1
|
Michicich M, Traylor Z, McCoy C, Valerio DM, Wilson A, Schneider M, Davis S, Barabas A, Mann RJ, LePage DF, Jiang W, Drumm ML, Kelley TJ, Conlon RA, Hodges CA. A W1282X cystic fibrosis mouse allows the study of pharmacological and gene-editing therapeutics to restore CFTR function. J Cyst Fibros 2025; 24:164-174. [PMID: 39532588 PMCID: PMC11788034 DOI: 10.1016/j.jcf.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/28/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND People with cystic fibrosis carrying two nonsense alleles lack CFTR-specific treatment. Growing evidence supports the hypothesis that nonsense mutation identity affects therapeutic response, calling for mutation-specific CF models. We describe a novel W1282X mouse model and compare it to an existing G542X mouse. METHODS The W1282X mouse was created using CRISPR/Cas9 to edit mouse Cftr. In this model, Cftr transcription was assessed using qRT-PCR and CFTR function was measured in the airway by nasal potential difference and in the intestine by short circuit current. Growth, survival, and intestinal motility were examined as well. Correction of W1282X CFTR was assessed pharmacologically and by gene-editing using a forskolin-induced swelling (FIS) assay in small intestine-derived organoids. RESULTS Homozygous W1282X mice demonstrate decreased Cftr mRNA, little to no CFTR function, and reduced survival, growth, and intestinal motility. W1282X organoids treated with various combinations of pharmacologic correctors display a significantly different amount of CFTR function than that of organoids from G542X mice. Successful gene editing of W1282X to wildtype sequence in intestinal organoids was achieved leading to restoration of CFTR function. CONCLUSIONS The W1282X mouse model recapitulates common human manifestations of CF similar to other CFTR null mice. Despite the similarities between the congenic W1282X and G542X models, they differ meaningfully in their response to identical pharmacological treatments. This heterogeneity highlights the importance of studying therapeutics across genotypes.
Collapse
Affiliation(s)
- Margaret Michicich
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Zachary Traylor
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Caitlan McCoy
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Dana M Valerio
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Alma Wilson
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Molly Schneider
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Sakeena Davis
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Amanda Barabas
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Rachel J Mann
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - David F LePage
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Weihong Jiang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Thomas J Kelley
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Ronald A Conlon
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Craig A Hodges
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States.
| |
Collapse
|
2
|
Anton-Păduraru DT, Murgu AM, Bozomitu LI, Mîndru DE, Iliescu Halițchi CO, Trofin F, Ciongradi CI, Sârbu I, Eṣanu IM, Azoicăi AN. Diagnosis and Management of Gastrointestinal Manifestations in Children with Cystic Fibrosis. Diagnostics (Basel) 2024; 14:228. [PMID: 38275475 PMCID: PMC10814426 DOI: 10.3390/diagnostics14020228] [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: 12/15/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
Cystic fibrosis (CF) is primarily known for its pulmonary consequences, which are extensively explored in the existing literature. However, it is noteworthy that individuals with CF commonly display gastrointestinal (G-I) manifestations due to the substantial presence of the cystic fibrosis transmembrane conductance regulator (CFTR) protein in the intestinal tract. Recognized as pivotal nonpulmonary aspects of CF, G-I manifestations exhibit a diverse spectrum. Identifying and effectively managing these manifestations are crucial for sustaining health and influencing the overall quality of life for CF patients. This review aims to synthesize existing knowledge, providing a comprehensive overview of the G-I manifestations associated with CF. Each specific G-I manifestation, along with the diagnostic methodologies and therapeutic approaches, is delineated, encompassing the impact of innovative treatments targeting the fundamental effects of CF on the G-I tract. The findings underscore the imperative for prompt diagnosis and meticulous management of G-I manifestations, necessitating a multidisciplinary team approach for optimal care and enhancement of the quality of life for affected individuals. In conclusion, the authors emphasize the urgency for further clinical studies to establish a more robust evidence base for managing G-I symptoms within the context of this chronic disease. Such endeavors are deemed essential for advancing understanding and refining the clinical care of CF patients with G-I manifestations.
