51
|
Almughem FA, Aldossary AM, Tawfik EA, Alomary MN, Alharbi WS, Alshahrani MY, Alshehri AA. Cystic Fibrosis: Overview of the Current Development Trends and Innovative Therapeutic Strategies. Pharmaceutics 2020; 12:E616. [PMID: 32630625 PMCID: PMC7407299 DOI: 10.3390/pharmaceutics12070616] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic Fibrosis (CF), an autosomal recessive genetic disease, is caused by a mutation in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR). This mutation reduces the release of chloride ions (Cl-) in epithelial tissues, and hyperactivates the epithelial sodium channels (ENaC) which aid in the absorption of sodium ions (Na+). Consequently, the mucus becomes dehydrated and thickened, making it a suitable medium for microbial growth. CF causes several chronic lung complications like thickened mucus, bacterial infection and inflammation, progressive loss of lung function, and ultimately, death. Until recently, the standard of clinical care in CF treatment had focused on preventing and treating the disease complications. In this review, we have summarized the current knowledge on CF pathogenesis and provided an outlook on the current therapeutic approaches relevant to CF (i.e., CFTR modulators and ENaC inhibitors). The enormous potential in targeting bacterial biofilms using antibiofilm peptides, and the innovative therapeutic strategies in using the CRISPR/Cas approach as a gene-editing tool to repair the CFTR mutation have been reviewed. Finally, we have discussed the wide range of drug delivery systems available, particularly non-viral vectors, and the optimal properties of nanocarriers which are essential for successful drug delivery to the lungs.
Collapse
Affiliation(s)
- Fahad A. Almughem
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (F.A.A.); (E.A.T.)
| | - Ahmad M. Aldossary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (A.M.A.); (M.N.A.)
| | - Essam A. Tawfik
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (F.A.A.); (E.A.T.)
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (A.M.A.); (M.N.A.)
| | - Waleed S. Alharbi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha 9088, Saudi Arabia;
| | - Abdullah A. Alshehri
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (F.A.A.); (E.A.T.)
| |
Collapse
|
52
|
Brinkmann F, Hanusch B, Ballmann M, Mayorandan S, Bollenbach A, Chobanyan-Jürgens K, Jansen K, Schmidt-Choudhury A, Derichs N, Tsikas D, Lücke T. Activated L-Arginine/Nitric Oxide Pathway in Pediatric Cystic Fibrosis and Its Association with Pancreatic Insufficiency, Liver Involvement and Nourishment: An Overview and New Results. J Clin Med 2020; 9:jcm9062012. [PMID: 32604946 PMCID: PMC7356307 DOI: 10.3390/jcm9062012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF; OMIM 219700) is a rare genetic disorder caused by a chloride channel defect, resulting in lung disease, pancreas insufficiency and liver impairment. Altered L-arginine (Arg)/nitric oxide (NO) metabolism has been observed in CF patients’ lungs and in connection with malnutrition. The aim of the present study was to investigate markers of the Arg/NO pathway in the plasma and urine of CF patients and to identify possible risk factors, especially associated with malnutrition. We measured the major NO metabolites nitrite and nitrate, Arg, a semi-essential amino acid and NO precursor, the NO synthesis inhibitor asymmetric dimethylarginine (ADMA) and its major urinary metabolite dimethylamine (DMA) in plasma and urine samples of 70 pediatric CF patients and 78 age-matched healthy controls. Biomarkers were determined by gas chromatography–mass spectrometry and high-performance liquid chromatography. We observed higher plasma Arg (90.3 vs. 75.6 µM, p < 0.0001), ADMA (0.62 vs. 0.57 µM, p = 0.03), Arg/ADMA ratio (148 vs. 135, p = 0.01), nitrite (2.07 vs. 1.95 µM, p = 0.03) and nitrate (43.3 vs. 33.1 µM, p < 0.001) concentrations, as well as higher urinary DMA (57.9 vs. 40.7 µM/mM creatinine, p < 0.001) and nitrate (159 vs. 115 µM/mM creatinine, p = 0.001) excretion rates in the CF patients compared to healthy controls. CF patients with pancreatic sufficiency showed plasma concentrations of the biomarkers comparable to those of healthy controls. Malnourished CF patients had lower Arg/ADMA ratios (p = 0.02), indicating a higher NO synthesis capacity in sufficiently nourished CF patients. We conclude that NO production, protein-arginine dimethylation, and ADMA metabolism is increased in pediatric CF patients. Pancreas and liver function influence Arg/NO metabolism. Good nutritional status is associated with higher NO synthesis capacity and lower protein-arginine dimethylation.
Collapse
Affiliation(s)
- Folke Brinkmann
- University Children’s Hospital, Ruhr University, 44791 Bochum, Germany; (F.B.); (M.B.); (K.J.); (A.S.-C.); (T.L.)
| | - Beatrice Hanusch
- University Children’s Hospital, Ruhr University, 44791 Bochum, Germany; (F.B.); (M.B.); (K.J.); (A.S.-C.); (T.L.)
- Correspondence: ; Tel.: +49-234-5092615
| | - Manfred Ballmann
- University Children’s Hospital, Ruhr University, 44791 Bochum, Germany; (F.B.); (M.B.); (K.J.); (A.S.-C.); (T.L.)
- Paediatric Clinic, University Medicine Rostock, 18057 Rostock, Germany
| | - Sebene Mayorandan
- Department of Paediatrics, Hannover Medical School, 30623 Hannover, Germany; (S.M.); (K.C.-J.); (N.D.)
- Department of Paediatrics, University Clinic Münster, 48149 Münster, Germany
| | - Alexander Bollenbach
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30623 Hannover, Germany; (A.B.); (D.T.)
| | - Kristine Chobanyan-Jürgens
- Department of Paediatrics, Hannover Medical School, 30623 Hannover, Germany; (S.M.); (K.C.-J.); (N.D.)
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Department of General Pediatrics, Neuropediatrics, Metabolism, Gastroenterology, Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Pediatric Clinical-Pharmacological Trial Center (paedKliPS), Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Kathrin Jansen
- University Children’s Hospital, Ruhr University, 44791 Bochum, Germany; (F.B.); (M.B.); (K.J.); (A.S.-C.); (T.L.)
| | - Anjona Schmidt-Choudhury
- University Children’s Hospital, Ruhr University, 44791 Bochum, Germany; (F.B.); (M.B.); (K.J.); (A.S.-C.); (T.L.)
| | - Nico Derichs
- Department of Paediatrics, Hannover Medical School, 30623 Hannover, Germany; (S.M.); (K.C.-J.); (N.D.)
- KinderPneumologieDerichs, Pediatric Pneumology and Allergology, CFTR & Pulmonary Research Center, 30173 Hannover, Germany
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30623 Hannover, Germany; (A.B.); (D.T.)
| | - Thomas Lücke
- University Children’s Hospital, Ruhr University, 44791 Bochum, Germany; (F.B.); (M.B.); (K.J.); (A.S.-C.); (T.L.)
| |
Collapse
|
53
|
CFTR deficiency causes cardiac dysplasia during zebrafish embryogenesis and is associated with dilated cardiomyopathy. Mech Dev 2020; 163:103627. [PMID: 32574800 DOI: 10.1016/j.mod.2020.103627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
Mutations in the CFTR gene cause cystic fibrosis (CF) with myocardial dysfunction. However, it remains unknown whether CF-related heart disease is a secondary effect of pulmonary disease, or an intrinsic primary defect in the heart. Here, we used zebrafish, which lack lung tissue, to investigate the role of CFTR in cardiogenesis. Our findings demonstrated that the loss of CFTR impairs cardiac development from the cardiac progenitor stage, resulting in cardiac looping defects, a dilated atrium, pericardial edema, and a decrease in heart rate. Furthermore, we found that cardiac development was perturbed in wild-type embryos treated with a gating-specific CFTR channel inhibitor, CFTRinh-172, at the blastula stage of development, but not at later stages. Gene expression analysis of blastulas indicated that transcript levels, including mRNAs associated with cardiovascular diseases, were significantly altered in embryos derived from cftr mutants relative to controls. To evaluate the role of CFTR in human heart failure, we performed a genetic association study on individuals with dilated cardiomyopathy and found that the I556V mutation in CFTR, which causes a channel defect, was associated with the disease. Similar to other well-studied channel-defective CFTR mutants, CFTR I556V mRNA failed to restore cardiac dysplasia in mutant embryos. The present study revealed an important role for the CFTR ion channel in regulating cardiac development during early embryogenesis, supporting the hypothesis that CF-related heart disease results from an intrinsic primary defect in the heart.
Collapse
|
54
|
Cicali B, Long T, Kim S, Cristofoletti R. Assessing the impact of cystic fibrosis on the antipyretic response of ibuprofen in children: Physiologically-based modeling as a candle in the dark. Br J Clin Pharmacol 2020; 86:2247-2255. [PMID: 32335930 DOI: 10.1111/bcp.14326] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 01/21/2023] Open
Abstract
AIM The goal of this study is to present the utility of quantitative modelling for extrapolation of drug safety and efficacy to underrepresented populations in controlled clinical trials. To illustrate this, the stepwise development of an integrated disease/pharmacokinetics/pharmacodynamics model of antipyretic efficacy of ibuprofen in children with cystic fibrosis (CF) is presented along with therapy optimization suggestions. METHOD Published clinical trials, in vitro data, and drug physiochemical properties were used to develop an ibuprofen-mediated antipyresis model for febrile children also having CF. Workflow included first developing a mechanistic absorption model using in vitro-in vivo extrapolation followed by physiologically-based pharmacokinetic (PBPK) modelling. The verified PBPK model was then scaled to paediatric patients with CF. Once verified, the PBPK model was linked to an indirect response model of antipyresis for simulation of the overall antipyretic efficacy of ibuprofen in CF children. RESULTS Model simulations showed therapeutic inequivalence between healthy children and paediatric patients with CF; Cmax and AUC decreased by 39% (32-46%) and 44% (36-52%), respectively, in patients. Further, and in agreement with literature reports, predicted pharmacodynamics time courses suggest a slower onset and faster offset of action in patients compared to healthy children, 30 and 60 minutes, respectively. Exploratory simulations suggest an increase in dosing frequency for CF children as a better therapeutic strategy. CONCLUSION Model-informed approaches to leveraging knowledge obtained throughout the life cycle of drug development may play a key role in extrapolating drug efficacy and safety to underrepresented populations.
