated PA absorption comparable to healthy subjects. HA bioavailability in CF without enzyme administration was 0.0292 (0.0192, 0.0459) and with enzymes increased to 0.606 (0.482, 0.823). In CF, compared with taking enzymes with the MBT, HA bioavailability was further decreased by factors of 0.829 (0.664, 0.979) and 0.78 (0.491, 1.13) with enzymes taken 30 and 60 minutes after MBT, respectively. The MBT detected differences in fat absorption in subjects with CF with and without enzyme administration and with changes in enzyme timing. Future studies will address application of the MBT in CF and other malabsorption diagnoses.
Collapse
Affiliation(s)
- Maria R Mascarenhas
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Jeffrey S Barrett
- Division of Clinical Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Sanofi-US, Bridgewater, NJ, USA
| | - Martha Wilson
- Department of Pathology and Lipid Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Virginia A Stallings
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Joan I Schall
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
49
|
Olivier AK, Gibson-Corley KN, Meyerholz DK. Animal models of gastrointestinal and liver diseases. Animal models of cystic fibrosis: gastrointestinal, pancreatic, and hepatobiliary disease and pathophysiology. Am J Physiol Gastrointest Liver Physiol 2015; 308:G459-71. [PMID: 25591863 PMCID: PMC4360044 DOI: 10.1152/ajpgi.00146.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Multiple organ systems, including the gastrointestinal tract, pancreas, and hepatobiliary systems, are affected by cystic fibrosis (CF). Many of these changes begin early in life and are difficult to study in young CF patients. Recent development of novel CF animal models has expanded opportunities in the field to better understand CF pathogenesis and evaluate traditional and innovative therapeutics. In this review, we discuss manifestations of CF disease in gastrointestinal, pancreatic, and hepatobiliary systems of humans and animal models. We also compare the similarities and limitations of animal models and discuss future directions for modeling CF.
Collapse
Affiliation(s)
- Alicia K. Olivier
- Department of Pathology and Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Katherine N. Gibson-Corley
- Department of Pathology and Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K. Meyerholz
- Department of Pathology and Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
50
|
Liu J, Walker NM, Ootani A, Strubberg AM, Clarke LL. Defective goblet cell exocytosis contributes to murine cystic fibrosis-associated intestinal disease. J Clin Invest 2015; 125:1056-68. [PMID: 25642775 DOI: 10.1172/jci73193] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/18/2014] [Indexed: 01/12/2023] Open
Abstract
Cystic fibrosis (CF) intestinal disease is associated with the pathological manifestation mucoviscidosis, which is the secretion of tenacious, viscid mucus that plugs ducts and glands of epithelial-lined organs. Goblet cells are the principal cell type involved in exocytosis of mucin granules; however, little is known about the exocytotic process of goblet cells in the CF intestine. Using intestinal organoids from a CF mouse model, we determined that CF goblet cells have altered exocytotic dynamics, which involved intrathecal granule swelling that was abruptly followed by incomplete release of partially decondensated mucus. Some CF goblet cells exhibited an ectopic granule location and distorted cellular morphology, a phenotype that is consistent with retrograde intracellular granule movement during exocytosis. Increasing the luminal concentration of bicarbonate, which mimics CF transmembrane conductance regulator-mediated anion secretion, increased spontaneous degranulation in WT goblet cells and improved exocytotic dynamics in CF goblet cells; however, there was still an apparent incoordination between granule decondensation and exocytosis in the CF goblet cells. Compared with those within WT goblet cells, mucin granules within CF goblet cells had an alkaline pH, which may adversely affect the polyionic composition of the mucins. Together, these findings indicate that goblet cell dysfunction is an epithelial-autonomous defect in the CF intestine that likely contributes to the pathology of mucoviscidosis and the intestinal manifestations of obstruction and inflammation.
Collapse
|