Collapse
Affiliation(s)
- Dana-Teodora Anton-Păduraru
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Alina Mariela Murgu
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Laura Iulia Bozomitu
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Dana Elena Mîndru
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| | - Codruța Olimpiada Iliescu Halițchi
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
| | - Felicia Trofin
- Department of Preventive Medicine and Interdisciplinarity–Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania;
| | - Carmen Iulia Ciongradi
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
- 2nd Department of Surgery, Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania
| | - Ioan Sârbu
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
- 2nd Department of Surgery, Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania
| | - Irina Mihaela Eṣanu
- Medical Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania;
| | - Alice Nicoleta Azoicăi
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaṣi, Romania; (D.-T.A.-P.); (L.I.B.); (D.E.M.); (C.O.I.H.); (A.N.A.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania; (C.I.C.); (I.S.)
| |
Collapse
|
3
|
Ferreira LMB, Cardoso VMO, Dos Santos Pedriz I, Souza MPC, Ferreira NN, Chorilli M, Gremião MPD, Zucolotto V. Understanding mucus modulation behavior of chitosan oligomers and dextran sulfate combining light scattering and calorimetric observations. Carbohydr Polym 2023; 306:120613. [PMID: 36746564 DOI: 10.1016/j.carbpol.2023.120613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
This study reports the fundamental understanding of mucus-modulatory strategies combining charged biopolymers with distinct molecular weights and surface charges. Here, key biophysical evidence supports that low-molecular-weight (Mw) polycation chitosan oligosaccharides (COSs) and high-Mw polyanion dextran sulfate (DS) exhibit distinct thermodynamic signatures upon interaction with mucin (MUC), the main protein of mucus. While the COS → MUC microcalorimetric titrations released ~14 kcal/mol and ~60 kcal/mol, the DS → MUC titrations released ~1200 and ~1450 kcal/mol at pH of 4.5 and 6.8, respectively. The MPT-2 titrations of COS → MUC and DS → MUC indicated a greater zeta potential variation at pH = 4.5 (relative variation = 815 % and 351 %, respectively) than at pH = 6.8 (relative variation = 282 % and 136 %, respectively). Further, the resultant binary (COS-MUC) and ternary (COS-DS-MUC) complexes showed opposite behavior (aggregation and charge inversion events) according to the pH environment. Most importantly, the results indicate that electrostatics could not be the driving force that governs COS-MUC interactions. To account for this finding, we proposed a two-level abstraction model. Macro features emerge collectively from individual interactions occurring at the molecular level. Therefore, to understand the outcomes of mucus modulatory strategy based on charged biopolymers it is necessary to integrate both visions into the same picture.
Collapse
Affiliation(s)
- Leonardo M B Ferreira
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil.
| | - Valéria M O Cardoso
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| | - Igor Dos Santos Pedriz
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| | - Maurício P C Souza
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Natália N Ferreira
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Maria P D Gremião
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo (USP), 13566-590 São Carlos, Brazil
| |
Collapse
|
4
|
Powell LC, Adams JYM, Quoraishi S, Py C, Oger A, Gazze SA, Francis LW, von Ruhland C, Owens D, Rye PD, Hill KE, Pritchard MF, Thomas DW. Alginate oligosaccharides enhance the antifungal activity of nystatin against candidal biofilms. Front Cell Infect Microbiol 2023; 13:1122340. [PMID: 36798083 PMCID: PMC9927220 DOI: 10.3389/fcimb.2023.1122340] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Background The increasing prevalence of invasive fungal infections in immuno-compromised patients is a considerable cause of morbidity and mortality. With the rapid emergence of antifungal resistance and an inadequate pipeline of new therapies, novel treatment strategies are now urgently required. Methods The antifungal activity of the alginate oligosaccharide OligoG in conjunction with nystatin was tested against a range of Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis and C. dubliniensis), in both planktonic and biofilm assays, to determine its potential clinical utility to enhance the treatment of candidal infections. The effect of OligoG (0-6%) ± nystatin on Candida spp. was examined in minimum inhibitory concentration (MIC) and growth curve assays. Antifungal effects of OligoG and nystatin treatment on biofilm formation and disruption were characterized using confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM) and ATP cellular viability assays. Effects on the cell membrane were determined using permeability assays and transmission electron microscopy (TEM). Results MIC and growth curve assays demonstrated the synergistic effects of OligoG (0-6%) with nystatin, resulting in an up to 32-fold reduction in MIC, and a significant reduction in the growth of C. parapsilosis and C. auris (minimum significant difference = 0.2 and 0.12 respectively). CLSM and SEM imaging demonstrated that the combination treatment of OligoG (4%) with nystatin (1 µg/ml) resulted