1
|
Ulloa-Aguirre A, Zariñán T, Gutiérrez-Sagal R, Tao YX. Targeting trafficking as a therapeutic avenue for misfolded GPCRs leading to endocrine diseases. Front Endocrinol (Lausanne) 2022; 13:934685. [PMID: 36093106 PMCID: PMC9452723 DOI: 10.3389/fendo.2022.934685] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/13/2022] [Indexed: 02/05/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are plasma membrane proteins associated with an array of functions. Mutations in these receptors lead to a number of genetic diseases, including diseases involving the endocrine system. A particular subset of loss-of-function mutant GPCRs are misfolded receptors unable to traffic to their site of function (i.e. the cell surface plasma membrane). Endocrine disorders in humans caused by GPCR misfolding include, among others, hypo- and hyper-gonadotropic hypogonadism, morbid obesity, familial hypocalciuric hypercalcemia and neonatal severe hyperparathyroidism, X-linked nephrogenic diabetes insipidus, congenital hypothyroidism, and familial glucocorticoid resistance. Several in vitro and in vivo experimental approaches have been employed to restore function of some misfolded GPCRs linked to endocrine disfunction. The most promising approach is by employing pharmacological chaperones or pharmacoperones, which assist abnormally and incompletely folded proteins to refold correctly and adopt a more stable configuration to pass the scrutiny of the cell's quality control system, thereby correcting misrouting. This review covers the most important aspects that regulate folding and traffic of newly synthesized proteins, as well as the experimental approaches targeted to overcome protein misfolding, with special focus on GPCRs involved in endocrine diseases.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
- *Correspondence: Alfredo Ulloa-Aguirre,
| | - Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology & Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| |
Collapse
|
2
|
Scano M, Benetollo A, Nogara L, Bondì M, Barba FD, Soardi M, Furlan S, Akyurek EE, Caccin P, Carotti M, Sacchetto R, Blaauw B, Sandonà D. CFTR corrector C17 is effective in muscular dystrophy, in vivo proof of concept in LGMDR3. Hum Mol Genet 2021; 31:499-509. [PMID: 34505136 PMCID: PMC8863415 DOI: 10.1093/hmg/ddab260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Limb-girdle muscular dystrophy R3 (LGMDR3) is caused by mutations in the SGCA gene coding for α-sarcoglycan (SG). Together with β- γ- and δ-SG, α-SG forms a tetramer embedded in the dystrophin associated protein complex crucial for protecting the sarcolemma from mechanical stresses elicited by muscle contraction. Most LGMDR3 cases are due to missense mutations, which result in non-properly folded, even though potentially functional α-SG. These mutants are prematurely discarded by the cell quality control. Lacking one subunit, the SG-complex is disrupted. The resulting loss of function leads to sarcolemma instability, muscle fiber damage and progressive limb muscle weakness. LGMDR3 is severely disabling and, unfortunately, still incurable. Here, we propose the use of small molecules, belonging to the class of cystic fibrosis transmembrane regulator (CFTR) correctors, for recovering mutants of α-SG defective in folding and trafficking. Specifically, CFTR corrector C17 successfully rerouted the SG-complex containing the human R98H-α-SG to the sarcolemma of hind-limb muscles of a novel LGMDR3 murine model. Notably, the muscle force of the treated model animals was fully recovered. To our knowledge, this is the first time that a compound designated for cystic fibrosis is successfully tested in a muscular dystrophy and may represent a novel paradigm of treatment for LGMDR3 as well as different other indications in which a potentially functional protein is prematurely discarded as folding-defective. Furthermore, the use of small molecules for recovering the endogenous mutated SG has an evident advantage over complex procedures such as gene or cell transfer.
Collapse
Affiliation(s)
- Martina Scano
- Department of Biomedical Sciences, University of Padova, Italy
| | | | - Leonardo Nogara
- Venetian Institute of Molecular Medicine, University of Padova, Italy
| | - Michela Bondì
- Department of Biomedical Sciences, University of Padova, Italy
| | | | - Michela Soardi
- Department of Biomedical Sciences, University of Padova, Italy
| | - Sandra Furlan
- Neuroscience Institute - Italian National Research Council (CNR), Italy
| | - Eylem Emek Akyurek
- Department of Comparative Biomedicine and Food Science, University of Padova, Italy
| | - Paola Caccin
- Department of Biomedical Sciences, University of Padova, Italy
| | | | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padova, Italy.,Venetian Institute of Molecular Medicine, University of Padova, Italy
| | | |
Collapse
|
3
|
Centko RM, Carlile GW, Barne I, Patrick BO, Blagojevic P, Thomas DY, Andersen RJ. Combination of Selective PARP3 and PARP16 Inhibitory Analogues of Latonduine A Corrects F508del-CFTR Trafficking. ACS OMEGA 2020; 5:25593-25604. [PMID: 33073085 PMCID: PMC7557227 DOI: 10.1021/acsomega.0c02467] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
The marine natural product latonduine A (1) shows F508del-cystic fibrosis transmembrane regulator (CFTR) corrector activity in cell-based assays. Pull-down experiments, enzyme inhibition assays, and siRNA knockdown experiments suggest that the F508del-CFTR corrector activities of latonduine A and a synthetic analogue MCG315 (4) result from simultaneous inhibition of PARP3 and PARP16. A library of synthetic latonduine A analogs has been prepared in an attempt to separate the PARP3 and PARP16 inhibitory properties of latonduine A with the goal of discovering selective small-molecule PARP3 and PARP16 inhibitory cell biology tools that could confirm the proposed dual-target F508del-CFTR corrector mechanism of action. The structure activity relationship (SAR) study reported herein has resulted in the discovery of the modestly potent (IC50 3.1 μM) PARP3 selective inhibitor (±)-5-hydroxy-4-phenyl-2,3,4,5-tetrahydro-1H-benzo[c]azepin-1-one (5) that shows 96-fold greater potency for inhibition of PARP3 compared with its inhibition of PARP16 in vitro and the potent (IC50 0.362 μM) PARP16 selective inhibitor (±)-7,8-dichloro-5-hydroxy-4-(pyridin-2-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-1-one (6) that shows 205-fold selectivity for PARP16 compared with PARP3 in vitro. At 1 or 10 μM, neither 5 or 6 alone showed F508del-CFTR corrector activity, but when added together at 1 or 10 μM each, the combination exhibited F508del-CFTR corrector activity identical to 1 or 10 μM latonduine A (1), respectively, supporting its novel dual PARP target mechanism of action. Latonduine A (1) showed additive in vitro corrector activity in combination with the clinically approved corrector VX809, making it a potential new partner for cystic fibrosis combination drug therapies.
