1
|
Nicosia L, Pranke I, Latorre RV, Murray JB, Lonetti L, Cavusoglu-Doran K, Dreano E, Costello JP, Carroll M, Melotti P, Sorio C, Sermet-Gaudelus I, Scallan MF, Harrison PT. Adenine base editing with engineered virus-like particles rescues the CFTR mutation G542X in patient-derived intestinal organoids. iScience 2025; 28:111979. [PMID: 40144632 PMCID: PMC11938077 DOI: 10.1016/j.isci.2025.111979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/05/2024] [Accepted: 02/05/2025] [Indexed: 03/28/2025] Open
Abstract
Cystic fibrosis (CF) is a life-shortening autosomal recessive disease, caused by loss-of-function mutations that affect the CF transmembrane conductance regulator (CFTR) anion channel. G542X is the second-most common CF-causing variant, and it does not respond to current CFTR modulator drugs. Our study explores the use of adenine base editing to edit G542X to a non-CF-causing variant, G542R, and recover CFTR function. Using base editor engineered virus-like particles (BE-eVLPs) in patient-derived intestinal organoids, we achieved ∼2% G542X-to-G542R editing efficiency and restored CFTR-mediated chloride transport to ∼6.4% of wild-type levels, independent of modulator treatment, and with no bystander edits. This proof-of-principle study demonstrates the potential of base editing to rescue G542X and provides a foundation for future in - vivo applications.
Collapse
Affiliation(s)
- Lucia Nicosia
- Department of Physiology, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Iwona Pranke
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
| | | | - Joss B. Murray
- Department of Physiology, University College Cork, Cork, Ireland
| | - Lisa Lonetti
- Department of Physiology, University College Cork, Cork, Ireland
| | | | - Elise Dreano
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
| | | | - Michael Carroll
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paola Melotti
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Claudio Sorio
- Department of Medicine, University of Verona, Verona, Italy
| | - Isabelle Sermet-Gaudelus
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
- Cystic Fibrosis National Pediatric Reference Center, Pneumo-Allergologie Pédiatrique, Hôpital Necker Enfants Malades, , Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
- European Reference Network, ERN-Lung CF, Frankfurt am Mein, Germany
| | | | - Patrick T. Harrison
- Department of Physiology, University College Cork, Cork, Ireland
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| |
Collapse
|
2
|
Yıldız CA, Selçuk Balcı M, Karabulut Ş, Başer ZM, Yüksel Kalyoncu M, Metin Çakar N, Akkitap Yiğit MM, Baysal EE, Özdemircioğlu F, Uzunoğlu B, Taştan G, Ergenekon P, Gökdemir Y, Erdem Eralp E, Karakoç F, Ata P, Karadağ B. Beyond the 10%: Unraveling the genetic diversity in Turkish cystic fibrosis patients not eligible for CFTR modulators. Pediatr Pulmonol 2024; 59:3250-3259. [PMID: 39031495 PMCID: PMC11601005 DOI: 10.1002/ppul.27181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Cystic fibrosis (CF) is an autosomal recessive disease caused by variants of CFTR gene. Over 2000 variants have been identified, and new drugs called CFTR modulators have been developed to target specific defects in the CFTR protein. However, these drugs are only suitable for patients with certain variants of CFTR, and eligibility rates vary depending on race and geographical region. This study aimed to reveal the detailed genotype and clinical characteristics of people with CF (pwCF) at our center in Turkey, a developing country, who are not eligible for CFTR modulators. METHODS A total of 445 pwCF followed up at Marmara University were reviewed retrospectively. Variants of the patients ineligible to CFTR modulators were classified based on American College of Medical Genetics guidelines, CFTR classification, the change in the encoded protein, and the variant type. RESULTS The study revealed that 139 (31.2%) patients weren't eligible for CFTR modulators. There were 60 different variants in the 276 alleles, as two were missing. The majority of patients had missense or nonsense variants, and that the most common variant was c.1545_1546del, which can be said unique to this geography. CONCLUSION The study highlights the importance of detecting the variants of ineligible patients in detail to guide future approaches for more targeted and effective interventions in CF care. Testing the effectiveness of CFTR modulators for rare or newly occurring variants is crucial to ensure equal access for pwCF to these therapies from different racial backgrounds and ethnic minorities.