in significant inhibition of candidal biofilm formation on glass and clinical grade silicone surfaces (p < 0.001), with increased cell death (p < 0.0001). The ATP biofilm disruption assay demonstrated a significant reduction in cell viability with OligoG (4%) alone and the combined OligoG/nystatin (MIC value) treatment (p < 0.04) for all Candida strains tested. TEM studies revealed the combined OligoG/nystatin treatment induced structural reorganization of the Candida cell membrane, with increased permeability when compared to the untreated control (p < 0.001). Conclusions Antimicrobial synergy between OligoG and nystatin against Candida spp. highlights the potential utility of this combination therapy in the prevention and topical treatment of candidal biofilm infections, to overcome the inherent tolerance of biofilm structures to antifungal agents.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- Microbiology and Infectious Disease group, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Lydia C. Powell,
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Sadik Quoraishi
- Otolaryngology Department, New Cross Hospital, Wolverhampton, United Kingdom
| | - Charlène Py
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Anaϊs Oger
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Salvatore A. Gazze
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Christopher von Ruhland
- Central Biotechnology Services, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David Owens
- Head and Neck Directorate, University Hospital of Wales, Cardiff, United Kingdom
| | | | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| |
Collapse
|
5
|
Cao S, Li L, Zhu B, Yao Z. Alginate modifying enzymes: An updated comprehensive review of the mannuronan C5-epimerases. ALGAL RES 2022. [DOI: 10.1016/j.algal.2022.102952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
6
|
Tan X, Kini A, Römermann D, Seidler U. The NHE3 Inhibitor Tenapanor Prevents Intestinal Obstructions in CFTR-Deleted Mice. Int J Mol Sci 2022; 23:ijms23179993. [PMID: 36077390 PMCID: PMC9456459 DOI: 10.3390/ijms23179993] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022] Open
Abstract
Mutations in the CFTR chloride channel result in intestinal obstructive episodes in cystic fibrosis (CF) patients and in CF animal models. In this study, we explored the possibility of reducing the frequency of obstructive episodes in cftr−/− mice through the oral application of a gut-selective NHE3 inhibitor tenapanor and searched for the underlying mechanisms involved. Sex- and age-matched cftr+/+ and cftr−/− mice were orally gavaged twice daily with 30 mg kg−1 tenapanor or vehicle for a period of 21 days. Body weight and stool water content was assessed daily and gastrointestinal transit time (GTT) once weekly. The mice were sacrificed when an intestinal obstruction was suspected or after 21 days, and stool and tissues were collected for further analysis. Twenty-one day tenapanor application resulted in a significant increase in stool water content and stool alkalinity and a significant decrease in GTT in cftr+/+ and cftr−/− mice. Tenapanor significantly reduced obstructive episodes to 8% compared to 46% in vehicle-treated cftr−/− mice and prevented mucosal inflammation. A decrease in cryptal hyperproliferation, mucus accumulation, and mucosal mast cell number was also observed in tenapanor- compared to vehicle-treated, unobstructed cftr−/− mice. Overall, oral tenapanor application prevented obstructive episodes in CFTR-deficient mice and was safe in cftr+/+ and cftr−/− mice. These results suggest that tenapanor may be a safe and affordable adjunctive therapy in cystic fibrosis patients to alleviate constipation and prevent recurrent DIOS.
Collapse
Affiliation(s)
| | | | | | - Ursula Seidler
- Correspondence: ; Tel.: +49-5115-329-427; Fax: +49-5115-328-428
| |
Collapse
|
7
|
Grubb BR, Livraghi-Butrico A. Animal models of cystic fibrosis in the era of highly effective modulator therapies. Curr Opin Pharmacol 2022; 64:102235. [DOI: 10.1016/j.coph.2022.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022]
|
8
|
Evaluating the alginate oligosaccharide (OligoG) as a therapy for Burkholderia cepacia complex cystic fibrosis lung infection. J Cyst Fibros 2022; 21:821-829. [DOI: 10.1016/j.jcf.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/15/2021] [Accepted: 01/09/2022] [Indexed: 11/15/2022]
|
9
|
Henen S, Denton C, Teckman J, Borowitz D, Patel D. Review of Gastrointestinal Motility in Cystic Fibrosis. J Cyst Fibros 2021; 20:578-585. [PMID: 34147362 DOI: 10.1016/j.jcf.2021.05.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Gastrointestinal manifestations in patients with cystic fibrosis (CF) are extremely common and have recently become a research focus. Gastrointestinal (GI) dysfunction is poorly understood in the CF population, despite many speculations including the role of luminal pH, bacterial overgrowth, and abnormal microbiome. Nevertheless, dysmotility is emerging as a possible key player in CF intestinal symptoms. Our review article aims to explore the sequelae of defective cystic fibrosis transmembrane conductance regulator (CFTR) genes on the GI tract as studied in both animals and humans, describe various presentations of intestinal dysmotility in CF, review newer diagnostic motility techniques including intraluminal manometry, and review the current literature regarding the potential role of dysmotility in CF-related intestinal pathologies.