Collapse
Affiliation(s)
- Ryan M. Centko
- Department
of Chemistry and Department of Earth, Ocean & Atmospheric
Sciences, University of British Columbia, Vancouver, British Columbia, Canada V6T1Z1
| | - Graeme W. Carlile
- Departments
of Biochemistry and Human Genetics and The Cystic Fibrosis Translational
Research Centre, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Isabel Barne
- Department
of Chemistry and Department of Earth, Ocean & Atmospheric
Sciences, University of British Columbia, Vancouver, British Columbia, Canada V6T1Z1
| | - Brian O. Patrick
- Department
of Chemistry and Department of Earth, Ocean & Atmospheric
Sciences, University of British Columbia, Vancouver, British Columbia, Canada V6T1Z1
| | - Polina Blagojevic
- Department
of Chemistry and Department of Earth, Ocean & Atmospheric
Sciences, University of British Columbia, Vancouver, British Columbia, Canada V6T1Z1
| | - David Y. Thomas
- Departments
of Biochemistry and Human Genetics and The Cystic Fibrosis Translational
Research Centre, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Raymond J. Andersen
- Department
of Chemistry and Department of Earth, Ocean & Atmospheric
Sciences, University of British Columbia, Vancouver, British Columbia, Canada V6T1Z1
| |
Collapse
|
4
|
Combined Use of CFTR Correctors in LGMD2D Myotubes Improves Sarcoglycan Complex Recovery. Int J Mol Sci 2020; 21:ijms21051813. [PMID: 32155735 PMCID: PMC7084537 DOI: 10.3390/ijms21051813] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/28/2020] [Indexed: 12/28/2022] Open
Abstract
Sarcoglycanopathies are rare limb girdle muscular dystrophies, still incurable, even though symptomatic treatments may slow down the disease progression. Most of the disease-causing defects are missense mutations leading to a folding defective protein, promptly removed by the cell’s quality control, even if possibly functional. Recently, we repurposed small molecules screened for cystic fibrosis as potential therapeutics in sarcoglycanopathy. Indeed, cystic fibrosis transmembrane regulator (CFTR) correctors successfully recovered the defective sarcoglycan-complex in vitro. Our aim was to test the combined administration of some CFTR correctors with C17, the most effective on sarcoglycans identified so far, and evaluate the stability of the rescued sarcoglycan-complex. We treated differentiated myogenic cells from both sarcoglycanopathy and healthy donors, evaluating the global rescue and the sarcolemma localization of the mutated protein, by biotinylation assays and western blot analyses. We observed the additive/synergistic action of some compounds, gathering the first ideas on possible mechanism/s of action. Our data also suggest that a defective α-sarcoglycan is competent for assembly into the complex that, if helped in cell traffic, can successfully reach the sarcolemma. In conclusion, our results strengthen the idea that CFTR correctors, acting probably as proteostasis modulators, have the potential to progress as therapeutics for sarcoglycanopathies caused by missense mutations.
Collapse
|
5
|
Deciphering the role of protein kinase CK2 in the maturation/stability of F508del-CFTR. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165611. [DOI: 10.1016/j.bbadis.2019.165611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 09/16/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022]
|
6
|
Protein Misfolding and Endoplasmic Reticulum Stress in Chronic Lung Disease: Will Cell-Specific Targeting Be the Key to the Cure? Chest 2019; 157:1207-1220. [PMID: 31778676 DOI: 10.1016/j.chest.2019.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic lung disease accounts for a significant global burden with respect to death, disability, and health-care costs. Due to the heterogeneous nature and limited treatment options for these diseases, it is imperative that the cellular and molecular mechanisms underlying the disease pathophysiology are further understood. The lung is a complex organ with a diverse cell population, and each cell type will likely have different roles in disease initiation, progression, and resolution. The effectiveness of a given therapeutic agent may depend on the net effect on each of these cell types. Over the past decade, it has been established that endoplasmic reticulum stress and the unfolded protein response are involved in the development of several chronic lung diseases. These conserved cellular pathways are important for maintaining cellular proteostasis, but their aberrant activation can result in pathology. This review discusses the current understanding of endoplasmic reticulum stress and the unfolded protein response at the cellular level in the development and progression of various chronic lung diseases. We highlight the need for increased understanding of the specific cellular contributions of unfolded protein response activation to these pathologies and suggest that the development of cell-specific targeted therapies is likely required to further decrease disease progression and to promote resolution of chronic lung disease.
Collapse
|
7
|
Djouadi F, Bastin J. Mitochondrial Genetic Disorders: Cell Signaling and Pharmacological Therapies. Cells 2019; 8:cells8040289. [PMID: 30925787 PMCID: PMC6523966 DOI: 10.3390/cells8040289] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/19/2019] [Accepted: 03/23/2019] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial fatty acid oxidation (FAO) and respiratory chain (RC) defects form a large group of inherited monogenic disorders sharing many common clinical and pathophysiological features, including disruption of mitochondrial bioenergetics, but also, for example, oxidative stress and accumulation of noxious metabolites. Interestingly, several transcription factors or co-activators exert transcriptional control on both FAO and RC genes, and can be activated by small molecules, opening to possibly common therapeutic approaches for FAO and RC deficiencies. Here, we review recent data on the potential of various drugs or small molecules targeting pivotal metabolic regulators: peroxisome proliferator activated receptors (PPARs), sirtuin 1 (SIRT1), AMP-activated protein kinase (AMPK), and protein kinase A (PKA)) or interacting with reactive oxygen species (ROS) signaling, to alleviate or to correct inborn FAO or RC deficiencies in cellular or animal models. The possible molecular mechanisms involved, in particular the contribution of mitochondrial biogenesis, are discussed. Applications of these pharmacological approaches as a function of genotype/phenotype are also addressed, which clearly orient toward personalized therapy. Finally, we propose that beyond the identification of individual candidate drugs/molecules, future pharmacological approaches should consider their combination, which could produce additive or synergistic effects that may further enhance their therapeutic potential.