Collapse
Affiliation(s)
- Brian Cicali
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Tao Long
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Sarah Kim
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
55
|
Scott P, Anderson K, Singhania M, Cormier R. Cystic Fibrosis, CFTR, and Colorectal Cancer. Int J Mol Sci 2020; 21:2891. [PMID: 32326161 PMCID: PMC7215855 DOI: 10.3390/ijms21082891] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF), caused by biallelic inactivating mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, has recently been categorized as a familial colorectal cancer (CRC) syndrome. CF patients are highly susceptible to early, aggressive colorectal tumor development. Endoscopic screening studies have revealed that by the age of forty 50% of CF patients will develop adenomas, with 25% developing aggressive advanced adenomas, some of which will have already advanced to adenocarcinomas. This enhanced risk has led to new CF colorectal cancer screening recommendations, lowering the initiation of endoscopic screening to age forty in CF patients, and to age thirty in organ transplant recipients. The enhanced risk for CRC also extends to the millions of people (more than 10 million in the US) who are heterozygous carriers of CFTR gene mutations. Further, lowered expression of CFTR is reported in sporadic CRC, where downregulation of CFTR is associated with poor survival. Mechanisms underlying the actions of CFTR as a tumor suppressor are not clearly understood. Dysregulation of Wnt/β-catenin signaling and disruption of intestinal stem cell homeostasis and intestinal barrier integrity, as well as intestinal dysbiosis, immune cell infiltration, stress responses, and intestinal inflammation have all been reported in human CF patients and in animal models. Notably, the development of new drug modalities to treat non-gastrointestinal pathologies in CF patients, especially pulmonary disease, offers hope that these drugs could be repurposed for gastrointestinal cancers.
Collapse
Affiliation(s)
| | | | | | - Robert Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; (P.S.); (K.A.); (M.S.)
| |
Collapse
|
56
|
Tawfik D, Zaccagnino A, Bernt A, Szczepanowski M, Klapper W, Schwab A, Kalthoff H, Trauzold A. The A818-6 system as an in-vitro model for studying the role of the transportome in pancreatic cancer. BMC Cancer 2020; 20:264. [PMID: 32228510 PMCID: PMC7106758 DOI: 10.1186/s12885-020-06773-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/23/2020] [Indexed: 02/08/2023] Open
Abstract
Background The human pancreatic cancer cell line A818–6 can be grown in vitro either as a highly malignant, undifferentiated monolayer (ML) or as three-dimensional (3D) single layer hollow spheres (HS) simulating a benign, highly differentiated, duct-like pancreatic epithelial structure. This characteristic allowing A818–6 cells to switch from one phenotype to another makes these cells a unique system to characterize the cellular and molecular modifications during differentiation on one hand and malignant transformation on the other hand. Ion channels and transport proteins (transportome) have been implicated in malignant transformation. Therefore, the current study aimed to analyse the transportome gene expression profile in the A818–6 cells growing as a monolayer or as hollow spheres. Methods & Results The study identified the differentially expressed transportome genes in both cellular states of A818–6 using Agilent and Nanostring arrays and some targets were validated via immunoblotting. Additionally, these results were compared to a tissue Affymetrix microarray analysis of pancreatic adenocarcinoma patients’ tissues. The overall transcriptional profile of the ML and HS cells confirmed the formerly described mesenchymal features of ML and epithelial nature of HS which was further verified via high expression of E-cadherin and low expression of vimentin found in HS in comparison to ML. Among the predicted features between HS and ML was the involvement of miRNA-9 in this switch. Importantly, the bioinformatics analysis also revealed substantial number (n = 126) of altered transportome genes. Interestingly, three genes upregulated in PDAC tissue samples (GJB2, GJB5 and SLC38A6) were found to be also upregulated in ML and 3 down-regulated transportome genes (KCNQ1, TRPV6 and SLC4A) were also reduced in ML. Conclusion This reversible HS/ML in vitro system might help in understanding the pathophysiological impact of the transportome in the dedifferentiation process in pancreatic carcinogenesis. Furthermore, the HS/ML model represents a novel system for studying the role of the transportome during the switch from a more benign, differentiated (HS) to a highly malignant, undifferentiated (ML) phenotype.
Collapse
Affiliation(s)
- Doaa Tawfik
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Arnold-Heller Str. 3, 24105, Kiel, Germany
| | - Angela Zaccagnino
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Arnold-Heller Str. 3, 24105, Kiel, Germany
| | - Alexander Bernt
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Arnold-Heller Str. 3, 24105, Kiel, Germany
| | - Monika Szczepanowski
- Clinic for Internal Medicine II, Christian-Albrechts-University of Kiel, UKSH, Kiel, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, Christian-Albrechts-University of Kiel, UKSH, Kiel, Germany
| | - Albrecht Schwab
- Institute of Physiology II, Westfälische Wilhelms-Universität, Münster, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Arnold-Heller Str. 3, 24105, Kiel, Germany
| | - Anna Trauzold
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Arnold-Heller Str. 3, 24105, Kiel, Germany.
| |
Collapse
|
57
|
Paranjapye A, Ruffin M, Harris A, Corvol H. Genetic variation in CFTR and modifier loci may modulate cystic fibrosis disease severity. J Cyst Fibros 2020; 19 Suppl 1:S10-S14. [PMID: 31734115 PMCID: PMC7036019 DOI: 10.1016/j.jcf.2019.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Abstract
In patients with cystic fibrosis (CF), genetic variants within and outside the CFTR locus contribute to the variability of the disease severity. CFTR transcription is tightly regulated by cis-regulatory elements (CREs) that control the three-dimensional structure of the locus, chromatin accessibility and transcription factor recruitment. Variants within these CREs may contribute to the pathophysiology and to the phenotypic heterogeneity by altering CFTR transcript abundance. In addition to the CREs, variants outside the CFTR locus, namely "modifiers genes", may also be associated with the clinical variability. This review addresses variants at the CFTR locus itself and CFTR CREs, together with the outcomes of the latest modifier gene studies with respect to the different CF phenotypes.
Collapse
Affiliation(s)
- Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University Medical School, 10900 Euclid Avenue, Cleveland, OH, USA
| | - Manon Ruffin
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University Medical School, 10900 Euclid Avenue, Cleveland, OH, USA.
| | - Harriet Corvol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France; AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France.
| |
Collapse
|
58
|
Abstract
Cystic fibrosis (CF) is a multisystem disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. CFTR is expressed in the apical surface of cholangiocytes. Homozygous CFTR gene mutation results in viscous and acidic bile secretions secondary to deficient surface fluid and bicarbonate efflux. Viscous, inspissated bile causes ductular obstruction and hepatotoxicity from retained bile components, leading to fibrosis and ultimately cirrhosis, known as CF liver disease (CFLD). CFLD is the third leading cause of death in CF patients. CFLD manifestations can take many forms. They range from asymptomatic elevation of transaminases to cirrhosis and end-stage liver disease. CFLD is diagnosed after excluding other causes of chronic liver disease. To date, there is no effective therapy to prevent or treat CFLD. Management of CFLD emphasizes on optimizing nutritional status. Ursodeoxycholic acid is the only available treatment that may prevent progression of CFLD at present. All CF patients with CFLD need annual investigations and follow-up for early detection of the disease. Liver transplantation should be considered in patients with decompensated cirrhosis and portal hypertension, with acceptable long-term outcomes. Novel therapies of CFLD are promising. This review article aims to summarize the published literature on CFLD, its pathophysiology, clinical features and complications, and management including new therapeutic options.
Collapse
Affiliation(s)
- Siham Al Sinani
- Graduate Medical Education Department, Oman Medical Specialty Board, Muscat, Oman.,Department of Child Health, Sultan Qaboos University Hospital, Muscat, Oman
| | - Sharef Al-Mulaabed
- Department of Pediatrics, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| | | | - Rabab Sultan
- Department of Child Health, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
59
|
Expression of Adenosine Receptors in Rodent Pancreas. Int J Mol Sci 2019; 20:ijms20215329. [PMID: 31717704 PMCID: PMC6862154 DOI: 10.3390/ijms20215329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/18/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Adenosine regulates exocrine and endocrine secretions in the pancreas. Adenosine is considered to play a role in acini-to-duct signaling in the exocrine pancreas. To identify the molecular basis of functional adenosine receptors in the exocrine pancreas, immunohistochemical analysis was performed in the rat, mouse, and guinea pig pancreas, and the secretory rate and concentration of HCO3− in pancreatic juice from the rat pancreas were measured. The A2A adenosine receptor colocalized with ezrin, an A-kinase anchoring protein, in the luminal membrane of duct cells in the mouse and guinea pig pancreas. However, a strong signal ascribed to A2B adenosine receptors was detected in insulin-positive β cells in islets of Langerhans. The A2A adenosine receptor agonist 4-[2-[[6-Amino-9-(N-ethyl-β-D-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl]benzenepropanoic acid (CGS 21680) stimulated HCO3−-rich fluid secretion from the rat pancreas. These results indicate that A2A adenosine receptors may be, at least in part, involved in the exocrine secretion of pancreatic duct cells via acini-to-duct signaling. The adenosine receptors may be a potential therapeutic target for cancer as well as exocrine dysfunctions of the pancreas.
Collapse
|
60
|
Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol 2019; 25:5732-5772. [PMID: 31636470 PMCID: PMC6801186 DOI: 10.3748/wjg.v25.i38.5732] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
In their seminal papers Hanahan and Weinberg described oncogenic processes a normal cell undergoes to be transformed into a cancer cell. The functions of ion channels in the gastrointestinal (GI) tract influence a variety of cellular processes, many of which overlap with these hallmarks of cancer. In this review we focus on the roles of the calcium (Ca2+), sodium (Na+), potassium (K+), chloride (Cl-) and zinc (Zn2+) transporters in GI cancer, with a special emphasis on the roles of the KCNQ1 K+ channel and CFTR Cl- channel in colorectal cancer (CRC). Ca2+ is a ubiquitous second messenger, serving as a signaling molecule for a variety of cellular processes such as control of the cell cycle, apoptosis, and migration. Various members of the TRP superfamily, including TRPM8, TRPM7, TRPM6 and TRPM2, have been implicated in GI cancers, especially through overexpression in pancreatic adenocarcinomas and down-regulation in colon cancer. Voltage-gated sodium channels (VGSCs) are classically associated with the initiation and conduction of action potentials in electrically excitable cells such as neurons and muscle cells. The VGSC NaV1.5 is abundantly expressed in human colorectal CRC cell lines as well as being highly expressed in primary CRC samples. Studies have demonstrated that conductance through NaV1.5 contributes significantly to CRC cell invasiveness and cancer progression. Zn2+ transporters of the ZIP/SLC39A and ZnT/SLC30A families are dysregulated in all major GI organ cancers, in particular, ZIP4 up-regulation in pancreatic cancer (PC). More than 70 K+ channel genes, clustered in four families, are found expressed in the GI tract, where they regulate a range of cellular processes, including gastrin secretion in the stomach and anion secretion and fluid balance in the intestinal tract. Several distinct types of K+ channels are found dysregulated in the GI tract. Notable are hERG1 upregulation in PC, gastric cancer (GC) and CRC, leading to enhanced cancer angiogenesis and invasion, and KCNQ1 down-regulation in CRC, where KCNQ1 expression is associated with enhanced disease-free survival in stage II, III, and IV disease. Cl- channels are critical for a range of cellular and tissue processes in the GI tract, especially fluid balance in the colon. Most notable is CFTR, whose deficiency leads to mucus blockage, microbial dysbiosis and inflammation in the intestinal tract. CFTR is a tumor suppressor in several GI cancers. Cystic fibrosis patients are at a significant risk for CRC and low levels of CFTR expression are associated with poor overall disease-free survival in sporadic CRC. Two other classes of chloride channels that are dysregulated in GI cancers are the chloride intracellular channels (CLIC1, 3 & 4) and the chloride channel accessory proteins (CLCA1,2,4). CLIC1 & 4 are upregulated in PC, GC, gallbladder cancer, and CRC, while the CLCA proteins have been reported to be down-regulated in CRC. In summary, it is clear, from the diverse influences of ion channels, that their aberrant expression and/or activity can contribute to malignant transformation and tumor progression. Further, because ion channels are often localized to the plasma membrane and subject to multiple layers of regulation, they represent promising clinical targets for therapeutic intervention including the repurposing of current drugs.