Collapse
Affiliation(s)
- Sara Henen
- Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104.
| | - Christine Denton
- Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104
| | - Jeff Teckman
- Interim Chair, Department of Pediatrics, Professor of Pediatrics and Biochemistry, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand BLVD, St. Louis, MO 63104.
| | - Drucy Borowitz
- Emeritus Professor of Clinical Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, 1001 Main Street, Buffalo, NY, 14203.
| | - Dhiren Patel
- Associate Professor and Medical Director, Neurogastroenterology and Motility Program, Department of Pediatrics, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Blvd, St. Louis, MO 63104.
| |
Collapse
|
10
|
Tøndervik A, Aarstad OA, Aune R, Maleki S, Rye PD, Dessen A, Skjåk-Bræk G, Sletta H. Exploiting Mannuronan C-5 Epimerases in Commercial Alginate Production. Mar Drugs 2020; 18:E565. [PMID: 33218095 PMCID: PMC7698916 DOI: 10.3390/md18110565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Alginates are one of the major polysaccharide constituents of marine brown algae in commercial manufacturing. However, the content and composition of alginates differ according to the distinct parts of these macroalgae and have a direct impact on the concentration of guluronate and subsequent commercial value of the final product. The Azotobacter vinelandii mannuronan C-5 epimerases AlgE1 and AlgE4 were used to determine their potential value in tailoring the production of high guluronate low-molecular-weight alginates from two sources of high mannuronic acid alginates, the naturally occurring harvested brown algae (Ascophyllum nodosum, Durvillea potatorum, Laminaria hyperborea and Lessonia nigrescens) and a pure mannuronic acid alginate derived from fermented production of the mutant strain of Pseudomonas fluorescens NCIMB 10,525. The mannuronan C-5 epimerases used in this study increased the content of guluronate from 32% up to 81% in both the harvested seaweed and bacterial fermented alginate sources. The guluronate-rich alginate oligomers subsequently derived from these two different sources showed structural identity as determined by proton nuclear magnetic resonance (1H NMR), high-performance anion-exchange chromatography with pulsed amperometric detection (HPAEC-PAD) and size-exclusion chromatography with online multi-angle static laser light scattering (SEC-MALS). Functional identity was determined by minimum inhibitory concentration (MIC) assays with selected bacteria and antibiotics using the previously documented low-molecular-weight guluronate enriched alginate OligoG CF-5/20 as a comparator. The alginates produced using either source showed similar antibiotic potentiation effects to the drug candidate OligoG CF-5/20 currently in development as a mucolytic and anti-biofilm agent. These findings clearly illustrate the value of using epimerases to provide an alternative production route for novel low-molecular-weight alginates.
Collapse
Affiliation(s)
- Anne Tøndervik
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Richard Birkelands vei 3B, N-7034 Trondheim, Norway; (R.A.); (S.M.); (H.S.)
| | - Olav A. Aarstad
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology, NTNU, Sem Sælands vei 6-8, N-7491 Trondheim, Norway; (O.A.A.); (G.S.-B.)
| | - Randi Aune
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Richard Birkelands vei 3B, N-7034 Trondheim, Norway; (R.A.); (S.M.); (H.S.)
| | - Susan Maleki
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Richard Birkelands vei 3B, N-7034 Trondheim, Norway; (R.A.); (S.M.); (H.S.)
| | - Philip D. Rye
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway; (P.D.R.); (A.D.)
| | - Arne Dessen
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway; (P.D.R.); (A.D.)
| | - Gudmund Skjåk-Bræk
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology, NTNU, Sem Sælands vei 6-8, N-7491 Trondheim, Norway; (O.A.A.); (G.S.-B.)
| | - Håvard Sletta
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Richard Birkelands vei 3B, N-7034 Trondheim, Norway; (R.A.); (S.M.); (H.S.)
| |
Collapse
|
11
|
van Koningsbruggen-Rietschel S, Davies JC, Pressler T, Fischer R, MacGregor G, Donaldson SH, Smerud K, Meland N, Mortensen J, Fosbøl MØ, Downey DG, Myrset AH, Flaten H, Rye PD. Inhaled dry powder alginate oligosaccharide in cystic fibrosis: a randomised, double-blind, placebo-controlled, crossover phase 2b study. ERJ Open Res 2020; 6:00132-2020. [PMID: 33123558 PMCID: PMC7569163 DOI: 10.1183/23120541.00132-2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/25/2020] [Indexed: 11/21/2022] Open
Abstract