Collapse
Affiliation(s)
- Fatima Djouadi
- Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006 Paris, France.
| | - Jean Bastin
- Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006 Paris, France.
| |
Collapse
|
8
|
Bodas M, Vij N. Adapting Proteostasis and Autophagy for Controlling the Pathogenesis of Cystic Fibrosis Lung Disease. Front Pharmacol 2019; 10:20. [PMID: 30774592 PMCID: PMC6367269 DOI: 10.3389/fphar.2019.00020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Cystic fibrosis (CF), a fatal genetic disorder predominant in the Caucasian population, is caused by mutations in the cystic fibrosis transmembrane conductance regulator (Cftr) gene. The most common mutation is the deletion of phenylalanine from the position-508 (F508del-CFTR), resulting in a misfolded-CFTR protein, which is unable to fold, traffic and retain its plasma membrane (PM) localization. The resulting CFTR dysfunction, dysregulates variety of key cellular mechanisms such as chloride ion transport, airway surface liquid (ASL) homeostasis, mucociliary-clearance, inflammatory-oxidative signaling, and proteostasis that includes ubiquitin-proteasome system (UPS) and autophagy. A collective dysregulation of these key homoeostatic mechanisms contributes to the development of chronic obstructive cystic fibrosis lung disease, instead of the classical belief focused exclusively on ion-transport defect. Hence, therapeutic intervention(s) aimed at rescuing chronic CF lung disease needs to correct underlying defect that mediates homeostatic dysfunctions and not just chloride ion transport. Since targeting all the myriad defects individually could be quite challenging, it will be prudent to identify a process which controls almost all disease-promoting processes in the CF airways including underlying CFTR dysfunction. There is emerging experimental and clinical evidence that supports the notion that impaired cellular proteostasis and autophagy plays a central role in regulating pathogenesis of chronic CF lung disease. Thus, correcting the underlying proteostasis and autophagy defect in controlling CF pulmonary disease, primarily via correcting the protein processing defect of F508del-CFTR protein has emerged as a novel intervention strategy. Hence, we discuss here both the rationale and significant therapeutic utility of emerging proteostasis and autophagy modulating drugs/compounds in controlling chronic CF lung disease, where targeted delivery is a critical factor-influencing efficacy.
Collapse
Affiliation(s)
- Manish Bodas
- Department of Medicine, University of Oklahoma, Oklahoma City, OK, United States
| | - Neeraj Vij
- Department of Pediatric Pulmonary Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- 4Dx Limited, Los Angeles, CA, United States
- VIJ Biotech LLC, Baltimore, MD, United States
| |
Collapse
|
9
|
Carotti M, Marsolier J, Soardi M, Bianchini E, Gomiero C, Fecchio C, Henriques SF, Betto R, Sacchetto R, Richard I, Sandonà D. Repairing folding-defective α-sarcoglycan mutants by CFTR correctors, a potential therapy for limb-girdle muscular dystrophy 2D. Hum Mol Genet 2019; 27:969-984. [PMID: 29351619 PMCID: PMC5886177 DOI: 10.1093/hmg/ddy013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/30/2017] [Indexed: 11/22/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2D (LGMD2D) is a rare autosomal-recessive disease, affecting striated muscle, due to mutation of SGCA, the gene coding for α-sarcoglycan. Nowadays, more than 50 different SGCA missense mutations have been reported. They are supposed to impact folding and trafficking of α-sarcoglycan because the defective polypeptide, although potentially functional, is recognized and disposed of by the quality control of the cell. The secondary reduction of α-sarcoglycan partners, β-, γ- and δ-sarcoglycan, disrupts a key membrane complex that, associated to dystrophin, contributes to assure sarcolemma stability during muscle contraction. The complex deficiency is responsible for muscle wasting and the development of a severe form of dystrophy. Here, we show that the application of small molecules developed to rescue ΔF508-CFTR trafficking, and known as CFTR correctors, also improved the maturation of several α-sarcoglycan mutants that were consequently rescued at the plasma membrane. Remarkably, in myotubes from a patient with LGMD2D, treatment with CFTR correctors induced the proper re-localization of the whole sarcoglycan complex, with a consequent reduction of sarcolemma fragility. Although the mechanism of action of CFTR correctors on defective α-sarcoglycan needs further investigation, this is the first report showing a quantitative and functional recovery of the sarcoglycan-complex in human pathologic samples, upon small molecule treatment. It represents the proof of principle of a pharmacological strategy that acts on the sarcoglycan maturation process and we believe it has a great potential to develop as a cure for most of the patients with LGMD2D.