Collapse
Affiliation(s)
- Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| |
Collapse
|
61
|
Patient-derived pancreas-on-a-chip to model cystic fibrosis-related disorders. Nat Commun 2019; 10:3124. [PMID: 31311920 PMCID: PMC6635497 DOI: 10.1038/s41467-019-11178-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 06/27/2019] [Indexed: 01/23/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder caused by defective CF Transmembrane Conductance Regulator (CFTR) function. Insulin producing pancreatic islets are located in close proximity to the pancreatic duct and there is a possibility of impaired cell-cell signaling between pancreatic ductal epithelial cells (PDECs) and islet cells as causative in CF. To study this possibility, we present an in vitro co-culturing system, pancreas-on-a-chip. Furthermore, we present an efficient method to micro dissect patient-derived human pancreatic ducts from pancreatic remnant cell pellets, followed by the isolation of PDECs. Here we show that defective CFTR function in PDECs directly reduced insulin secretion in islet cells significantly. This uniquely developed pancreatic function monitoring tool will help to study CF-related disorders in vitro, as a system to monitor cell-cell functional interaction of PDECs and pancreatic islets, characterize appropriate therapeutic measures and further our understanding of pancreatic function. Defective CFTR protein, responsible for Cystic Fibrosis (CF), is highly expressed in pancreatic ductal epithelial cells (PDECs) but their impact on insulin secreting pancreatic islets is not fully understood. Here the authors develop a non-CF and CF patient derived pancreas-on-a-chip model to study how CF affects insulin secretion.
Collapse
|
62
|
Layer P, Kashirskaya N, Gubergrits N. Contribution of pancreatic enzyme replacement therapy to survival and quality of life in patients with pancreatic exocrine insufficiency. World J Gastroenterol 2019; 25:2430-2441. [PMID: 31171887 PMCID: PMC6543241 DOI: 10.3748/wjg.v25.i20.2430] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/02/2019] [Accepted: 04/20/2019] [Indexed: 02/06/2023] Open
Abstract
The objective of this study was to analyze the current evidence for the use of pancreatic enzyme replacement therapy (PERT) in affecting survival and quality of life in patients with pancreatic exocrine insufficiency (PEI). Systematic searches of the literature were performed using the PubMed database. Articles were selected for inclusion if they reported findings from trials assessing the effects of PERT on quality of life, survival, malabsorption, growth parameters (such as height, body weight and body mass index), or gastrointestinal symptoms (such as abdominal pain, stool consistency and flatulence). PERT improved PEI-related malabsorption and weight maintenance in patients with cystic fibrosis, chronic pancreatitis, pancreatic cancer, and post-surgical states. In patients with chronic pancreatitis, PERT improved PEI-related symptoms and quality of life measures. Several small retrospective studies have also suggested that PERT may have a positive impact on survival, but long-term studies assessing this effect were not identified. PERT is effective for treating malnutrition and supporting weight maintenance, and it is associated with improved quality of life and possibly with enhanced survival in patients with PEI. However, there is evidence that not all patients with PEI receive adequate PERT. Future work should aim to assess the long-term effects of PERT on the survival of patients with PEI.
Collapse
Affiliation(s)
- Peter Layer
- Department of Internal Medicine, Israelitic Hospital, Hamburg 22297, Germany
| | - Nataliya Kashirskaya
- Department of Genetic Epidemiology (Cystic Fibrosis group), Federal State Budgetary Institution “Research Centre for Medical Genetics”, Moscow 115522, Russia
| | - Natalya Gubergrits
- Department of Internal Medicine, Donetsk National Medical University, Lyman 83001, Ukraine
| |
Collapse
|
63
|
Kelsey R, Manderson Koivula FN, McClenaghan NH, Kelly C. Cystic Fibrosis-Related Diabetes: Pathophysiology and Therapeutic Challenges. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2019; 12:1179551419851770. [PMID: 31191067 PMCID: PMC6539575 DOI: 10.1177/1179551419851770] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022]
Abstract
Cystic fibrosis–related diabetes (CFRD) is among the most common extrapulmonary co-morbidity associated with cystic fibrosis (CF), affecting an estimated 50% of adults with the condition. Cystic fibrosis is prevalent in 1 in every 2500 Caucasian live births and is caused by a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Mutated CFTR leads to dehydrated epithelial surfaces and a build-up of mucus in a variety of tissues including the lungs and pancreas. The leading cause of mortality in CF is repeated respiratory bacterial infections, which prompts a decline in lung function. Co-morbid diabetes promotes bacterial colonisation of the airways and exacerbates the deterioration in respiratory health. Cystic fibrosis–related diabetes is associated with a 6-fold higher mortality rate compared with those with CF alone. The management of CFRD adds a further burden for the patient and creates new therapeutic challenges for the clinical team. Several proposed hypotheses on how CFRD develops have emerged, including exocrine-driven fibrosis and destruction of the entire pancreas and contrasting theories on the direct or indirect impact of CFTR mutation on islet function. The current review outlines recent data on the impact of CFTR on endocrine pancreatic function and discusses the use of conventional diabetic therapies and new CFTR-correcting drugs on the treatment of CFRD.
Collapse
Affiliation(s)
- Ryan Kelsey
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, University of Ulster, Derry/Londonderry, UK
| | - Fiona N Manderson Koivula
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, University of Ulster, Derry/Londonderry, UK
| | | | - Catriona Kelly
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, University of Ulster, Derry/Londonderry, UK
| |
Collapse
|
64
|
Bono-Neri F, Romano C, Isedeh A. Cystic Fibrosis: Advancing Along the Continuum. J Pediatr Health Care 2019; 33:242-254. [PMID: 30529125 DOI: 10.1016/j.pedhc.2018.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/09/2018] [Accepted: 08/19/2018] [Indexed: 12/12/2022]
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder resulting from a mutation in the gene which encodes a cellular transmembrane protein channel known as the CF transmembrane conductance regulator. Located systemically on the surface of numerous cells, these altered channels yield multisystem dysfunction. Typical manifestations seen are chronic, progressive, obstructive lung disease, pancreatic insufficiency, CF-related diabetes mellitus, malabsorption and malnutrition, liver disease, and infertility.Once considered a pediatric disorder, through developments in innovative care and therapeutic modalities, CF now spans the life continuum and has established itself as an ageless disease. Facing management of maturing-life issues, advanced practice nurses (APNs) in pediatrics now find themselves needing to collaborate with or facilitate transition of care to other APNs, such as nurse midwives and adult APNs, as well as their counterpart specialists in medicine, all while maintaining open communication with the patient, family and managing CF center.
Collapse
|
65
|
Norris AW, Ode KL, Merjaneh L, Sanda S, Yi Y, Sun X, Engelhardt JF, Hull RL. Survival in a bad neighborhood: pancreatic islets in cystic fibrosis. J Endocrinol 2019; 241:JOE-18-0468.R1. [PMID: 30759072 PMCID: PMC6675675 DOI: 10.1530/joe-18-0468] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
In cystic fibrosis (CF), ductal plugging and acinar loss result in rapid decline of exocrine pancreatic function. This destructive process results in remodeled islets, with only a modest reduction in insulin producing β cells. However, β-cell function is profoundly impaired, with decreased insulin release and abnormal glucose tolerance being present even in infants with CF. Ultimately, roughly half of CF subjects develop diabetes (termed CF-related diabetes, CFRD). Importantly, CFRD increases CF morbidity and mortality via worsening catabolism and pulmonary disease. Current accepted treatment options for CFRD are aimed at insulin replacement, thereby improving glycemia as well as preventing nutritional losses and lung decline. CFRD is a unique form of diabetes with a distinct pathophysiology that is as yet incompletely understood. Recent studies highlight emerging areas of interest. First, islet inflammation and lymphocyte infiltration are common even in young children with CF and may contribute to β-cell failure. Second, controversy exists in the literature regarding the presence/importance of β-cell intrinsic functions of CFTR and its direct role in modulating insulin release. Third, loss of the CF transmembrane conductance regulator (CFTR) from pancreatic ductal epithelium, the predominant site of its synthesis, results in paracrine effects that impair insulin release. Finally, the degree of β-cell loss in CFRD does not appear sufficient to explain the deficit in insulin release. Thus, it may be possible to enhance the function of the remaining β cells using strategies such as targeting islet inflammation or ductal CFTR deficiency to effectively treat or even prevent CFRD.