Background OligoG is a low molecular-weight alginate oligosaccharide that improves the viscoelastic properties of cystic fibrosis (CF) mucus and disrupts biofilms, thereby potentiating the activity of antimicrobial agents. The efficacy of inhaled OligoG was evaluated in adult patients with CF. Methods A randomised, double-blind, placebo-controlled multicentre crossover study was used to demonstrate safety and efficacy of inhaled dry powder OligoG. Subjects were randomly allocated to receive OligoG 1050 mg per day (10 capsules three times daily) or matching placebo for 28 days, with 28-day washout periods following each treatment period. The primary end-point was absolute change in percentage predicted forced expiratory volume in 1 s (FEV1) at the end of 28-day treatment. The intention-to-treat (ITT) population (n=65) was defined as randomised to treatment with at least one administration of study medication and post-dosing evaluation. Results In this study, 90 adult subjects were screened and 65 were randomised. Statistically significant improvement in FEV1 was not observed in the ITT population. Adverse events included nasopharyngitis, cough and pulmonary exacerbation. The number and proportions of patients with adverse events and serious adverse events were similar between OligoG and placebo group. Conclusions Inhalation of OligoG-dry powder over 28 days was safe in adult CF subjects. Statistically significant improvement of FEV1 was not reached. The planned analyses did not indicate a significant treatment benefit with OligoG compared to placebo. Post hoc exploratory analyses showed subgroup results that indicate that further studies of OligoG in this patient population are justified. Inhalation of OligoG-DPI over 28 days was shown to be safe in adult CF subjects. Statistically significant improvement of FEV1 was not reached. Post hoc subgroup analyses support mechanism of action for OligoG and warrant further prospective studies.https://bit.ly/2PHq6Z0
Collapse
Affiliation(s)
| | - Jane C Davies
- Dept of Paediatric Respiratory Medicine, National Heart and Lung Institute, Imperial College London, and Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | | | | | - Gordon MacGregor
- Dept of Respiratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | | | - Knut Smerud
- SMERUD Medical Research International AS, Oslo, Norway
| | - Nils Meland
- SMERUD Medical Research International AS, Oslo, Norway
| | - Jann Mortensen
- Copenhagen CF Centre, Rigshospitalet, Copenhagen, Denmark
| | - Marie Ø Fosbøl
- Copenhagen CF Centre, Rigshospitalet, Copenhagen, Denmark
| | - Damian G Downey
- Centre for Experimental Medicine, Queen's University, Belfast, UK
| | | | | | | |
Collapse
|
12
|
Liu J, Yang S, Li X, Yan Q, Reaney MJT, Jiang Z. Alginate Oligosaccharides: Production, Biological Activities, and Potential Applications. Compr Rev Food Sci Food Saf 2019; 18:1859-1881. [DOI: 10.1111/1541-4337.12494] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/09/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Jun Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthCollege of Food Science and Nutritional EngineeringChina Agricultural Univ. Beijing 100083 China
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthBeijing Technology and Business Univ. Beijing 100048 China
| | - Shaoqing Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthCollege of Food Science and Nutritional EngineeringChina Agricultural Univ. Beijing 100083 China
| | - Xiuting Li
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthBeijing Technology and Business Univ. Beijing 100048 China
| | - Qiaojuan Yan
- Bioresource Utilization LaboratoryCollege of EngineeringChina Agricultural Univ. Beijing 100083 China
| | - Martin J. T. Reaney
- Dept. of Plant SciencesUniv. of Saskatchewan Saskatoon SK S7N 5A8 Canada
- Guangdong Saskatchewan Oilseed Joint Laboratory (GUSTO)Dept. of Food Science and EngineeringJinan Univ. Guangzhou 510632 China
| | - Zhengqiang Jiang
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthCollege of Food Science and Nutritional EngineeringChina Agricultural Univ. Beijing 100083 China
| |
Collapse
|
13
|
Fernandez-Petty CM, Hughes GW, Bowers HL, Watson JD, Rosen BH, Townsend SM, Santos C, Ridley CE, Chu KK, Birket SE, Li Y, Leung HM, Mazur M, Garcia BA, Evans TIA, Libby EF, Hathorne H, Hanes J, Tearney GJ, Clancy JP, Engelhardt JF, Swords WE, Thornton DJ, Wiesmann WP, Baker SM, Rowe SM. A glycopolymer improves vascoelasticity and mucociliary transport of abnormal cystic fibrosis mucus. JCI Insight 2019; 4:125954. [PMID: 30996141 DOI: 10.1172/jci.insight.125954] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/28/2019] [Indexed: 01/05/2023] Open
Abstract
Cystic fibrosis (CF) is characterized by increased mucus viscosity and delayed mucociliary clearance that contributes to progressive decline of lung function. Mucus in the respiratory and GI tract is excessively adhesive in the presence of airway dehydration and excess extracellular Ca2+ upon mucin release, promoting hyperviscous, densely packed mucins characteristic of CF. Therapies that target mucins directly through ionic interactions remain unexploited. Here we show that poly (acetyl, arginyl) glucosamine (PAAG), a polycationic biopolymer suitable for human use, interacts directly with mucins in a Ca2+-sensitive manner to reduce CF mucus viscoelasticity and improve its transport. Notably, PAAG induced a linear structure of purified MUC5B and altered its sedimentation profile and viscosity, indicative of proper mucin expansion. In vivo, PAAG nebulization improved mucociliary transport in CF rats with delayed mucus clearance, and cleared mucus plugging in CF ferrets. This study demonstrates the potential use of a synthetic glycopolymer PAAG as a molecular agent that could benefit patients with a broad array of mucus diseases.