Collapse
Affiliation(s)
- Ceren Ayça Yıldız
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Merve Selçuk Balcı
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Şeyda Karabulut
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Zeynep Münteha Başer
- Department of Medical GeneticsMarmara University School of MedicineIstanbulTurkey
| | - Mine Yüksel Kalyoncu
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Neval Metin Çakar
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | | | - Eda Esra Baysal
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Fulya Özdemircioğlu
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Burcu Uzunoğlu
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Gamze Taştan
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Pınar Ergenekon
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Yasemin Gökdemir
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Ela Erdem Eralp
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Fazilet Karakoç
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| | - Pınar Ata
- Department of Medical GeneticsMarmara University School of MedicineIstanbulTurkey
| | - Bülent Karadağ
- Division of Pediatric PulmonologyMarmara University School of MedicineIstanbulTurkey
| |
Collapse
|
3
|
Morais P, Zhang R, Yu YT. Therapeutic Nonsense Suppression Modalities: From Small Molecules to Nucleic Acid-Based Approaches. Biomedicines 2024; 12:1284. [PMID: 38927491 PMCID: PMC11201248 DOI: 10.3390/biomedicines12061284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Nonsense mutations are genetic mutations that create premature termination codons (PTCs), leading to truncated, defective proteins in diseases such as cystic fibrosis, neurofibromatosis type 1, Dravet syndrome, Hurler syndrome, Beta thalassemia, inherited bone marrow failure syndromes, Duchenne muscular dystrophy, and even cancer. These mutations can also trigger a cellular surveillance mechanism known as nonsense-mediated mRNA decay (NMD) that degrades the PTC-containing mRNA. The activation of NMD can attenuate the consequences of truncated, defective, and potentially toxic proteins in the cell. Since approximately 20% of all single-point mutations are disease-causing nonsense mutations, it is not surprising that this field has received significant attention, resulting in a remarkable advancement in recent years. In fact, since our last review on this topic, new examples of nonsense suppression approaches have been reported, namely new ways of promoting the translational readthrough of PTCs or inhibiting the NMD pathway. With this review, we update the state-of-the-art technologies in nonsense suppression, focusing on novel modalities with therapeutic potential, such as small molecules (readthrough agents, NMD inhibitors, and molecular glue degraders); antisense oligonucleotides; tRNA suppressors; ADAR-mediated RNA editing; targeted pseudouridylation; and gene/base editing. While these various modalities have significantly advanced in their development stage since our last review, each has advantages (e.g., ease of delivery and specificity) and disadvantages (manufacturing complexity and off-target effect potential), which we discuss here.
Collapse
Affiliation(s)
- Pedro Morais
- Drug Metabolism and Pharmacokinetics, Research and Development, Bayer Pharmaceuticals, 42113 Wuppertal, Germany
| | - Rui Zhang
- Center for RNA Biology, Department of Biochemistry and Biophysics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA;
| | - Yi-Tao Yu
- Center for RNA Biology, Department of Biochemistry and Biophysics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA;
| |
Collapse
|
4
|
Strandgren C, Wiman KG. Therapeutic targeting of TP53 nonsense mutations in cancer. Ups J Med Sci 2024; 129:10719. [PMID: 38863730 PMCID: PMC11165251 DOI: 10.48101/ujms.v129.10719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/13/2024] Open
Abstract
Mutations in the TP53 tumor suppressor gene occur with high prevalence in a wide range of human tumors. A significant fraction of these mutations (around 10%) are nonsense mutations, creating a premature termination codon (PTC) that leads to the expression of truncated inactive p53 protein. Induction of translational readthrough across a PTC in nonsense mutant TP53 allows the production of full-length protein and potentially restoration of normal p53 function. Aminoglycoside antibiotics and a number of novel compounds have been shown to induce full-length p53 in tumor cells carrying various TP53 nonsense mutations. Full-length p53 protein generated by translational readthrough retains the capacity to transactivate p53 target genes and trigger tumor cell death. These findings raise hopes for efficient therapy of TP53 nonsense mutant tumors in the future.