Collapse
Affiliation(s)
- Andrew W. Norris
- Department of Pediatrics, University of Iowa, Iowa City, Iowa 52242
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242
| | - Katie Larson Ode
- Department of Pediatrics, University of Iowa, Iowa City, Iowa 52242
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242
| | - Lina Merjaneh
- Division of Endocrinology & Diabetes, Seattle Children’s Hospital, Seattle, Washington 98105
| | - Srinath Sanda
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
- Diabetes Center, University of California San Francisco, San Francisco, CA
| | - Yaling Yi
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242
| | - John F. Engelhardt
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242
| | - Rebecca L. Hull
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
66
|
Ye R, Onodera T, Blanchard PG, Kusminski CM, Esser V, Brekken RA, Scherer PE. β1 Syntrophin Supports Autophagy Initiation and Protects against Cerulein-Induced Acute Pancreatitis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:813-825. [PMID: 30653956 DOI: 10.1016/j.ajpath.2019.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/26/2018] [Accepted: 01/02/2019] [Indexed: 01/23/2023]
Abstract
Syntrophins are a family of proteins forming membrane-anchored scaffolds and serving as adaptors for various transmembrane and intracellular signaling molecules. To understand the physiological roles of β1 syntrophin, one of the least characterized members, we generated mouse models to eliminate β1 syntrophin specifically in the endocrine or exocrine pancreas. β1 syntrophin is dispensable for the morphology and function of insulin-producing β cells. However, mice with β1 syntrophin deletion in exocrine acinar cells exhibit increased severity of cerulein-induced acute pancreatitis. Reduced expression of cystic fibrosis transmembrane conductance regulator and dilation of acinar lumen are potential predisposition factors. During the disease progression, a relative lack of autophagy is associated with deficiencies in both actin assembly and endoplasmic reticulum nucleation. Our findings reveal, for the first time, that β1 syntrophin is a critical regulator of actin cytoskeleton and autophagy in pancreatic acinar cells and is potently protective against cerulein-induced acute pancreatitis.
Collapse
Affiliation(s)
- Risheng Ye
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Medical Education, Texas Tech University Health Sciences Center Paul L. Foster School of Medicine, El Paso, Texas
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Pierre-Gilles Blanchard
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Victoria Esser
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
67
|
Semaniakou A, Croll RP, Chappe V. Animal Models in the Pathophysiology of Cystic Fibrosis. Front Pharmacol 2019; 9:1475. [PMID: 30662403 PMCID: PMC6328443 DOI: 10.3389/fphar.2018.01475] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/03/2018] [Indexed: 01/28/2023] Open
Abstract
Our understanding of the multiorgan pathology of cystic fibrosis (CF) has improved impressively during the last decades, but we still lack a full comprehension of the disease progression. Animal models have greatly contributed to the elucidation of specific mechanisms involved in CF pathophysiology and the development of new therapies. Soon after the cloning of the CF transmembrane conductance regulator (CFTR) gene in 1989, the first mouse model was generated and this model has dominated in vivo CF research ever since. Nonetheless, the failure of murine models to mirror human disease severity in the pancreas and lung has led to the generation of larger animal models such as pigs and ferrets. The following review presents and discusses data from the current animal models used in CF research.
Collapse
Affiliation(s)
- Anna Semaniakou
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Valerie Chappe
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
68
|
Madácsy T, Pallagi P, Maleth J. Cystic Fibrosis of the Pancreas: The Role of CFTR Channel in the Regulation of Intracellular Ca 2+ Signaling and Mitochondrial Function in the Exocrine Pancreas. Front Physiol 2018; 9:1585. [PMID: 30618777 PMCID: PMC6306458 DOI: 10.3389/fphys.2018.01585] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
Cystic fibrosis (CF) is the most common genetic disorder that causes a significant damage in secretory epithelial cells due to the defective ion flux across the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel. Pancreas is one of the organs most frequently damaged by the disease leading to pancreatic insufficiency, abdominal pain and an increased risk of acute pancreatitis in CF patients causing a significant decrease in the quality of life. CFTR plays a central role in the pancreatic ductal secretory functions by carrying Cl- and HCO3 - ions across the apical membrane. Therefore pathophysiological studies in CF mostly focused on the effects of impaired ion secretion by pancreatic ductal epithelial cells leading to exocrine pancreatic damage. However, several studies indicated that CFTR has a central role in the regulation of intracellular signaling processes and is now more widely considered as a signaling hub in epithelial cells. In contrast, elevated intracellular Ca2+ level was observed in the lack of functional CFTR in different cell types including airway epithelial cells. In addition, impaired CFTR expression has been correlated with damaged mitochondrial function in epithelial cells. These alterations of intracellular signaling in CF are not well characterized in the exocrine pancreas yet. Therefore in this review we would like to summarize the complex role of CFTR in the exocrine pancreas with a special focus on the intracellular signaling and mitochondrial function.
Collapse
Affiliation(s)
- Tamara Madácsy
- First Department of Medicine, University of Szeged, Szeged, Hungary.,HAS-USZ Momentum Epithel Cell Signalling and Secretion Research Group, Szeged, Hungary
| | - Petra Pallagi
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Jozsef Maleth
- First Department of Medicine, University of Szeged, Szeged, Hungary.,HAS-USZ Momentum Epithel Cell Signalling and Secretion Research Group, Szeged, Hungary.,Department of Public Health, University of Szeged, Szeged, Hungary
| |
Collapse
|
69
|
Conese M, Beccia E, Carbone A, Castellani S, Di Gioia S, Corti F, Angiolillo A, Colombo C. The role of stem cells in cystic fibrosis disease modeling and drug discovery. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1549480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Massimo Conese
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elisa Beccia
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Annalucia Carbone
- Division of Internal Medicine and Chronobiology Unit, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo (FG), Italy
| | - Stefano Castellani
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Fabiola Corti
- Department of Pathophysiology and Transplantation, Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Antonella Angiolillo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Carla Colombo
- Department of Pathophysiology and Transplantation, Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
70
|
Webster MJ, Tarran R. Slippery When Wet: Airway Surface Liquid Homeostasis and Mucus Hydration. CURRENT TOPICS IN MEMBRANES 2018; 81:293-335. [PMID: 30243435 DOI: 10.1016/bs.ctm.2018.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability to regulate cell volume is crucial for normal physiology; equally the regulation of extracellular fluid homeostasis is of great importance. Alteration of normal extracellular fluid homeostasis contributes to the development of several diseases including cystic fibrosis. With regard to the airway surface liquid (ASL), which lies apically on top of airway epithelia, ion content, pH, mucin and protein abundance must be tightly regulated. Furthermore, airway epithelia must be able to switch from an absorptive to a secretory state as required. A heterogeneous population of airway epithelial cells regulate ASL solute and solvent composition, and directly secrete large mucin molecules, antimicrobials, proteases and soluble mediators into the airway lumen. This review focuses on how epithelial ion transport influences ASL hydration and ASL pH, with a specific focus on the roles of anion and cation channels and exchangers. The role of ions and pH in mucin expansion is also addressed. With regard to fluid volume regulation, we discuss the roles of nucleotides, adenosine and the short palate lung and nasal epithelial clone 1 (SPLUNC1) as soluble ASL mediators. Together, these mechanisms directly influence ciliary beating and in turn mucociliary clearance to maintain sterility and to detoxify the airways. Whilst all of these components are regulated in normal airways, defective ion transport and/or mucin secretion proves detrimental to lung homeostasis as such we address how defective ion and fluid transport, and a loss of homeostatic mechanisms, contributes to the development of pathophysiologies associated with cystic fibrosis.
Collapse
Affiliation(s)
- Megan J Webster
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Robert Tarran
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
71
|
O'Neal WK, Knowles MR. Cystic Fibrosis Disease Modifiers: Complex Genetics Defines the Phenotypic Diversity in a Monogenic Disease. Annu Rev Genomics Hum Genet 2018; 19:201-222. [DOI: 10.1146/annurev-genom-083117-021329] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In many respects, genetic studies in cystic fibrosis (CF) serve as a paradigm for a human Mendelian genetic success story. From recognition of the condition as a heritable pathological entity to implementation of personalized treatments based on genetic findings, this multistep pathway of progress has focused on the genetic underpinnings of CF clinical disease. Along this path was the recognition that not all CFTR gene mutations produce the same disease and the recognition of the complex, multifactorial nature of CF genotype–phenotype relationships. The non- CFTR genetic components (gene modifiers) that contribute to variation in phenotype are the focus of this review. A multifaceted approach involving candidate gene studies, genome-wide association studies, and gene expression studies has revealed significant gene modifiers for multiple CF phenotypes. The bold challenges for the future are to integrate the findings into our understanding of CF pathogenesis and to use the knowledge to develop novel therapies.
Collapse
Affiliation(s)
- Wanda K. O'Neal
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;,
| | - Michael R. Knowles
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;,
| |
Collapse
|
72
|
Principe DR, Overgaard NH, Park AJ, Diaz AM, Torres C, McKinney R, Dorman MJ, Castellanos K, Schwind R, Dawson DW, Rana A, Maker A, Munshi HG, Rund LA, Grippo PJ, Schook LB. KRAS G12D and TP53 R167H Cooperate to Induce Pancreatic Ductal Adenocarcinoma in Sus scrofa Pigs. Sci Rep 2018; 8:12548. [PMID: 30135483 PMCID: PMC6105629 DOI: 10.1038/s41598-018-30916-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022] Open
Abstract
Although survival has improved in recent years, the prognosis of patients with advanced pancreatic ductal adenocarcinoma (PDAC) remains poor. Despite substantial differences in anatomy, physiology, genetics, and metabolism, the overwhelming majority of preclinical testing relies on transgenic mice. Hence, while mice have allowed for tremendous advances in cancer biology, they have been a poor predictor of drug performance/toxicity in the clinic. Given the greater similarity of sus scrofa pigs to humans, we engineered transgenic sus scrofa expressing a LSL-KRASG12D-TP53R167H cassette. By applying Adeno-Cre to pancreatic duct cells in vitro, cells self-immortalized and established tumors in immunocompromised mice. When Adeno-Cre was administered to the main pancreatic duct in vivo, pigs developed extensive PDAC at the injection site hallmarked by excessive proliferation and desmoplastic stroma. This serves as the first large animal model of pancreatic carcinogenesis, and may allow for insight into new avenues of translational research not before possible in rodents.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nana Haahr Overgaard
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Copenhagen, Denmark
- University of Illinois Department of Animal Sciences, Urbana-Champaign, IL, USA
| | - Alex J Park
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew M Diaz
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Carolina Torres
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ronald McKinney
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthew J Dorman
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Karla Castellanos
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Regina Schwind
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - David W Dawson
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | - Ajay Maker
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Lauretta A Rund
- University of Illinois Department of Animal Sciences, Urbana-Champaign, IL, USA
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| | - Lawrence B Schook
- University of Illinois Department of Animal Sciences, Urbana-Champaign, IL, USA.