Collapse
Affiliation(s)
| | - Gareth W Hughes
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, United Kingdom
| | - Hannah L Bowers
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - John D Watson
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Bradley H Rosen
- Department of Anatomy & Cell Biology and.,Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | | | | | - Caroline E Ridley
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, United Kingdom
| | - Kengyeh K Chu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA,Harvard Medical School, Boston, Massachusetts, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Susan E. Birket
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center
| | - Yao Li
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center
| | - Hui Min Leung
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA,Harvard Medical School, Boston, Massachusetts, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marina Mazur
- Gregory Fleming James Cystic Fibrosis Research Center
| | - Bryan A Garcia
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center
| | | | | | - Heather Hathorne
- Gregory Fleming James Cystic Fibrosis Research Center,Department of Pediatrics, UAB, Birmingham, Alabama, USA
| | - Justin Hanes
- Center for Nanomedicine and Departments of Biomedical Engineering, Chemical & Biomolecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Guillermo J Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA,Harvard Medical School, Boston, Massachusetts, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John P Clancy
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - John F Engelhardt
- Department of Anatomy & Cell Biology and.,Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - William E Swords
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center
| | - David J Thornton
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, United Kingdom
| | | | | | - Steven M Rowe
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center,Department of Pediatrics, UAB, Birmingham, Alabama, USA.,Department of Cell Developmental & Integrative Biology, UAB, Birmingham, Alabama, USA
| |
Collapse
|
14
|
McHugh DR, Cotton CU, Moss FJ, Vitko M, Valerio DM, Kelley TJ, Hao S, Jafri A, Drumm ML, Boron WF, Stern RC, McBennett K, Hodges CA. Linaclotide improves gastrointestinal transit in cystic fibrosis mice by inhibiting sodium/hydrogen exchanger 3. Am J Physiol Gastrointest Liver Physiol 2018; 315:G868-G878. [PMID: 30118317 PMCID: PMC9925117 DOI: 10.1152/ajpgi.00261.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastrointestinal dysfunction in cystic fibrosis (CF) is a prominent source of pain among patients with CF. Linaclotide, a guanylate cyclase C (GCC) receptor agonist, is a US Food and Drug Administration-approved drug prescribed for chronic constipation but has not been widely used in CF, as the cystic fibrosis transmembrane conductance regulator (CFTR) is the main mechanism of action. However, anecdotal clinical evidence suggests that linaclotide may be effective for treating some gastrointestinal symptoms in CF. The goal of this study was to determine the effectiveness and mechanism of linaclotide in treating CF gastrointestinal disorders using CF mouse models. Intestinal transit, chloride secretion, and intestinal lumen fluidity were assessed in wild-type and CF mouse models in response to linaclotide. CFTR and sodium/hydrogen exchanger 3 (NHE3) response to linaclotide was also evaluated. Linaclotide treatment improved intestinal transit in mice carrying either F508del or null Cftr mutations but did not induce detectable Cl- secretion. Linaclotide increased fluid retention and fluidity of CF intestinal contents, suggesting inhibition of fluid absorption. Targeted inhibition of sodium absorption by the NHE3 inhibitor tenapanor produced improvements in gastrointestinal transit similar to those produced by linaclotide treatment, suggesting that inhibition of fluid absorption by linaclotide contributes to improved gastrointestinal transit in CF. Our results demonstrate that linaclotide improves gastrointestinal transit in CF mouse models by increasing luminal fluidity through inhibiting NHE3-mediated sodium absorption. Further studies are necessary to assess whether linaclotide could improve CF intestinal pathologies in patients. GCC signaling and NHE3 inhibition may be therapeutic targets for CF intestinal manifestations. NEW & NOTEWORTHY Linaclotide's primary mechanism of action in alleviating chronic constipation is through cystic fibrosis transmembrane conductance regulator (CFTR), negating its use in patients with cystic fibrosis (CF). For the first time, our findings suggest that in the absence of CFTR, linaclotide can improve fluidity of the intestinal lumen through the inhibition of sodium/hydrogen exchanger 3. These findings suggest that linaclotide could improve CF intestinal pathologies in patients.