Collapse
Affiliation(s)
- Marcello Carotti
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Justine Marsolier
- Genethon, Evry F-91002, France.,INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | - Michela Soardi
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Elisa Bianchini
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy.,Aptuit, 37135 Verona, Italy
| | - Chiara Gomiero
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, 35020 Legnaro, Padova, Italy
| | - Chiara Fecchio
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Sara F Henriques
- Genethon, Evry F-91002, France.,INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | - Romeo Betto
- Neuroscience Institute (CNR Padova), 35131 Padova, Italy
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, 35020 Legnaro, Padova, Italy
| | - Isabelle Richard
- Genethon, Evry F-91002, France.,INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | - Dorianna Sandonà
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
10
|
Hou ZS, Ulloa-Aguirre A, Tao YX. Pharmacoperone drugs: targeting misfolded proteins causing lysosomal storage-, ion channels-, and G protein-coupled receptors-associated conformational disorders. Expert Rev Clin Pharmacol 2018; 11:611-624. [DOI: 10.1080/17512433.2018.1480367] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM) and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
11
|
Gene editing & stem cells. J Cyst Fibros 2017; 17:10-16. [PMID: 29233638 DOI: 10.1016/j.jcf.2017.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 12/26/2022]
|
12
|
Delion M, Braux J, Jourdain ML, Guillaume C, Bour C, Gangloff S, Pimpec-Barthes FL, Sermet-Gaudelus I, Jacquot J, Velard F. Overexpression of RANKL in osteoblasts: a possible mechanism of susceptibility to bone disease in cystic fibrosis. J Pathol 2017; 240:50-60. [PMID: 27235726 DOI: 10.1002/path.4753] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/09/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Bone fragility and loss are a significant cause of morbidity in patients with cystic fibrosis (CF), and the lack of effective therapeutic options means that treatment is more often palliative rather than curative. A deeper understanding of the pathogenesis of CF-related bone disease (CFBD) is necessary to develop new therapies. Defective CF transmembrane conductance regulator (CFTR) protein and chronic inflammation in bone are important components of the CFBD development. The receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG) drive the regulation of bone turnover. To investigate their roles in CFBD, we evaluated the involvement of defective CFTR in their production level in CF primary human osteoblasts with and without inflammatory stimulation, in the presence or not of pharmacological correctors of the CFTR. No major difference in cell ultrastructure was noted between cultured CF and non-CF osteoblasts, but a delayed bone matrix mineralization was observed in CF osteoblasts. Strikingly, resting CF osteoblasts exhibited strong production of RANKL protein, which was highly localized at the cell membrane and was enhanced in TNF (TNF-α) or IL-17-stimulated conditions. Under TNF stimulation, a defective response in OPG production was observed in CF osteoblasts in contrast to the elevated OPG production of non-CF osteoblasts, leading to an elevated RANKL-to-OPG protein ratio in CF osteoblasts. Pharmacological inhibition of CFTR chloride channel conductance in non-CF osteoblasts replicated both the decreased OPG production and the enhanced RANKL-to-OPG ratio. Interestingly, using CFTR correctors such as C18, we significantly reduced the production of RANKL by CF osteoblasts, in both resting and TNF-stimulated conditions. In conclusion, the overexpression of RANKL and high membranous RANKL localization in osteoblasts are related to defective CFTR, and may worsen bone resorption, leading to bone loss in patients with CF. Targeting osteoblasts with CFTR correctors may represent an effective strategy to treat CFBD. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Martial Delion
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Julien Braux
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Marie-Laure Jourdain
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Christine Guillaume
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Camille Bour
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Sophie Gangloff
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | | | - Isabelle Sermet-Gaudelus
- Unité de Pneumo-Pédiatrie Allergologie, Hôpital Necker, Inserm U1551, Université Paris Sorbonne, Paris, France
| | - Jacky Jacquot
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Frédéric Velard
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| |
Collapse
|
13
|
Venkatesh D, Boehm C, Barlow LD, Nankissoor NN, O'Reilly A, Kelly S, Dacks JB, Field MC. Evolution of the endomembrane systems of trypanosomatids - conservation and specialisation. J Cell Sci 2017; 130:1421-1434. [PMID: 28386020 PMCID: PMC5399786 DOI: 10.1242/jcs.197640] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/09/2017] [Indexed: 12/20/2022] Open
Abstract
Parasite surfaces support multiple functions required for survival within their hosts, and maintenance and functionality of the surface depends on membrane trafficking. To understand the evolutionary history of trypanosomatid trafficking, where multiple lifestyles and mechanisms of host interactions are known, we examined protein families central to defining intracellular compartments and mediating transport, namely Rabs, SNAREs and RabGAPs, across all available Euglenozoa genomes. Bodonids possess a large trafficking repertoire, which is mainly retained by the Trypanosoma cruzi group, with extensive losses in other lineages, particularly African trypanosomes and phytomonads. There are no large-scale expansions or contractions from an inferred ancestor, excluding direct associations between parasitism or host range. However, we observe stepwise secondary losses within Rab and SNARE cohorts (but not RabGAPs). Major changes are associated with endosomal and late exocytic pathways, consistent with the diversity in surface proteomes between trypanosomatids and mechanisms of interaction with the host. Along with the conserved core family proteins, several lineage-specific members of the Rab (but not SNARE) family were found. Significantly, testing predictions of SNARE complex composition by proteomics confirms generalised retention of function across eukaryotes.