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
73
|
Arda HE, Tsai J, Rosli YR, Giresi P, Bottino R, Greenleaf WJ, Chang HY, Kim SK. A Chromatin Basis for Cell Lineage and Disease Risk in the Human Pancreas. Cell Syst 2018; 7:310-322.e4. [PMID: 30145115 DOI: 10.1016/j.cels.2018.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/28/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022]
Abstract
Understanding the genomic logic that underlies cellular diversity and developmental potential in the human pancreas will accelerate the growth of cell replacement therapies and reveal genetic risk mechanisms in diabetes. Here, we identified and characterized thousands of chromatin regions governing cell-specific gene regulation in human pancreatic endocrine and exocrine lineages, including islet β cells, α cells, duct, and acinar cells. Our findings have captured cellular ontogenies at the chromatin level, identified lineage-specific regulators potentially acting on these sites, and uncovered hallmarks of regulatory plasticity between cell types that suggest mechanisms to regenerate β cells from pancreatic endocrine or exocrine cells. Our work shows that disease risk variants related to pancreas are significantly enriched in these regulatory regions and reveals previously unrecognized links between endocrine and exocrine pancreas in diabetes risk.
Collapse
Affiliation(s)
- H Efsun Arda
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA
| | - Jennifer Tsai
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA
| | - Yenny R Rosli
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA
| | - Paul Giresi
- Center for Personal Dynamic Regulomes, 269 Campus Drive CCSR 2145, Stanford, CA 94305, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, 279 Campus Drive West Beckman Center, B-257, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, 269 Campus Drive CCSR 2145, Stanford, CA 94305, USA; Program in Epithelial Biology, Stanford University School of Medicine, 269 Campus Drive CCSR 2145, Stanford, CA 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
74
|
Deconstructing the principles of ductal network formation in the pancreas. PLoS Biol 2018; 16:e2002842. [PMID: 30048442 PMCID: PMC6080801 DOI: 10.1371/journal.pbio.2002842] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/07/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022] Open
Abstract
The mammalian pancreas is a branched organ that does not exhibit stereotypic branching patterns, similarly to most other glands. Inside branches, it contains a network of ducts that undergo a transition from unconnected microlumen to a mesh of interconnected ducts and finally to a treelike structure. This ductal remodeling is poorly understood, both on a microscopic and macroscopic level. In this article, we quantify the network properties at different developmental stages. We find that the pancreatic network exhibits stereotypic traits at each stage and that the network properties change with time toward the most economical and optimized delivery of exocrine products into the duodenum. Using in silico modeling, we show how steps of pancreatic network development can be deconstructed into two simple rules likely to be conserved for many other glands. The early stage of the network is explained by noisy, redundant duct connection as new microlumens form. The later transition is attributed to pruning of the network based on the flux of fluid running through the pancreatic network into the duodenum. In the pancreas of mammals, digestive enzymes are transported from their production site in acini (clusters of cells that secrete the enzymes) to the intestine via a network of ducts. During organ development in fetuses, the ducts initially form by the coordinated polarization of cells to form small holes, which will connect and fuse, to constitute a meshwork. This hyperconnected network further develops into a treelike structure by the time of birth. In this article, we use methods originally developed to analyze road, rail, web, or river networks to quantify the network properties at different developmental stages. We find that the pancreatic network properties are similar between individuals at specific time points but eventually change to achieve the most economical and optimized structure to deliver pancreatic juice into the duodenum. Using in silico modeling, we show how the stages of pancreatic network development follow two simple rules, which are likely to be conserved for the development of other glands. The early stage of the network is explained by noisy, redundant duct connection as new small ductal holes form. Later on, the secretion of fluid that runs through the pancreatic network into the duodenum leads to the widening of ducts with the greatest flow, while nonnecessary ducts are eliminated, akin to how river beds are formed.
Collapse
|
75
|
Moore PJ, Tarran R. The epithelial sodium channel (ENaC) as a therapeutic target for cystic fibrosis lung disease. Expert Opin Ther Targets 2018; 22:687-701. [PMID: 30028216 DOI: 10.1080/14728222.2018.1501361] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Cystic fibrosis is an autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that codes for the CFTR anion channel. In the absence of functional CFTR, the epithelial Na+ channel is also dysregulated. Airway surface liquid (ASL) hydration is maintained by a balance between epithelial sodium channel (ENaC)-led Na+ absorption and CFTR-dependent anion secretion. This finely tuned homeostatic mechanism is required to maintain sufficient airway hydration to permit the efficient mucus clearance necessary for a sterile lung environment. In CF airways, the lack of CFTR and increased ENaC activity lead to ASL/mucus dehydration that causes mucus obstruction, neutrophilic infiltration, and chronic bacterial infection. Rehydration of ASL/mucus in CF airways can be achieved by inhibiting Na+ absorption with pharmacological inhibitors of ENaC. Areas covered: In this review, we discuss ENaC structure and function and its role in CF lung disease and focus on ENaC inhibition as a potential therapeutic target to rehydrate CF mucus. We also discuss the failure of the first generation of pharmacological inhibitors of ENaC and recent alternate strategies to attenuate ENaC activity in the CF lung. Expert opinion: ENaC is an attractive therapeutic target to rehydrate CF ASL that may serve as a monotherapy or function in parallel with other treatments. Given the increased number of strategies being employed to inhibit ENaC, this is an exciting and optimistic time to be in this field.
Collapse
Affiliation(s)
- Patrick J Moore
- a Marsico Lung Institute , University of North Carolina , Chapel Hill , NC , USA
| | - Robert Tarran
- a Marsico Lung Institute , University of North Carolina , Chapel Hill , NC , USA.,b Department of Cell Biology & Physiology , University of North Carolina , Chapel Hill , NC , USA
| |
Collapse
|
76
|
Amaral MD, Boj SF, Shaw J, Leipziger J, Beekman JM. Cystic fibrosis: Beyond the airways. Report on the meeting of the basic science working group in Loutraki, Greece. J Cyst Fibros 2018; 17:441-443. [PMID: 29866530 DOI: 10.1016/j.jcf.2018.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 11/16/2022]
Abstract
The European Cystic Fibrosis Society (ECFS) Basic Science Working Group (BSWG) organized a session on the topic "Cystic Fibrosis: Beyond the Airways", within the 15th ECFS Basic Science Conference which gathered around 200 researchers working in the basic science of CF. The session was organized and chaired by Margarida Amaral (BioISI, University of Lisboa, Portugal) and Jeffrey Beekman (University Medical Centre Utrecht, Netherlands) as Chair and Vice-Chair of the BSWG and its purpose was to bring attention of participants of the ECFS Basic Science Conference to "more forgotten" organs in CF disease. In this report we attempt to review and integrate the ideas that emerged at the session.
Collapse
Affiliation(s)
- Margarida D Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal.
| | - Sylvia F Boj
- Hubrecht Organoid Technology, Yalelaan 62, 3584 CM Utrecht, The Netherlands
| | - James Shaw
- Institute of Cellular Medicine, Newcastle University Institute, The Medical School, 4(th) Floor William Leech Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Jens Leipziger
- Department of Biomedicine, Physiology and Biophysics, University of Aahrus, Ole Worms Alle 3, build 1170, 8000 Aarhus, Denmark
| | - Jeffrey M Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
77
|
CFTR mutation enhances Dishevelled degradation and results in impairment of Wnt-dependent hematopoiesis. Cell Death Dis 2018; 9:275. [PMID: 29449653 PMCID: PMC5833403 DOI: 10.1038/s41419-018-0311-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 01/16/2023]
Abstract
Mutations of cystic fibrosis transmembrane conductance regulator (CFTR) cause cystic fibrosis (CF) with a multitude of clinical manifestations. Some CF patients develop clinically significant anemia, suggesting that CFTR may regulate hematopoiesis. Here, we report that cftr mutant zebrafish model exhibits primitive and definitive hematopoietic defects with impaired Wnt signaling. Cftr is found to interact, via its PDZ-binding domain (PDZBD), with Dishevelled (Dvl), a key component of Wnt signaling required for hematopoietic progenitor specification, thus protecting Dvl from Dapper1 (Dpr1)-induced lysosomal degradation. Defective hematopoiesis and impaired Wnt signaling in cftr mutant can be rescued by overexpression of wild-type or channel function-defective G551D mutant CFTR with an intact PDZBD, but not Cftr with mutations in the PDZBD. Analysis of human database (http://r2.amc.nl) shows that CFTR is positively correlated with DVL2 and Wnt-related hematopoietic factors in human blood system. The results reveal a previously unrecognized role of CFTR, which is independent of its channel function, in regulating DVL degradation and thus Wnt signaling required for hematopoiesis in both zebrafish and humans, providing an explanation for the anemic phenotype of CF patients.
Collapse
|
78
|
Andharia N, Hayashi M, Matsuda H. Electrophysiological properties of anion exchangers in the luminal membrane of guinea pig pancreatic duct cells. Pflugers Arch 2018; 470:897-907. [PMID: 29399744 PMCID: PMC5945753 DOI: 10.1007/s00424-018-2116-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/10/2018] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
The pancreatic duct epithelium secretes the HCO3−-rich pancreatic juice. The HCO3− transport across the luminal membrane has been proposed to be mediated by SLC26A Cl−–HCO3− exchangers. To examine the electrophysiological properties of Cl−–HCO3− exchangers, we directly measured HCO3− conductance in the luminal membrane of the interlobular pancreatic duct cells from guinea pigs using an inside-out patch-clamp technique. Intracellular HCO3− increased the HCO3− conductance with a half-maximal effective concentration value of approximately 30 mM. The selectivity sequence based on permeability ratios was SCN− (1.4) > Cl− (1.2) = gluconate (1.1) = I− (1.1) = HCO3− (1.0) > methanesulfonate (0.6). The sequence of the relative conductance was HCO3− (1.0) > SCN− (0.7) = I− (0.7) > Cl− (0.5) = gluconate (0.4) > methanesulfonate (0.2). The current dependent on intracellular HCO3− was reduced by replacement of extracellular Cl− with gluconate or by H2DIDS, an inhibitor of Cl−–HCO3− exchangers. RT-PCR analysis revealed that the interlobular and main ducts expressed all SLC26A family members except Slc26a5 and Slc26a8. SLC26A1, SLC26A4, SLC26A6, and SLC26A10 were found to be localized to the luminal membrane of the guinea pig pancreatic duct by immunohistochemistry. These results demonstrate that these SLC26A Cl−–HCO3− exchangers may mediate the electrogenic HCO3− transport through the luminal membrane and may be involved in pancreatic secretion in guinea pig ducts.