Collapse
Affiliation(s)
| | - Klas G Wiman
- Karolinska Institutet, Departement of Oncology-Pathology, Stockholm, Sweden
| |
Collapse
|
5
|
Wong KM, Wegener E, Baradaran-Heravi A, Huppke B, Gärtner J, Huppke P. Evaluation of Novel Enhancer Compounds in Gentamicin-Mediated Readthrough of Nonsense Mutations in Rett Syndrome. Int J Mol Sci 2023; 24:11665. [PMID: 37511424 PMCID: PMC10380790 DOI: 10.3390/ijms241411665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Rett syndrome (RTT), a severe X-linked neurodevelopmental disorder, is primarily caused by mutations in the methyl CpG binding protein 2 gene (MECP2). Over 35% RTT patients carry nonsense mutation in MECP2, making it a suitable candidate disease for nonsense suppression therapy. In our previous study, gentamicin was found to induce readthrough of MECP2 nonsense mutations with modest efficiency. Given the recent discovery of readthrough enhancers, CDX compounds, we herein evaluated the potentiation effect of CDX5-1, CDX5-288, and CDX6-180 on gentamicin-mediated readthrough efficiency in transfected HeLa cell lines bearing the four most common MECP2 nonsense mutations. We showed that all three CDX compounds potentiated gentamicin-mediated readthrough and increased full-length MeCP2 protein levels in cells expressing the R168X, R255X, R270X, and R294X nonsense mutations. Among all three CDX compounds, CDX5-288 was the most potent enhancer and enabled the use of reduced doses of gentamicin, thus mitigating the toxicity. Furthermore, we successfully demonstrated the upregulation of full-length Mecp2 protein expression in fibroblasts derived from Mecp2R255X/Y mice through combinatorial treatment. Taken together, findings demonstrate the feasibility of this combinatorial approach to nonsense suppression therapy for a subset of RTT patients.
Collapse
Affiliation(s)
- Keit Men Wong
- Department of Neuropediatrics, Jena University Hospital, 07747 Jena, Germany
- Center for Rare Diseases, Jena University Hospital, 07747 Jena, Germany
| | - Eike Wegener
- Department of Pediatrics and Adolescent Medicine, Division of Neuropediatrics, Pediatric Neurology University Medical Center Göttingen, Georg August University Göttingen, 37075 Göttingen, Germany
| | - Alireza Baradaran-Heravi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver BC V6T 1Z3, Canada
| | - Brenda Huppke
- Department of Neuropediatrics, Jena University Hospital, 07747 Jena, Germany
- Center for Rare Diseases, Jena University Hospital, 07747 Jena, Germany
| | - Jutta Gärtner
- Department of Pediatrics and Adolescent Medicine, Division of Neuropediatrics, Pediatric Neurology University Medical Center Göttingen, Georg August University Göttingen, 37075 Göttingen, Germany
| | - Peter Huppke
- Department of Neuropediatrics, Jena University Hospital, 07747 Jena, Germany
- Center for Rare Diseases, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
6
|
Recoding of Nonsense Mutation as a Pharmacological Strategy. Biomedicines 2023; 11:biomedicines11030659. [PMID: 36979640 PMCID: PMC10044939 DOI: 10.3390/biomedicines11030659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Approximately 11% of genetic human diseases are caused by nonsense mutations that introduce a premature termination codon (PTC) into the coding sequence. The PTC results in the production of a potentially harmful shortened polypeptide and activation of a nonsense-mediated decay (NMD) pathway. The NMD pathway reduces the burden of unproductive protein synthesis by lowering the level of PTC mRNA. There is an endogenous rescue mechanism that produces a full-length protein from a PTC mRNA. Nonsense suppression therapies aim to increase readthrough, suppress NMD, or are a combination of both strategies. Therefore, treatment with translational readthrough-inducing drugs (TRIDs) and NMD inhibitors may increase the effectiveness of PTC suppression. Here we discuss the mechanism of PTC readthrough and the development of novel approaches to PTC suppression. We also discuss the toxicity and bioavailability of therapeutics used to stimulate PTC readthrough.