Collapse
Affiliation(s)
- Divya Venkatesh
- Wellcome Trust Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK.,Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PQ, UK
| | - Cordula Boehm
- Wellcome Trust Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Lael D Barlow
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Nerissa N Nankissoor
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Amanda O'Reilly
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PQ, UK
| | - Steven Kelly
- Department of Plant Sciences, University of Oxford, Oxford OX1 6JP, UK
| | - Joel B Dacks
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Mark C Field
- Wellcome Trust Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
14
|
Ubiquitin Ligase RNF138 Promotes Episodic Ataxia Type 2-Associated Aberrant Degradation of Human Ca v2.1 (P/Q-Type) Calcium Channels. J Neurosci 2017; 37:2485-2503. [PMID: 28167673 DOI: 10.1523/jneurosci.3070-16.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/09/2017] [Accepted: 01/31/2017] [Indexed: 11/21/2022] Open
Abstract
Voltage-gated CaV2.1 channels comprise a pore-forming α1A subunit with auxiliary α2δ and β subunits. CaV2.1 channels play an essential role in regulating synaptic signaling. Mutations in the human gene encoding the CaV2.1 subunit are associated with the cerebellar disease episodic ataxia type 2 (EA2). Several EA2-causing mutants exhibit impaired protein stability and exert dominant-negative suppression of CaV2.1 wild-type (WT) protein expression via aberrant proteasomal degradation. Here, we set out to delineate the protein degradation mechanism of human CaV2.1 subunit by identifying RNF138, an E3 ubiquitin ligase, as a novel CaV2.1-binding partner. In neurons, RNF138 and CaV2.1 coexist in the same protein complex and display notable subcellular colocalization at presynaptic and postsynaptic regions. Overexpression of RNF138 promotes polyubiquitination and accelerates protein turnover of CaV2.1. Disrupting endogenous RNF138 function with a mutant (RNF138-H36E) or shRNA infection significantly upregulates the CaV2.1 protein level and enhances CaV2.1 protein stability. Disrupting endogenous RNF138 function also effectively rescues the defective protein expression of EA2 mutants, as well as fully reversing EA2 mutant-induced excessive proteasomal degradation of CaV2.1 WT subunits. RNF138-H36E coexpression only partially restores the dominant-negative effect of EA2 mutants on CaV2.1 WT functional expression, which can be attributed to defective membrane trafficking of CaV2.1 WT in the presence of EA2 mutants. We propose that RNF138 plays a critical role in the homeostatic regulation of CaV2.1 protein level and functional expression and that RNF138 serves as the primary E3 ubiquitin ligase promoting EA2-associated aberrant degradation of human CaV2.1 subunits.SIGNIFICANCE STATEMENT Loss-of-function mutations in the human CaV2.1 subunit are linked to episodic ataxia type 2 (EA2), a dominantly inherited disease characterized by paroxysmal attacks of ataxia and nystagmus. EA2-causing mutants may exert dominant-negative effects on the CaV2.1 wild-type subunit via aberrant proteasomal degradation. The molecular nature of the CaV2.1 ubiquitin-proteasome degradation pathway is currently unknown. The present study reports the first identification of an E3 ubiquitin ligase for CaV2.1, RNF138. CaV2.1 protein stability is dynamically regulated by RNF138 and auxiliary α2δ and β subunits. We provide a proof of concept that protecting the human CaV2.1 subunit from excessive proteasomal degradation with specific interruption of endogenous RNF138 function may partially contribute to the future development of a novel therapeutic strategy for EA2 patients.
Collapse
|
15
|
Bourdin B, Segura E, Tétreault MP, Lesage S, Parent L. Determination of the Relative Cell Surface and Total Expression of Recombinant Ion Channels Using Flow Cytometry. J Vis Exp 2016. [PMID: 27768059 DOI: 10.3791/54732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Inherited or de novo mutations in cation-selective channels may lead to sudden cardiac death. Alteration in the plasma membrane trafficking of these multi-spanning transmembrane proteins, with or without change in channel gating, is often postulated to contribute significantly in this process. It has thus become critical to develop a method to quantify the change of the relative cell surface expression of cardiac ion channels on a large scale. Herein, a detailed protocol is provided to determine the relative total and cell surface expression of cardiac L-type calcium channels CaV1.2 and membrane-associated subunits in tsA-201 cells using two-color fluorescent cytometry assays. Compared with other microscopy-based or immunoblotting-based qualitative methods, flow cytometry experiments are fast, reproducible, and large-volume assays that deliver quantifiable end-points on large samples of live cells (ranging from 104 to 106 cells) with similar cellular characteristics in a single flow. Constructs were designed to constitutively express mCherry at the intracellular C-terminus (thus allowing a rapid assessment of the total protein expression) and express an extracellular-facing hemagglutinin (HA) epitope to estimate the cell surface expression of membrane proteins using an anti-HA fluorescence conjugated antibody. To avoid false negative, experiments were also conducted in permeabilized cells to confirm the accessibility and proper expression of the HA epitope. The detailed procedure provides: (1) design of tagged DNA (deoxyribonucleic acid) constructs, (2) lipid-mediated transfection of constructs in tsA-201 cells, (3) culture, harvest, and staining of non-permeabilized and permeabilized cells, and (4) acquisition and analysis of fluorescent signals. Additionally, the basic principles of flow cytometry are explained and the experimental design, including the choice of fluorophores, titration of the HA antibody and control experiments, is thoroughly discussed. This specific approach offers objective relative quantification of the total and cell surface expression of ion channels that can be extended to study ion pumps and plasma membrane transporters.
Collapse
Affiliation(s)
- Benoîte Bourdin
- Département de Physiologie Moléculaire et Intégrative, Montreal Heart Institute Research Centre
| | - Emilie Segura
- Département de Physiologie Moléculaire et Intégrative, Montreal Heart Institute Research Centre
| | | | - Sylvie Lesage
- Département de Microbiologie, Infectiologie, Immunologie, Centre de recherche de l'Hôpital Maisonneuve-Rosemont
| | - Lucie Parent
- Département de Physiologie Moléculaire et Intégrative, Montreal Heart Institute Research Centre;
| |
Collapse
|
16
|
Peng YJ, Huang JJ, Wu HH, Hsieh HY, Wu CY, Chen SC, Chen TY, Tang CY. Regulation of CLC-1 chloride channel biosynthesis by FKBP8 and Hsp90β. Sci Rep 2016; 6:32444. [PMID: 27580824 PMCID: PMC5007535 DOI: 10.1038/srep32444] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/10/2016] [Indexed: 01/23/2023] Open
Abstract
Mutations in human CLC-1 chloride channel are associated with the skeletal muscle disorder myotonia congenita. The disease-causing mutant A531V manifests enhanced proteasomal degradation of CLC-1. We recently found that CLC-1 degradation is mediated by cullin 4 ubiquitin ligase complex. It is currently unclear how quality control and protein degradation systems coordinate with each other to process the biosynthesis of CLC-1. Herein we aim to ascertain the molecular nature of the protein quality control system for CLC-1. We identified three CLC-1-interacting proteins that are well-known heat shock protein 90 (Hsp90)-associated co-chaperones: FK506-binding protein 8 (FKBP8), activator of Hsp90 ATPase homolog 1 (Aha1), and Hsp70/Hsp90 organizing protein (HOP). These co-chaperones promote both the protein level and the functional expression of CLC-1 wild-type and A531V mutant. CLC-1 biosynthesis is also facilitated by the molecular chaperones Hsc70 and Hsp90β. The protein stability of CLC-1 is notably increased by FKBP8 and the Hsp90β inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) that substantially suppresses cullin 4 expression. We further confirmed that cullin 4 may interact with Hsp90β and FKBP8. Our data are consistent with the idea that FKBP8 and Hsp90β play an essential role in the late phase of CLC-1 quality control by dynamically coordinating protein folding and degradation.