Collapse
Affiliation(s)
- N Andharia
- Department of Physiology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, 573-1010, Japan
| | - M Hayashi
- Department of Physiology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, 573-1010, Japan.
| | - H Matsuda
- Department of Physiology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, 573-1010, Japan
| |
Collapse
|
79
|
Rotti PG, Xie W, Poudel A, Yi Y, Sun X, Tyler SR, Uc A, Norris AW, Hara M, Engelhardt JF, Gibson-Corley KN. Pancreatic and Islet Remodeling in Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Knockout Ferrets. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:876-890. [PMID: 29366680 DOI: 10.1016/j.ajpath.2017.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/05/2017] [Accepted: 12/28/2017] [Indexed: 12/25/2022]
Abstract
In cystic fibrosis (CF), there is early destruction of the exocrine pancreas, and this results in a unique form of diabetes that affects approximately half of adult CF individuals. An animal model of cystic fibrosis-related diabetes has been developed in the ferret, which progresses through phases of glycemic abnormalities because of islet remodeling during and after exocrine destruction. Herein, we quantified the pancreatic histopathological changes that occur during these phases. There was an increase in percentage ductal, fat, and islet area in CF ferrets over time compared with age-matched wild-type controls. We also quantified islet size, shape, islet cell composition, cell proliferation (Ki-67), and expression of remodeling markers (matrix metalloprotease-7, desmin, and α-smooth muscle actin). Pancreatic ducts were dilated with scattered proliferating cells and were surrounded by activated stellate cells, indicative of tissue remodeling. The timing of islet and duct proliferation, stellate cell activation, and matrix remodeling coincided with the previously published stages of glycemic crisis and inflammation. This mapping of remodeling events in the CF ferret pancreas provides insights into early changes that control glycemic intolerance and subsequent recovery during the evolution of CF pancreatic disease.
Collapse
Affiliation(s)
- Pavana G Rotti
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa; Department of Biomedical Engineering, The University of Iowa, Iowa City, Iowa
| | - Weiliang Xie
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Ananta Poudel
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yaling Yi
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Scott R Tyler
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Aliye Uc
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Andrew W Norris
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | | |
Collapse
|
80
|
Abstract
Cystic fibrosis (CF) is the most common autosomal recessive disorder in Caucasian populations. Individuals with CF have seen significant increases in life expectancy in the last 60 years. As a result, previously rare complications are now coming to light. The most common of these is cystic fibrosis-related diabetes (CFRD), which affects 40-50% of CF adults. CFRD significantly impacts the pulmonary function and longevity of CF patients, yet a lack of consensus on the best methods to diagnose and treat CFRD remains. We begin by reviewing our understanding of the pathogenesis of CFRD, as emerging evidence shows the cystic fibrosis transmembrane conductance regulator (CFTR) also has important roles in the release of insulin and glucagon and in the protection of β cells from oxidative stress. We then discuss how current recommended methods of CFRD diagnosis are not appropriate, as continuous glucose monitoring becomes more effective, practical, and cost-effective. Finally, we evaluate emerging treatments which have narrowed the mortality gap within the CF patient group. In the future, pharmacological potentiators and correctors directly targeting CFTR show huge promise for both CFRD and the wider CF patient groups.
Collapse
Affiliation(s)
- Kayani Kayani
- Faculty of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Raihan Mohammed
- Faculty of Medicine, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Raihan Mohammed,
| | - Hasan Mohiaddin
- Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
81
|
Conese M, Beccia E, Castellani S, Di Gioia S, Colombo C, Angiolillo A, Carbone A. The long and winding road: stem cells for cystic fibrosis. Expert Opin Biol Ther 2017; 18:281-292. [PMID: 29216777 DOI: 10.1080/14712598.2018.1413087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic syndrome with a high mortality rate due to severe lung disease. Despite having several drugs targeting specific mutated CFTR proteins already in clinical trials, new therapies, based on stem cells, are also emerging to treat those patients. AREAS COVERED The authors review the main sources of stem cells, including embryonic stem cells (ESCs), induced-pluripotent stem cells (iPSCs), gestational stem cells, and adult stem cells, such as mesenchymal stem cells (MSCs) in the context of CF. Furthermore, they describe the main animal and human models of lung physiology and pathology, involved in the optimization of these stem cell-applied therapies in CF. EXPERT OPINION ESCs and iPSCs are emerging sources for disease modeling and drug discovery purposes. The allogeneic transplant of healthy MSCs, that acts independently to specific mutations, is under intense scrutiny due to their secretory, immunomodulatory, anti-inflammatory and anti-bacterial properties. The main challenge for future developments will be to get exogenous stem cells into the appropriate lung location, where they can regenerate endogenous stem cells and act as inflammatory modulators. The clinical application of stem cells for the treatment of CF certainly warrants further insight into pre-clinical models, including large animals, organoids, decellularized organs and lung bioengineering.
Collapse
Affiliation(s)
- Massimo Conese
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Elisa Beccia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy.,b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Stefano Castellani
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Sante Di Gioia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Carla Colombo
- c Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation , University of Milan , Milan , Italy
| | - Antonella Angiolillo
- b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Annalucia Carbone
- d Division of Internal Medicine and Chronobiology Unit , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo (FG) , Italy
| |
Collapse
|
82
|
Yoon JC. Evolving Mechanistic Views and Emerging Therapeutic Strategies for Cystic Fibrosis-Related Diabetes. J Endocr Soc 2017; 1:1386-1400. [PMID: 29264462 PMCID: PMC5686691 DOI: 10.1210/js.2017-00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/25/2017] [Indexed: 12/19/2022] Open
Abstract
Diabetes is a common and important complication of cystic fibrosis, an autosomal recessive genetic disease due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Cystic fibrosis-related diabetes (CFRD) is associated with profound detrimental effects on the disease course and mortality and is expected to increase in prevalence as the survival of patients with cystic fibrosis continues to improve. Despite progress in the functional characterization of CFTR molecular defects, the mechanistic basis of CFRD is not well understood, in part because of the relative inaccessibility of the pancreatic tissue and the limited availability of representative animal models. This review presents a concise overview of the current understanding of CFRD pathogenesis and provides a cutting-edge update on novel findings from human and animal studies. Potential contributions from paracrine mechanisms and β-cell compensatory mechanisms are highlighted, as well as functional β-cell and α-cell defects, incretin defects, exocrine pancreatic insufficiency, and loss of islet cell mass. State-of-the-art and emerging treatment options are explored, including advances in insulin administration, CFTR modulators, cell replacement, gene replacement, and gene editing therapies.
Collapse
Affiliation(s)
- John C Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616
| |
Collapse
|
83
|
Hohwieler M, Perkhofer L, Liebau S, Seufferlein T, Müller M, Illing A, Kleger A. Stem cell-derived organoids to model gastrointestinal facets of cystic fibrosis. United European Gastroenterol J 2017; 5:609-624. [PMID: 28815024 PMCID: PMC5548342 DOI: 10.1177/2050640616670565] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/25/2016] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) is one of the most frequently occurring inherited human diseases caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) which lead to ample defects in anion transport and epithelial fluid secretion. Existing models lack both access to early stages of CF development and a coeval focus on the gastrointestinal CF phenotypes, which become increasingly important due increased life span of the affected individuals. Here, we provide a comprehensive overview of gastrointestinal facets of CF and the opportunity to model these in various systems in an attempt to understand and treat CF. A particular focus is given on forward-leading organoid cultures, which may circumvent current limitations of existing models and thereby provide a platform for drug testing and understanding of disease pathophysiology in gastrointestinal organs.
Collapse
Affiliation(s)
- Meike Hohwieler
- Department of Internal Medicine 1, University Medical Centre Ulm, Ulm, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine 1, University Medical Centre Ulm, Ulm, Germany
| | - Stefan Liebau
- Institute of Neuroanatomy, Eberhard Karls University Tuebingen, Oesterbergstr. 3, 72074 Tuebingen, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine 1, University Medical Centre Ulm, Ulm, Germany
| | - Martin Müller
- Department of Internal Medicine 1, University Medical Centre Ulm, Ulm, Germany
| | - Anett Illing
- Department of Internal Medicine 1, University Medical Centre Ulm, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine 1, University Medical Centre Ulm, Ulm, Germany
| |
Collapse
|
84
|
Pedersen SF, Novak I, Alves F, Schwab A, Pardo LA. Alternating pH landscapes shape epithelial cancer initiation and progression: Focus on pancreatic cancer. Bioessays 2017; 39. [DOI: 10.1002/bies.201600253] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Stine F. Pedersen
- Section for Cell Biology and Physiology; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - Ivana Novak
- Section for Cell Biology and Physiology; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - Frauke Alves
- Max Planck Institute of Experimental Medicine; Göttingen Germany
- Institute for Diagnostic and Interventional Radiology; University Medical Center; Göttingen Germany
- Department of Hematology and Medical Oncology; University Medical Center; Göttingen Germany
| | - Albrecht Schwab
- Institute of Physiology II; University of Münster; Münster Germany
| | - Luis A. Pardo
- Max Planck Institute of Experimental Medicine; Göttingen Germany
| |
Collapse
|
85
|
Abstract
OBJECTIVES Caffeine is contained in coffee, tea, and numerous beverages and foods. We examined the direct effects of caffeine on the physiological function of pancreatic duct cells by using interlobular duct segments isolated from guinea pig pancreas. METHODS The rate of fluid secretion was continuously measured by monitoring the luminal volume of isolated duct segments. Changes in intracellular Ca concentration ([Ca]i) were estimated by microfluorometry in ducts loaded with Fura-2. RESULTS Both secretin-stimulated and acetylcholine (ACh)-stimulated fluid secretions were substantially and reversibly inhibited by relatively low concentrations of caffeine as low as 0.03 mM relevant to blood levels after ingestion of caffeine-containing beverages. Caffeine inhibited ACh-induced elevation of [Ca]i and secretin-induced fluctuation of [Ca]i. Caffeine abolished thapsigargin-induced intracellular Ca release but did not affect the entry of extracellular Ca. Caffeine (0.05 mM) abolished ethanol (1 mM)-induced fluid hypersecretion in secretin-stimulated pancreatic duct. CONCLUSIONS Low concentrations of caffeine directly inhibit pancreatic ductal fluid secretion stimulated by secretin or ACh and also ethanol-induced fluid hypersecretion. The inhibition by caffeine seems to be mediated by the blockade of intracellular Ca mobilization. Daily intake of caffeine may reduce the volume of pancreatic juice secretion.
Collapse
|
86
|
Abstract
OBJECTIVES The aim of this study was to determine if comprehensive genetic testing was useful to identify genetic variants that discriminate chronic pancreatitis (CP) from acute recurrent pancreatitis (ARP) in a pediatric population. METHODS We conducted a retrospective review of 50 patients enrolled in our institutional pancreatitis registry between April 2013 and January 2015. Genetic analysis of PRSS1, CFTR, SPINK1, and CTRC classified variants as mutations or variants of unknown clinical significance and the minor allele frequency of variants in our cohort was obtained. RESULTS Genetic testing was obtained in 16/16 (100%) of CP and 29/34 (85%) of ARP patients. A total of 39 genetic variants were found in 27 (60%) of 45 subjects tested with 5 (11%) subjects having 2 different genes affected. Variant frequency was greatest in patients for CFTR (17/45, 38%) followed by SPINK1 (11/44, 25%), CTRC (2/27, 7%), and PRSS1 (2/44, 4%). CFTR variants were more likely in those with CP compared to ARP (63% and 24%, P = 0.01). CONCLUSIONS This study is the first to find a higher rate of CFTR mutations in CP versus ARP groups using comprehensive genetic testing in a pediatric population.