Collapse
Affiliation(s)
- Daniel R. McHugh
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Calvin U. Cotton
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fraser J. Moss
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Megan Vitko
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Dana M. Valerio
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Thomas J. Kelley
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,4Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Shuyu Hao
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Anjum Jafri
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mitchell L. Drumm
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Walter F. Boron
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio,5Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio,6Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Robert C. Stern
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,7Rainbow Babies and Children’s Hospital, Cleveland, Ohio
| | - Kimberly McBennett
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,7Rainbow Babies and Children’s Hospital, Cleveland, Ohio
| | - Craig A. Hodges
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
15
|
Powell LC, Pritchard MF, Ferguson EL, Powell KA, Patel SU, Rye PD, Sakellakou SM, Buurma NJ, Brilliant CD, Copping JM, Menzies GE, Lewis PD, Hill KE, Thomas DW. Targeted disruption of the extracellular polymeric network of Pseudomonas aeruginosa biofilms by alginate oligosaccharides. NPJ Biofilms Microbiomes 2018; 4:13. [PMID: 29977590 PMCID: PMC6026129 DOI: 10.1038/s41522-018-0056-3] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/20/2018] [Accepted: 06/06/2018] [Indexed: 11/29/2022] Open
Abstract
Acquisition of a mucoid phenotype by Pseudomonas sp. in the lungs of cystic fibrosis (CF) patients, with subsequent over-production of extracellular polymeric substance (EPS), plays an important role in mediating the persistence of multi-drug resistant (MDR) infections. The ability of a low molecular weight (Mn = 3200 g mol−1) alginate oligomer (OligoG CF-5/20) to modify biofilm structure of mucoid Pseudomonas aeruginosa (NH57388A) was studied in vitro using scanning electron microscopy (SEM), confocal laser scanning microscopy (CLSM) with Texas Red (TxRd®)-labelled OligoG and EPS histochemical staining. Structural changes in treated biofilms were quantified using COMSTAT image-analysis software of CLSM z-stack images, and nanoparticle diffusion. Interactions between the oligomers, Ca2+ and DNA were studied using molecular dynamics (MD) simulations, Fourier transform infrared spectroscopy (FTIR) and isothermal titration calorimetry (ITC). Imaging demonstrated that OligoG treatment (≥0.5%) inhibited biofilm formation, revealing a significant reduction in both biomass and biofilm height (P < 0.05). TxRd®-labelled oligomers readily diffused into established (24 h) biofilms. OligoG treatment (≥2%) induced alterations in the EPS of established biofilms; significantly reducing the structural quantities of EPS polysaccharides, and extracellular (e)DNA (P < 0.05) with a corresponding increase in nanoparticle diffusion (P < 0.05) and antibiotic efficacy against established biofilms. ITC demonstrated an absence of rapid complex formation between DNA and OligoG and confirmed the interactions of OligoG with Ca2+ evident in FTIR and MD modelling. The ability of OligoG to diffuse into biofilms, potentiate antibiotic activity, disrupt DNA-Ca2+-DNA bridges and biofilm EPS matrix highlights its potential for the treatment of biofilm-related infections. Small carbohydrate molecules derived from marine algae show potential for inhibiting biofilm formation in multi-drug resistant infections. A research team led by Lydia Powell at Cardiff University, UK, investigated the action of carbohydrates called alginate oligosaccharides, composed of a small number of linked sugar molecules. The oligosaccharides modified and disrupted the structure of cultured biofilms of Pseudomonas aeruginosa, the cause of many serious drug resistant infections. This effect significantly inhibited the formation and maintenance of the biofilm state, which is known to be a crucial factor allowing the bacteria to resist drug treatment. Antibiotics proved more effective following the oligosaccharide intervention. The researchers uncovered key molecular details involved in the ability of the oligosaccharides to diffuse into and disrupt biofilms. The therapeutic potential of these small carbohydrates is currently being investigated in clinical trials.
Collapse
Affiliation(s)
- Lydia C Powell
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| | - Manon F Pritchard
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| | - Elaine L Ferguson
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| | - Kate A Powell
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| | - Shree U Patel
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| | | | | | - Niklaas J Buurma
- 3Physical Organic Chemistry Centre, School of Chemistry, Cardiff University, Cardiff, UK
| | | | - Jack M Copping
- 4Respiratory Diagnostics Group, Swansea University, Swansea, UK
| | | | - Paul D Lewis
- 4Respiratory Diagnostics Group, Swansea University, Swansea, UK
| | - Katja E Hill
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| | - David W Thomas
- 1Advanced Therapies Group, Cardiff University School of Dentistry, Heath Park, Cardiff, CF14 4XY UK
| |
Collapse
|
16
|
Nordgård CT, Draget KI. Co association of mucus modulating agents and nanoparticles for mucosal drug delivery. Adv Drug Deliv Rev 2018; 124:175-183. [PMID: 29307632 DOI: 10.1016/j.addr.2018.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/26/2017] [Accepted: 01/02/2018] [Indexed: 01/27/2023]
Abstract
Nanoparticulate drug delivery systems (nDDS) offer a variety of options when it comes to routes of administration. One possible path is crossing mucosal barriers, such as in the airways and in the GI tract, for systemic distribution or local treatment. The main challenge with this administration route is that the size and surface properties of the nanoparticles, as opposed to small molecular drugs, very often results in mucosal capture, immobilization and removal, which in turn results in a very low bioavailability. Strategies to overcome this challenge do exist, like surface 'stealth' modification with PEG. Here we review an alternative or supplemental strategy, co-association of mucus modulating agents with the nDDS to improve bioavailability, where the nDDS may be surface modified or unmodified. This contribution presents some examples on how possible co-association systems may be achieved, using currently marketed mucolytic drugs, alternative formulations or novel agents.