Collapse
Affiliation(s)
- Yi-Jheng Peng
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jing-Jia Huang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hao-Han Wu
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Ying Hsieh
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Ying Wu
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Ching Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsung-Yu Chen
- Neuroscience Center, University of California, Davis, USA
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Jacquot J, Delion M, Gangloff S, Braux J, Velard F. Bone disease in cystic fibrosis: new pathogenic insights opening novel therapies. Osteoporos Int 2016; 27:1401-1412. [PMID: 26431978 DOI: 10.1007/s00198-015-3343-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/24/2015] [Indexed: 01/17/2023]
Abstract
Mutations within the gene encoding for the chloride ion channel cystic fibrosis transmembrane conductance regulator (CFTR) results in cystic fibrosis (CF), the most common lethal autosomal recessive genetic disease that causes a number of long-term health problems, as the bone disease. Osteoporosis and increased vertebral fracture risk associated with CF disease are becoming more important as the life expectancy of patients continues to improve. The etiology of low bone density is multifactorial, most probably a combination of inadequate peak bone mass during puberty and increased bone losses in adults. Body mass index, male sex, advanced pulmonary disease, malnutrition and chronic therapies are established additional risk factors for CF-related bone disease (CFBD). Consistently, recent evidence has confirmed that CFTR plays a major role in the osteoprotegerin (OPG) and COX-2 metabolite prostaglandin E2 (PGE2) production, two key regulators in the bone formation and regeneration. Several others mechanisms were also recognized from animal and cell models contributing to malfunctions of osteoblast (cell that form bone) and indirectly of bone-resorpting osteoclasts. Understanding such mechanisms is crucial for the development of therapies in CFBD. Innovative therapeutic approaches using CFTR modulators such as C18 have recently shown in vitro capacity to enhance PGE2 production and normalized the RANKL-to-OPG ratio in human osteoblasts bearing the mutation F508del-CFTR and therefore potential clinical utility in CFBD. This review focuses on the recently identified pathogenic mechanisms leading to CFBD and potential future therapies for treating CFBD.
Collapse
Affiliation(s)
- J Jacquot
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR CAP-Santé (FED 4231), Université Reims Champagne Ardenne, 1, Avenue du Maréchal Juin, 51095, Reims, France.
| | | | | | | | | |
Collapse
|
18
|
Nieddu E, Pollarolo B, Mazzei MT, Anzaldi M, Schenone S, Pedemonte N, Galietta LJV, Mazzei M. Phenylhydrazones as Correctors of a Mutant Cystic Fibrosis Transmembrane Conductance Regulator. Arch Pharm (Weinheim) 2015; 349:112-23. [DOI: 10.1002/ardp.201500352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Erika Nieddu
- Department of Pharmacy; University of Genova; Genova Italy
| | | | | | - Maria Anzaldi
- Department of Pharmacy; University of Genova; Genova Italy
| | | | | | | | - Mauro Mazzei
- Department of Pharmacy; University of Genova; Genova Italy
| |
Collapse
|
19
|
Martin U. Pluripotent stem cells for disease modeling and drug screening: new perspectives for treatment of cystic fibrosis? Mol Cell Pediatr 2015; 2:15. [PMID: 26666881 PMCID: PMC4678132 DOI: 10.1186/s40348-015-0023-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/16/2015] [Indexed: 12/13/2022] Open
Abstract
Despite continuous improvements in treating clinical symptoms and the identification of single compounds that effectively rescue some rare mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), associated lung and liver pathologies remain largely untreatable and no real breakthrough is visible for the majority of patients suffering from cystic fibrosis (CF). Novel compounds have to be identified and tailored in combination to specific CFTR mutations, to different tissues, or even to the individual patient. Immortalized cell lines overexpressing mutant CFTR are typically used to screen candidate molecules but have proven to be poor predictors of clinical efficacy. The complexity of CFTR maturation and turnover requires the use of cellular models that closely recapitulate the specific properties of the clinically most affected organs. Importantly, current screening efforts based on primary airway cells or intestinal organoids cannot specifically target single rare CFTR mutations or mimic multiple cell types. In the near future, genetically engineered induced pluripotent stem cells will provide an excellent basis for personalized organotypic models of CF disease and biological screens for identification of CFTR potentiators and correctors.
Collapse
Affiliation(s)
- Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany. .,REBIRTH Cluster of Excellence, Hannover, Germany. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
20
|
McShane AJ, Bajrami B, Ramos AA, Diego-Limpin PA, Farrokhi V, Coutermarsh BA, Stanton BA, Jensen T, Riordan JR, Wetmore D, Joseloff E, Yao X. Targeted proteomic quantitation of the absolute expression and turnover of cystic fibrosis transmembrane conductance regulator in the apical plasma membrane. J Proteome Res 2014; 13:4676-85. [PMID: 25227318 PMCID: PMC4227562 DOI: 10.1021/pr5006795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Deficient
chloride transport through cystic fibrosis (CF) transmembrane
conductance regulator (CFTR) causes lethal complications in CF patients.
CF is the most common autosomal recessive genetic disease, which is
caused by mutations in the CFTR gene; thus, CFTR mutants can serve
as primary targets for drugs to modulate and rescue the ion channel’s
function. The first step of drug modulation is to increase the expression
of CFTR in the apical plasma membrane (PM); thus, accurate measurement
of CFTR in the PM is desired. This work reports a tandem enrichment
strategy to prepare PM CFTR and uses a stable isotope labeled CFTR
sample as the quantitation reference to measure the absolute amount
of apical PM expression of CFTR in CFBE 41o- cells. It was found that
CFBE 41o- cells expressing wild-type CFTR (wtCFTR), when cultured
on plates, had 2.9 ng of the protein in the apical PM per million
cells; this represented 10% of the total CFTR found in the cells.