Collapse
|
87
|
Zaccagnino A, Pilarsky C, Tawfik D, Sebens S, Trauzold A, Novak I, Schwab A, Kalthoff H. In silico analysis of the transportome in human pancreatic ductal adenocarcinoma. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:749-763. [PMID: 27652669 DOI: 10.1007/s00249-016-1171-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/18/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022]
Abstract
The altered expression and/or activity of ion channels and transporters (transportome) have been associated with malignant behavior of cancer cells and were proposed to be a hallmark of cancer. However, the impact of altered transportome in epithelial cancers, such as pancreatic ductal adenocarcinoma (PDAC), as well as its pathophysiological consequences, still remains unclear. Here, we report the in silico analysis of 840 transportome genes in PDAC patients' tissues. Our study was focused on the transportome changes and their correlation with functional and behavioral responses in PDAC tumor and stromal compartments. The dysregulated gene expression datasets were filtered using a cut-off of fold-change values ≤-2 or ≥2 (adjusted p value ≤0.05). The dysregulated transportome genes were clearly associated with impaired physiological secretory mechanisms and/or pH regulation, control of cell volume, and cell polarity. Additionally, some down-regulated transportome genes were found to be closely linked to epithelial cell differentiation. Furthermore, the observed decrease in genes coding for calcium and chloride transport might be a mechanism for evasion of apoptosis. In conclusion, the current work provides a comprehensive overview of the altered transportome expression and its association with predicted PDAC malignancy with special focus on the epithelial compartment.
Collapse
Affiliation(s)
- A Zaccagnino
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| | - C Pilarsky
- Department of Surgery, University Clinic, Krankenhausstr. 12, 91054, Erlangen, Germany
| | - D Tawfik
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - S Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - A Trauzold
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - I Novak
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - A Schwab
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27 b, 48149, Muenster, Germany
| | - H Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| |
Collapse
|
88
|
Schnúr A, Hegyi P, Rousseau S, Lukacs GL, Veit G. Epithelial Anion Transport as Modulator of Chemokine Signaling. Mediators Inflamm 2016; 2016:7596531. [PMID: 27382190 PMCID: PMC4921137 DOI: 10.1155/2016/7596531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/03/2016] [Accepted: 05/12/2016] [Indexed: 12/16/2022] Open
Abstract
The pivotal role of epithelial cells is to secrete and absorb ions and water in order to allow the formation of a luminal fluid compartment that is fundamental for the epithelial function as a barrier against environmental factors. Importantly, epithelial cells also take part in the innate immune system. As a first line of defense they detect pathogens and react by secreting and responding to chemokines and cytokines, thus aggravating immune responses or resolving inflammatory states. Loss of epithelial anion transport is well documented in a variety of diseases including cystic fibrosis, chronic obstructive pulmonary disease, asthma, pancreatitis, and cholestatic liver disease. Here we review the effect of aberrant anion secretion with focus on the release of inflammatory mediators by epithelial cells and discuss putative mechanisms linking these transport defects to the augmented epithelial release of chemokines and cytokines. These mechanisms may contribute to the excessive and persistent inflammation in many respiratory and gastrointestinal diseases.
Collapse
Affiliation(s)
- Andrea Schnúr
- Department of Physiology, McGill University, Montréal, QC, Canada H3G 1Y6
| | - Péter Hegyi
- Institute for Translational Medicine and 1st Department of Medicine, University of Pécs, Pécs 7624, Hungary
- MTA-SZTE Translational Gastroenterology Research Group, Szeged 6720, Hungary
| | - Simon Rousseau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada H2X 2P2
| | - Gergely L. Lukacs
- Department of Physiology, McGill University, Montréal, QC, Canada H3G 1Y6
- Department of Biochemistry, McGill University, Montréal, QC, Canada H3G 1Y6
- Groupe de Recherche Axé sur la Structure des Protéines (GRASP), McGill University, Montréal, QC, Canada H3G 1Y6
| | - Guido Veit
- Department of Physiology, McGill University, Montréal, QC, Canada H3G 1Y6
| |
Collapse
|
89
|
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
|
90
|
Lahiri T, Hempstead SE, Brady C, Cannon CL, Clark K, Condren ME, Guill MF, Guillerman RP, Leone CG, Maguiness K, Monchil L, Powers SW, Rosenfeld M, Schwarzenberg SJ, Tompkins CL, Zemanick ET, Davis SD. Clinical Practice Guidelines From the Cystic Fibrosis Foundation for Preschoolers With Cystic Fibrosis. Pediatrics 2016; 137:peds.2015-1784. [PMID: 27009033 DOI: 10.1542/peds.2015-1784] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/29/2015] [Indexed: 11/24/2022] Open
Abstract
Cystic fibrosis (CF) clinical care guidelines exist for the care of infants up to age 2 years and for individuals ≥6 years of age. An important gap exists for preschool children between the ages of 2 and 5 years. This period marks a time of growth and development that is critical to achieve optimal nutritional status and maintain lung health. Given that disease often progresses in a clinically silent manner, objective and sensitive tools that detect and track early disease are important in this age group. Several challenges exist that may impede the delivery of care for these children, including adherence to therapies. A multidisciplinary committee was convened by the CF Foundation to develop comprehensive evidence-based and consensus recommendations for the care of preschool children, ages 2 to 5 years, with CF. This document includes recommendations in the following areas: routine surveillance for pulmonary disease, therapeutics, and nutritional and gastrointestinal care.
Collapse
Affiliation(s)
- Thomas Lahiri
- Pediatric Pulmonology, University of Vermont Children's Hospital and Department of Pediatrics, University of Vermont College of Medicine, Burlington, Vermont;
| | - Sarah E Hempstead
- The Dartmouth Institute for Health Policy and Clinical Practice, Lebanon, New Hampshire; Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Cynthia Brady
- Children's Respiratory and Critical Care Specialists and Children's Hospitals and Clinics of Minnesota, Minneapolis, Minnesota
| | | | - Kelli Clark
- Department of Pediatrics, University of North Carolina, Charlotte, North Carolina
| | - Michelle E Condren
- University of Oklahoma College of Pharmacy and School of Community Medicine, Tulsa, Oklahoma
| | - Margaret F Guill
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; Allergy and Pediatric Pulmonology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - R Paul Guillerman
- Department of Radiology, Baylor College of Medicine and Department of Pediatric Radiology, Texas Children's Hospital, Houston, Texas
| | - Christina G Leone
- Cystic Fibrosis Center, Children's Hospital Colorado, Aurora, Colorado
| | - Karen Maguiness
- Section of Pediatric Pulmonology, Allergy and Sleep Medicine, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lisa Monchil
- Armond V. Mascia, MD Cystic Fibrosis Center, Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, New York
| | - Scott W Powers
- Department of Pediatrics and Cincinnati Children's Research Foundation, University of Cincinnati College of Medicine and Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Margaret Rosenfeld
- Division of Pulmonary Medicine, Seattle Children's Hospital and Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Sarah Jane Schwarzenberg
- Pediatric Gastroenterology, Hepatology and Nutrition, University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - Connie L Tompkins
- Department of Rehabilitation and Movement Sciences, University of Vermont College of Nursing and Health Sciences, Burlington, Vermont; and
| | - Edith T Zemanick
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Stephanie D Davis
- Section of Pediatric Pulmonology, Allergy and Sleep Medicine, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
91
|
The adenosine A2B receptor is involved in anion secretion in human pancreatic duct Capan-1 epithelial cells. Pflugers Arch 2016; 468:1171-1181. [PMID: 26965147 PMCID: PMC4943985 DOI: 10.1007/s00424-016-1806-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 12/13/2022]
Abstract
Adenosine modulates a wide variety of biological processes via adenosine receptors. In the exocrine pancreas, adenosine regulates transepithelial anion secretion in duct cells and is considered to play a role in acini-to-duct signaling. To identify the functional adenosine receptors and Cl− channels important for anion secretion, we herein performed experiments on Capan-1, a human pancreatic duct cell line, using open-circuit Ussing chamber and gramicidin-perforated patch-clamp techniques. The luminal addition of adenosine increased the negative transepithelial potential difference (Vte) in Capan-1 monolayers with a half-maximal effective concentration value of approximately 10 μM, which corresponded to the value obtained on whole-cell Cl− currents in Capan-1 single cells. The effects of adenosine on Vte, an equivalent short-circuit current (Isc), and whole-cell Cl− currents were inhibited by CFTRinh-172, a cystic fibrosis transmembrane conductance regulator (CFTR) Cl− channel inhibitor. The adenosine A2B receptor agonist, BAY 60-6583, increased Isc and whole-cell Cl− currents through CFTR Cl− channels, whereas the A2A receptor agonist, CGS 21680, had negligible effects. The A2B receptor antagonist, PSB 603, inhibited the response of Isc to adenosine. Immunohistochemical analysis showed that the A2A and A2B receptors colocalized with Ezrin in the luminal membranes of Capan-1 monolayers and in rat pancreatic ducts. Adenosine elicited the whole-cell Cl− currents in guinea pig duct cells. These results demonstrate that luminal adenosine regulates anion secretion by activating CFTR Cl− channels via adenosine A2B receptors on the luminal membranes of Capan-1 cells. The present study endorses that purinergic signaling is important in the regulation of pancreatic secretion.
Collapse
|
92
|
Pallagi P, Hegyi P, Rakonczay Z. The Physiology and Pathophysiology of Pancreatic Ductal Secretion: The Background for Clinicians. Pancreas 2015; 44:1211-1233. [PMID: 26465950 DOI: 10.1097/mpa.0000000000000421] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human exocrine pancreas consists of 2 main cell types: acinar and ductal cells. These exocrine cells interact closely to contribute to the secretion of pancreatic juice. The most important ion in terms of the pancreatic ductal secretion is HCO3. In fact, duct cells produce an alkaline fluid that may contain up to 140 mM NaHCO3, which is essential for normal digestion. This article provides an overview of the basics of pancreatic ductal physiology and pathophysiology. In the first part of the article, we discuss the ductal electrolyte and fluid transporters and their regulation. The central role of cystic fibrosis transmembrane conductance regulator (CFTR) is highlighted, which is much more than just a Cl channel. We also review the role of pancreatic ducts in severe debilitating diseases such as cystic fibrosis (caused by various genetic defects of cftr), pancreatitis, and diabetes mellitus. Stimulation of ductal secretion in cystic fibrosis and pancreatitis may have beneficial effects in their treatment.