Collapse
|
7
|
Bentur L, Pollak M. Trikafta—Extending Its Success to Less Common Mutations. J Pers Med 2022; 12:jpm12091528. [PMID: 36143317 PMCID: PMC9504046 DOI: 10.3390/jpm12091528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Lea Bentur
- Pediatric Pulmonary Institute, Ruth Children’s Hospital, Rambam Health Care Center, Haifa 31096, Israel
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
- Correspondence: (L.B.); (M.P.)
| | - Mordechai Pollak
- Pediatric Pulmonary Institute, Ruth Children’s Hospital, Rambam Health Care Center, Haifa 31096, Israel
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
- Correspondence: (L.B.); (M.P.)
| |
Collapse
|
8
|
Khalid AM, Hosny KM, Mirjalili S. COVIDOA: a novel evolutionary optimization algorithm based on coronavirus disease replication lifecycle. Neural Comput Appl 2022; 34:22465-22492. [PMID: 36043205 PMCID: PMC9411047 DOI: 10.1007/s00521-022-07639-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 07/18/2022] [Indexed: 11/13/2022]
Abstract
This paper presents a novel bio-inspired optimization algorithm called Coronavirus Optimization Algorithm (COVIDOA). COVIDOA is an evolutionary search strategy that mimics the mechanism of coronavirus when hijacking human cells. COVIDOA is inspired by the frameshifting technique used by the coronavirus for replication. The proposed algorithm is tested using 20 standard benchmark optimization functions with different parameter values. Besides, we utilized five IEEE Congress of Evolutionary Computation (CEC) benchmark test functions (CECC06, 2019 Competition) and five CEC 2011 real-world problems to prove the proposed algorithm's efficiency. The proposed algorithm is compared to eight of the most popular and recent metaheuristic algorithms from the state-of-the-art in terms of best cost, average cost (AVG), corresponding standard deviation (STD), and convergence speed. The results demonstrate that COVIDOA is superior to most existing metaheuristics.
Collapse
|
9
|
Khalid AM, Hamza HM, Mirjalili S, Hosny KM. BCOVIDOA: A Novel Binary Coronavirus Disease Optimization Algorithm for Feature Selection. Knowl Based Syst 2022; 248:108789. [PMID: 35464666 PMCID: PMC9014647 DOI: 10.1016/j.knosys.2022.108789] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 11/20/2022]
Abstract
The increased use of digital tools such as smart phones, Internet of Things devices, cameras, and microphones, has led to the produuction of big data. Large data dimensionality, redundancy, and irrelevance are inherent challenging problems when it comes to big data. Feature selection is a necessary process to select the optimal subset of features when addressing such problems. In this paper, the authors propose a novel Binary Coronavirus Disease Optimization Algorithm (BCOVIDOA) for feature selection, where the Coronavirus Disease Optimization Algorithm (COVIDOA) is a new optimization technique that mimics the replication mechanism used by Coronavirus when hijacking human cells. The performance of the proposed algorithm is evaluated using twenty-six standard benchmark datasets from UCI Repository. The results are compared with nine recent wrapper feature selection algorithms. The experimental results demonstrate that the proposed BCOVIDOA significantly outperforms the existing algorithms in terms of accuracy, best cost, the average cost (AVG), standard deviation (STD), and size of selected features. Additionally, the Wilcoxon rank-sum test is calculated to prove the statistical significance of the results.