When these cells were polarized on filters, the apical PM expression
of CFTR increased to 14%. Turnover of CFTR in the apical PM of baby
hamster kidney cells overexpressing wtCFTR (BHK-wtCFTR) was also quantified
by targeted proteomics based on multiple reaction monitoring mass
spectrometry; wtCFTR had a half-life of 29.0 ± 2.5 h in the apical
PM. This represents the first direct measurement of CFTR turnover
using stable isotopes. The absolute quantitation and turnover measurements
of CFTR in the apical PM can significantly facilitate understanding
the disease mechanism of CF and thus the development of new disease-modifying
drugs. Absolute CFTR quantitation allows for direct result comparisons
among analyses, analysts, and laboratories and will greatly amplify
the overall outcome of CF research and therapy.
Collapse
Affiliation(s)
- Adam J McShane
- Department of Chemistry, University of Connecticut , Storrs, Connecticut 06269, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Myint M, Limberis MP, Bell P, Somanathan S, Haczku A, Wilson JM, Diamond SL. In vivo evaluation of adeno-associated virus gene transfer in airways of mice with acute or chronic respiratory infection. Hum Gene Ther 2014; 25:966-76. [PMID: 25144316 DOI: 10.1089/hum.2014.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Patients with cystic fibrosis (CF) often suffer chronic lung infection with concomitant inflammation, a setting that may reduce the efficacy of gene transfer. While gene therapy development for CF often involves viral-based vectors, little is known about gene transfer in the context of an infected airway. In this study, three mouse models were established to evaluate adeno-associated virus (AAV) gene transfer in such an environment. Bordetella bronchiseptica RB50 was used in a chronic, nonlethal respiratory infection in C57BL/6 mice. An inoculum of ∼10(5) CFU allowed B. bronchiseptica RB50 to persist in the upper and lower respiratory tracts for at least 21 days. In this infection model, administration of an AAV vector on day 2 resulted in 2.8-fold reduction of reporter gene expression compared with that observed in uninfected controls. Postponement of AAV administration to day 14 resulted in an even greater (eightfold) reduction of reporter gene expression, when compared with uninfected controls. In another infection model, Pseudomonas aeruginosa PAO1 was used to infect surfactant protein D (SP-D) or surfactant protein A (SP-A) knockout (KO) mice. With an inoculum of ∼10(5) CFU, infection persisted for 2 days in the nasal cavity of either mouse model. Reporter gene expression was approximately ∼2.5-fold lower compared with uninfected mice. In the SP-D KO model, postponement of AAV administration to day 9 postinfection resulted in only a two fold reduction in reporter gene expression, when compared with expression seen in uninfected controls. These results confirm that respiratory infections, both ongoing and recently resolved, decrease the efficacy of AAV-mediated gene transfer.
Collapse
Affiliation(s)
- Melissa Myint
- 1 Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania , Philadelphia, PA 19104
| | | | | | | | | | | | | |
Collapse
|
22
|
Boucherle B, Bertrand J, Maurin B, Renard BL, Fortuné A, Tremblier B, Becq F, Norez C, Décout JL. A new 9-alkyladenine-cyclic methylglyoxal diadduct activates wt- and F508del-cystic fibrosis transmembrane conductance regulator (CFTR) in vitro and in vivo. Eur J Med Chem 2014; 83:455-65. [PMID: 24992073 DOI: 10.1016/j.ejmech.2014.06.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 01/28/2023]
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is the main chloride channel present in the apical membrane of epithelial cells and the F508 deletion (F508del-CFTR) in the CF gene is the most common cystic fibrosis-causing mutation. In the search for a pharmacotherapy of cystic fibrosis caused by the F508del-CFTR, a bi-therapy could be developed associating a corrector of F508del-CFTR trafficking and an activator of the channel activity of CFTR. Here, we report on the synthesis of 9-alkyladenine derivatives analogues of our previously discovered activator of wt-CFTR and F508del-CFTR, GPact-11a, and the identification of a new activator of these channels, GPact-26a, through various flux assays on human airway epithelial CF and non-CF cell lines and in vivo measurement of rat salivary secretion. This study reveals that the possible modifications of the side chain introduced at the N9 position of the main pharmacophore are highly limited since only an allyl group can replace the propyl side chain present in GPact-11a to lead to a strong activation of wt-CFTR in CHO cells. Docking simulations of the synthesised compounds and of four described modulators performed using a 3D model of the wt-type CFTR protein suggest five possible binding sites located at the interface of the nucleotide binding domains NBD1/NBD2. However, the docking study did not allow the differentiation between active and non-active compounds.
Collapse
Affiliation(s)
- Benjamin Boucherle
- Université Grenoble Alpes, Joseph Fourier/CNRS, UMR 5063, Département de Pharmacochimie Moléculaire, 470 rue de la Chimie, F-38041 Grenoble, France
| | - Johanna Bertrand
- Université de Poitiers/CNRS, Laboratoire Signalisation et Transports Ioniques Membranaires, 1 rue Georges Bonnet, F-86022 Poitiers, France
| | - Bruno Maurin
- Université Grenoble Alpes, Joseph Fourier/CNRS, UMR 5063, Département de Pharmacochimie Moléculaire, 470 rue de la Chimie, F-38041 Grenoble, France
| | - Brice-Loïc Renard
- Université Grenoble Alpes, Joseph Fourier/CNRS, UMR 5063, Département de Pharmacochimie Moléculaire, 470 rue de la Chimie, F-38041 Grenoble, France
| | - Antoine Fortuné
- Université Grenoble Alpes, Joseph Fourier/CNRS, UMR 5063, Département de Pharmacochimie Moléculaire, 470 rue de la Chimie, F-38041 Grenoble, France
| | - Brice Tremblier
- Université de Poitiers/CNRS, Laboratoire Signalisation et Transports Ioniques Membranaires, 1 rue Georges Bonnet, F-86022 Poitiers, France
| | - Frédéric Becq
- Université de Poitiers/CNRS, Laboratoire Signalisation et Transports Ioniques Membranaires, 1 rue Georges Bonnet, F-86022 Poitiers, France
| | - Caroline Norez
- Université de Poitiers/CNRS, Laboratoire Signalisation et Transports Ioniques Membranaires, 1 rue Georges Bonnet, F-86022 Poitiers, France
| | - Jean-Luc Décout
- Université Grenoble Alpes, Joseph Fourier/CNRS, UMR 5063, Département de Pharmacochimie Moléculaire, 470 rue de la Chimie, F-38041 Grenoble, France.