Collapse
Affiliation(s)
- Petra Pallagi
- From the *First Department of Medicine, University of Szeged; and †Hungarian Academy of Sciences-University of Szeged Translational Gastroenterology Research Group, Szeged, Hungary
| | | | | |
Collapse
|
93
|
Gibson-Corley KN, Meyerholz DK, Engelhardt JF. Pancreatic pathophysiology in cystic fibrosis. J Pathol 2015; 238:311-20. [PMID: 26365583 DOI: 10.1002/path.4634] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/04/2015] [Accepted: 09/08/2015] [Indexed: 12/28/2022]
Abstract
The pancreas is one of the earliest, and most commonly affected, organs in patients with cystic fibrosis (CF). Studying the pathogenesis of pancreatic disease is limited in CF patients, due to its early clinical onset, co-morbidities and lack of tissue samples from the early phases of disease. In recent years, several new CF animal models have been developed that have advanced our understanding of both CF exocrine and endocrine pancreatic disease. Additionally, these models have helped us to better define the influence of pancreatic lesions on CF disease progression in other organs, such as the gastrointestinal tract and lung.
Collapse
Affiliation(s)
| | - David K Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
94
|
Watson P. Pancreatitis in dogs and cats: definitions and pathophysiology. J Small Anim Pract 2015; 56:3-12. [PMID: 25586802 DOI: 10.1111/jsap.12293] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 12/10/2013] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
Pancreatitis, or inflammation of the pancreas, is commonly seen in dogs and cats and presents a spectrum of disease severities from acute to chronic and mild to severe. It is usually sterile, but the causes and pathophysiology remain poorly understood. The acute end of the disease spectrum is associated with a high mortality but the potential for complete recovery of organ structure and function if the animal survives. At the other end of the spectrum, chronic pancreatitis in either species can cause refractory pain and reduce quality of life. It may also result in progressive exocrine and endocrine functional impairment. There is confusion in the veterinary literature about definitions of acute and chronic pancreatitis and there are very few studies on the pathophysiology of naturally occurring pancreatitis in dogs and cats. This article reviews histological and clinical definitions and current understanding of the pathophysiology and causes in small animals by comparison with the much more extensive literature in humans, and suggests many areas that need further study in dogs and cats.
Collapse
Affiliation(s)
- P Watson
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES
| |
Collapse
|
95
|
Genetic susceptibility factors for alcohol-induced chronic pancreatitis. Pancreatology 2015; 15:S23-31. [PMID: 26149858 DOI: 10.1016/j.pan.2015.05.476] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/10/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022]
Abstract
Chronic pancreatitis is a progressive inflammatory disease of the pancreas and frequently associated with immoderate alcohol consumption. Since only a small proportion of alcoholics eventually develop chronic pancreatitis genetic susceptibility factors have long been suspected to contribute to the pathogenesis of the disease. Smaller studies in ethnically defined populations have found that not only polymorphism in proteins involved in the metabolism of ethanol, such as Alcohol Dehydrogenase and Aldehyde Dehydrogenase, can confer a risk for developing chronic pancreatitis but also mutations that had previously been reported in association with idiopathic pancreatitis, such as SPINK1 mutations. In a much broader approach employing genome wide search strategies the NAPS study found that polymorphisms in the Trypsin locus (PRSS1 rs10273639), and the Claudin 2 locus (CLDN2-RIPPLY1-MORC4 locus rs7057398 and rs12688220) confer an increased risk of developing alcohol-induced pancreatitis. These results from North America have now been confirmed by a European consortium. In another genome wide approach polymorphisms in the genes encoding Fucosyltransferase 2 (FUT2) non-secretor status and blood group B were not only found in association with higher serum lipase levels in healthy volunteers but also to more than double the risk for developing alcohol-associated chronic pancreatitis. These novel genetic associations will allow to investigate the pathophysiological and biochemical basis of alcohol-induced chronic pancreatitis on a cellular level and in much more detail than previously possible.
Collapse
|
96
|
Wang J, Barbuskaite D, Tozzi M, Giannuzzo A, Sørensen CE, Novak I. Proton Pump Inhibitors Inhibit Pancreatic Secretion: Role of Gastric and Non-Gastric H+/K+-ATPases. PLoS One 2015; 10:e0126432. [PMID: 25993003 PMCID: PMC4436373 DOI: 10.1371/journal.pone.0126432] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/02/2015] [Indexed: 02/07/2023] Open
Abstract
The mechanism by which pancreas secretes high HCO3- has not been fully resolved. This alkaline secretion, formed in pancreatic ducts, can be achieved by transporting HCO3- from serosa to mucosa or by moving H+ in the opposite direction. The aim of the present study was to determine whether H+/K+-ATPases are expressed and functional in human pancreatic ducts and whether proton pump inhibitors (PPIs) have effect on those. Here we show that the gastric HKα1 and HKβ subunits (ATP4A; ATP4B) and non-gastric HKα2 subunits (ATP12A) of H+/K+-ATPases are expressed in human pancreatic cells. Pumps have similar localizations in duct cell monolayers (Capan-1) and human pancreas, and notably the gastric pumps are localized on the luminal membranes. In Capan-1 cells, PPIs inhibited recovery of intracellular pH from acidosis. Furthermore, in rats treated with PPIs, pancreatic secretion was inhibited but concentrations of major ions in secretion follow similar excretory curves in control and PPI treated animals. In addition to HCO3-, pancreas also secretes K+. In conclusion, this study calls for a revision of the basic model for HCO3- secretion. We propose that proton transport is driving secretion, and that in addition it may provide a protective pH buffer zone and K+ recirculation. Furthermore, it seems relevant to re-evaluate whether PPIs should be used in treatment therapies where pancreatic functions are already compromised.
Collapse
Affiliation(s)
- Jing Wang
- Department of Biology, Section for Molecular Integrative Physiology, August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Dagne Barbuskaite
- Department of Biology, Section for Molecular Integrative Physiology, August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Marco Tozzi
- Department of Biology, Section for Molecular Integrative Physiology, August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Giannuzzo
- Department of Biology, Section for Molecular Integrative Physiology, August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Christiane E. Sørensen
- Department of Biology, Section for Molecular Integrative Physiology, August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Ivana Novak
- Department of Biology, Section for Molecular Integrative Physiology, August Krogh Building, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
97
|
Navis A, Bagnat M. Loss of cftr function leads to pancreatic destruction in larval zebrafish. Dev Biol 2015; 399:237-48. [PMID: 25592226 DOI: 10.1016/j.ydbio.2014.12.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 12/14/2022]
Abstract
The development and function of many internal organs requires precisely regulated fluid secretion. A key regulator of vertebrate fluid secretion is an anion channel, the cystic fibrosis transmembrane conductance regulator (CFTR). Loss of CFTR function leads to defects in fluid transport and cystic fibrosis (CF), a complex disease characterized by a loss of fluid secretion and mucus buildup in many organs including the lungs, liver, and pancreas. Several animal models including mouse, ferret and pig have been generated to investigate the pathophysiology of CF. However, these models have limited accessibility to early processes in the development of CF and are not amenable for forward genetic or chemical screens. Here, we show that Cftr is expressed and localized to the apical membrane of the zebrafish pancreatic duct and that loss of cftr function leads to destruction of the exocrine pancreas and a cystic fibrosis phenotype that mirrors human disease. Our analyses reveal that the cftr mutant pancreas initially develops normally, then rapidly loses pancreatic tissue during larval life, reflecting pancreatic disease in CF. Altogether, we demonstrate that the cftr mutant zebrafish is a powerful new model for pancreatitis and pancreatic destruction in CF. This accessible model will allow more detailed investigation into the mechanisms that drive CF of the pancreas and facilitate development of new therapies to treat the disease.
Collapse
Affiliation(s)
- Adam Navis
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - Michel Bagnat
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States.
| |
Collapse
|
98
|
Mélo SKM, Santiago TA, Duarte TDL, Abreu JMG, Manso Filho HC. A Proton-Pump Inhibitor Modifies the Concentration of Digestion Biomarkers in Healthy Horses. J Equine Vet Sci 2014. [DOI: 10.1016/j.jevs.2014.09.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
99
|
Molloy K, McElvaney NG. Ivacaftor: from bench to bedside... and back again. Am J Respir Crit Care Med 2014; 190:128-9. [PMID: 25025350 DOI: 10.1164/rccm.201406-1122ed] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kevin Molloy
- 1 Respiratory Research Division Royal College of Surgeons in Ireland Dublin, Ireland and
| | | |
Collapse
|
100
|
Kong SC, Giannuzzo A, Gianuzzo A, Novak I, Pedersen SF. Acid-base transport in pancreatic cancer: molecular mechanisms and clinical potential. Biochem Cell Biol 2014; 92:449-59. [PMID: 25372771 DOI: 10.1139/bcb-2014-0078] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Solid tumors are characterized by a microenvironment that is highly acidic, while intracellular pH (pHi) is normal or even elevated. This is the result of elevated metabolic rates in the highly proliferative cancer cells, in conjunction with often greatly increased rates of net cellular acid extrusion. Studies in various cancers have suggested that while the acid extrusion mechanisms employed are generally the same as those in healthy cells, the specific transporters upregulated vary with the cancer type. The main such transporters include Na(+)/H(+) exchangers, various HCO3(-) transporters, H(+) pumps, and lactate-H(+) cotransporters. The mechanisms leading to their dysregulation in cancer are incompletely understood but include changes in transporter expression levels, trafficking and membrane localization, and posttranslational modifications. In turn, accumulating evidence has revealed that in addition to supporting their elevated metabolic rate, their increased acid efflux capacity endows the cancer cells with increased capacity for invasiveness, proliferation, and chemotherapy resistance. The pancreatic duct exhibits an enormous capacity for acid-base transport, rendering pHi dysregulation a potentially very important topic in pancreatic ductal adenocarcinoma (PDAC). PDAC - accounting for about 90% of all pancreatic cancers - has one of the highest cancer mortality rates known, and new diagnostic and treatment options are highly needed. However, very little is known about whether pH regulation is altered in PDAC and, if so, the possible role of this in cancer development. Here, we review current models for pancreatic acid-base transport and pH homeostasis and summarize current views on acid-base dysregulation in cancer, focusing where possible on the few studies to date in PDAC. Finally, we present new data-mining analyses of acid-base transporter expression changes in PDAC and discuss essential directions for future work.
Collapse
Affiliation(s)
- Su Chii Kong
- a Section for Cell and Developmental Biology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|