Collapse
Affiliation(s)
- Catherine Taylor Nordgård
- NOBIPOL, Department of Biotechnology and Food Science, Norwegian University of Science and Technology NTNU, 7491 Trondheim, Norway.
| | - Kurt I Draget
- NOBIPOL, Department of Biotechnology and Food Science, Norwegian University of Science and Technology NTNU, 7491 Trondheim, Norway.
| |
Collapse
|
17
|
Alginate Oligomers and Their Use as Active Pharmaceutical Drugs. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2018. [DOI: 10.1007/978-981-10-6910-9_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
18
|
Lee YE, Kim H, Seo C, Park T, Lee KB, Yoo SY, Hong SC, Kim JT, Lee J. Marine polysaccharides: therapeutic efficacy and biomedical applications. Arch Pharm Res 2017; 40:1006-1020. [PMID: 28918561 PMCID: PMC7090684 DOI: 10.1007/s12272-017-0958-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/10/2017] [Indexed: 12/22/2022]
Abstract
The ocean contains numerous marine organisms, including algae, animals, and plants, from which diverse marine polysaccharides with useful physicochemical and biological properties can be extracted. In particular, fucoidan, carrageenan, alginate, and chitosan have been extensively investigated in pharmaceutical and biomedical fields owing to their desirable characteristics, such as biocompatibility, biodegradability, and bioactivity. Various therapeutic efficacies of marine polysaccharides have been elucidated, including the inhibition of cancer, inflammation, and viral infection. The therapeutic activities of these polysaccharides have been demonstrated in various settings, from in vitro laboratory-scale experiments to clinical trials. In addition, marine polysaccharides have been exploited for tissue engineering, the immobilization of biomolecules, and stent coating. Their ability to detect and respond to external stimuli, such as pH, temperature, and electric fields, has enabled their use in the design of novel drug delivery systems. Thus, along with the promising characteristics of marine polysaccharides, this review will comprehensively detail their various therapeutic, biomedical, and miscellaneous applications.
Collapse
Affiliation(s)
- Young-Eun Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Hyeongmin Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Changwon Seo
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Taejun Park
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Kyung Bin Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Seung-Yup Yoo
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Seong-Chul Hong
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Jeong Tae Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea.
| |
Collapse
|
19
|
Darrah R, Bederman I, Vitko M, Valerio DM, Drumm ML, Hodges CA. Growth deficits in cystic fibrosis mice begin in utero prior to IGF-1 reduction. PLoS One 2017; 12:e0175467. [PMID: 28384265 PMCID: PMC5383306 DOI: 10.1371/journal.pone.0175467] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/27/2017] [Indexed: 12/16/2022] Open
Abstract
Growth deficits are common in cystic fibrosis (CF), but their cause is complex, with contributions from exocrine pancreatic insufficiency, pulmonary complications, gastrointestinal obstructions, and endocrine abnormalities. The CF mouse model displays similar growth impairment despite exocrine pancreatic function and in the absence of chronic pulmonary infection. The high incidence of intestinal obstruction in the CF mouse has been suggested to significantly contribute to the observed growth deficits. Previous studies by our group have shown that restoration of the cystic fibrosis transmembrane conductance regulator (CFTR) in the intestinal epithelium prevents intestinal obstruction but does not improve growth. In this study, we further investigate growth deficits in CF and gut-corrected CF mice by assessing insulin-like growth factor 1 (IGF-1). IGF-1 levels were significantly decreased in CF and gut-corrected CF adult mice compared to wildtype littermates and were highly correlated with weight. Interestingly, perinatal IGF-1 levels were not significantly different between CF and wildtype littermates, even though growth deficits in CF mice could be detected late in gestation. Since CFTR has been suggested to play a role in water and nutrient exchange in the placenta through its interaction with aquaporins, we analyzed placental aquaporin expression in late-gestation CF and control littermates. While significant differences were observed in Aquaporin 9 expression in CF placentas in late gestation, there was no evidence of placental fluid exchange differences between CF and control littermates. The results from this study indicate that decreased IGF-1 levels are highly correlated with growth in CF mice, independent of CF intestinal obstruction. However, the perinatal growth deficits that are observed in CF mice are not due to decreased IGF-1 levels or differences in placenta-mediated fluid exchange. Further investigation is necessary to understand the etiology of early growth deficits in CF, as growth has been shown to be a significant factor in disease outcomes.
Collapse
Affiliation(s)
- Rebecca Darrah
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Ilya Bederman
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Megan Vitko
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Dana M. Valerio
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mitchell L. Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Craig A. Hodges
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|