| |
Collapse
|
23
|
Compain P, Bodlenner A. The Multivalent Effect in Glycosidase Inhibition: A New, Rapidly Emerging Topic in Glycoscience. Chembiochem 2014; 15:1239-51. [DOI: 10.1002/cbic.201402026] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Indexed: 11/07/2022]
|
24
|
Correction of chloride transport and mislocalization of CFTR protein by vardenafil in the gastrointestinal tract of cystic fibrosis mice. PLoS One 2013; 8:e77314. [PMID: 24204804 PMCID: PMC3811977 DOI: 10.1371/journal.pone.0077314] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 09/07/2013] [Indexed: 12/13/2022] Open
Abstract
Although lung disease is the major cause of mortality in cystic fibrosis (CF), gastrointestinal (GI) manifestations are the first hallmarks in 15–20% of affected newborns presenting with meconium ileus, and remain major causes of morbidity throughout life. We have previously shown that cGMP-dependent phosphodiesterase type 5 (PDE5) inhibitors rescue defective CF Transmembrane conductance Regulator (CFTR)-dependent chloride transport across the mouse CF nasal mucosa. Using F508del-CF mice, we examined the transrectal potential difference 1 hour after intraperitoneal injection of the PDE5 inhibitor vardenafil or saline to assess the amiloride-sensitive sodium transport and the chloride gradient and forskolin-dependent chloride transport across the GI tract. In the same conditions, we performed immunohistostaining studies in distal colon to investigate CFTR expression and localization. F508del-CF mice displayed increased sodium transport and reduced chloride transport compared to their wild-type littermates. Vardenafil, applied at a human therapeutic dose (0.14 mg/kg) used to treat erectile dysfunction, increased chloride transport in F508del-CF mice. No effect on sodium transport was detected. In crypt colonocytes of wild-type mice, the immunofluorescence CFTR signal was mostly detected in the apical cell compartment. In F508del-CF mice, a 25% reduced signal was observed, located mostly in the subapical region. Vardenafil increased the peak of intensity of the fluorescence CFTR signal in F508del-CF mice and displaced it towards the apical cell compartment. Our findings point out the intestinal mucosa as a valuable tissue to study CFTR transport function and localization and to evaluate efficacy of therapeutic strategies in CF. From our data we conclude that vardenafil mediates potentiation of the CFTR chloride channel and corrects mislocalization of the mutant protein. The study provides compelling support for targeting the cGMP signaling pathway in CF pharmacotherapy.
Collapse
|
25
|
Patel AC. Clinical relevance of target identity and biology: implications for drug discovery and development. ACTA ACUST UNITED AC 2013; 18:1164-85. [PMID: 24080260 DOI: 10.1177/1087057113505906] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many of the most commonly used drugs precede techniques for target identification and drug specificity and were developed on the basis of efficacy and safety, an approach referred to as classical pharmacology and, more recently, phenotypic drug discovery. Although substantial gains have been made during the period of focus on target-based approaches, particularly in oncology, these approaches have suffered a high overall failure rate and lower productivity in terms of new drugs when compared with phenotypic approaches. This review considers the importance of target identity and biology in clinical practice from the prescriber's viewpoint. In evaluating influences on prescribing behavior, studies suggest that target identity and mechanism of action are not significant factors in drug choice. Rather, patients and providers consistently value efficacy, safety, and tolerability. Similarly, the Food and Drug Administration requires evidence of safety and efficacy for new drugs but does not require knowledge of drug target identity or target biology. Prescribers do favor drugs with novel mechanisms, but this preference is limited to diseases for which treatments are either not available or suboptimal. Thus, while understanding of drug target and target biology is important from a scientific perspective, it is not particularly important to prescribers, who prioritize efficacy and safety.
Collapse
Affiliation(s)
- Anand C Patel
- 1Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
26
|
Compain P, Decroocq C, Joosten A, de Sousa J, Rodríguez-Lucena D, Butters TD, Bertrand J, Clément R, Boinot C, Becq F, Norez C. Rescue of functional CFTR channels in cystic fibrosis: a dramatic multivalent effect using iminosugar cluster-based correctors. Chembiochem 2013; 14:2050-8. [PMID: 24038832 DOI: 10.1002/cbic.201300312] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Indexed: 12/23/2022]
Abstract
Cystic fibrosis is caused by a mutation in the gene for the cystic fibrosis transmembrane conductance regulator (CFTR) protein. N-butyl 1-deoxynojirimycin (N-Bu DNJ), a clinical candidate for the treatment of cystic fibrosis, is able to act as a CFTR corrector by overcoming the processing defect of the mutant protein. To explore the potential of multivalency on CFTR correction activity, a library of twelve DNJ click clusters with valencies ranging from 3 to 14 were synthesized. Significantly, the trivalent analogues were found to be up to 225-fold more potent than N-Bu DNJ and up to 1000-fold more potent than the corresponding monovalent models. These results provide the first description of a multivalent effect for correcting protein folding defects in cells and should have application for the treatment of a number of protein folding disorders. Preliminary mechanistic studies indicated that CFTR correction activity enhancement was not due to a multivalent effect in ER-glucosidase inhibition or to a different mode of action of the multivalent iminosugars.
Collapse
Affiliation(s)
- Philippe Compain
- Laboratoire de Synthèse Organique et Molécules Bioactives, Université de Strasbourg et CNRS (UMR 7509), Ecole Européenne de Chimie, Polymères et Matériaux, 25 rue Becquerel, 67087 Strasbourg (France); Institut Universitaire de France, 103 Bd Saint-Michel, 75005 Paris (France).
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|