Collapse
Affiliation(s)
- Asmaa M Khalid
- Department of Information Technology, Faculty of Computers and Informatics, Zagazig University, Zagazig 44519, Egypt
| | - Hanaa M Hamza
- Department of Information Technology, Faculty of Computers and Informatics, Zagazig University, Zagazig 44519, Egypt
| | - Seyedali Mirjalili
- Centre for Artificial Intelligence Research and Optimization, Torrens University Australia, Fortitude Valley, Brisbane 4006, QLD, Australia
- Yonsei Frontier Lab, Yonsei University, Seoul, South Korea
| | - Khalid M Hosny
- Department of Information Technology, Faculty of Computers and Informatics, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
10
|
Ensinck MM, Carlon MS. One Size Does Not Fit All: The Past, Present and Future of Cystic Fibrosis Causal Therapies. Cells 2022; 11:cells11121868. [PMID: 35740997 PMCID: PMC9220995 DOI: 10.3390/cells11121868] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 02/04/2023] Open
Abstract
Cystic fibrosis (CF) is the most common monogenic disorder, caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Over the last 30 years, tremendous progress has been made in understanding the molecular basis of CF and the development of treatments that target the underlying defects in CF. Currently, a highly effective CFTR modulator treatment (Kalydeco™/Trikafta™) is available for 90% of people with CF. In this review, we will give an extensive overview of past and ongoing efforts in the development of therapies targeting the molecular defects in CF. We will discuss strategies targeting the CFTR protein (i.e., CFTR modulators such as correctors and potentiators), its cellular environment (i.e., proteostasis modulation, stabilization at the plasma membrane), the CFTR mRNA (i.e., amplifiers, nonsense mediated mRNA decay suppressors, translational readthrough inducing drugs) or the CFTR gene (gene therapies). Finally, we will focus on how these efforts can be applied to the 15% of people with CF for whom no causal therapy is available yet.
Collapse
Affiliation(s)
- Marjolein M. Ensinck
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Flanders, Belgium;
| | - Marianne S. Carlon
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Flanders, Belgium;
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Flanders, Belgium
- Correspondence:
| |
Collapse
|
11
|
Rouf MA, Wen L, Mahendra Y, Wang J, Zhang K, Liang S, Wang Y, Li Z, Wang Y, Wang G. The recent advances and future perspectives of genetic compensation studies in the zebrafish model. Genes Dis 2022; 10:468-479. [DOI: 10.1016/j.gendis.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 10/19/2022] Open
|
12
|
McKay IR, Ooi CY. The Exocrine Pancreas in Cystic Fibrosis in the Era of CFTR Modulation: A Mini Review. Front Pediatr 2022; 10:914790. [PMID: 35832587 PMCID: PMC9271761 DOI: 10.3389/fped.2022.914790] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) is a common disorder of autosomal recessive inheritance, that once conferred a life expectancy of only a few months. Over recent years, significant advances have been made to CF therapeutic approaches, changing the face of the disease, and facilitating the partial restoration of pancreatic function. This mini review summarizes the current landscape of exocrine pancreatic management in CF and explores areas for future direction and development.
Collapse
Affiliation(s)
| | - Chee Y Ooi
- School of Clinical Medicine, Discipline of Paediatrics and Child Health, Randwick Clinical Campus, University of New South Wales (UNSW) Medicine and Health, University of New South Wales, Sydney, NSW, Australia.,Department of Gastroenterology, Sydney Children's Hospital Randwick, Randwick, NSW, Australia
| |
Collapse
|
13
|
Bongiorno R, Colombo MP, Lecis D. Deciphering the nonsense-mediated mRNA decay pathway to identify cancer cell vulnerabilities for effective cancer therapy. J Exp Clin Cancer Res 2021; 40:376. [PMID: 34852841 PMCID: PMC8638473 DOI: 10.1186/s13046-021-02192-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
Nonsense-mediated mRNA decay (NMD) is a highly conserved cellular surveillance mechanism, commonly studied for its role in mRNA quality control because of its capacity of degrading mutated mRNAs that would produce truncated proteins. However, recent studies have proven that NMD hides more complex tasks involved in a plethora of cellular activities. Indeed, it can control the stability of mutated as well as non-mutated transcripts, tuning transcriptome regulation. NMD not only displays a pivotal role in cell physiology but also in a number of genetic diseases. In cancer, the activity of this pathway is extremely complex and it is endowed with both pro-tumor and tumor suppressor functions, likely depending on the genetic context and tumor microenvironment. NMD inhibition has been tested in pre-clinical studies showing favored production of neoantigens by cancer cells, which can stimulate the triggering of an anti-tumor immune response. At the same time, NMD inhibition could result in a pro-tumor effect, increasing cancer cell adaptation to stress. Since several NMD inhibitors are already available in the clinic to treat genetic diseases, these compounds could be redirected to treat cancer patients, pending the comprehension of these variegated NMD regulation mechanisms. Ideally, an effective strategy should exploit the anti-tumor advantages of NMD inhibition and simultaneously preserve its intrinsic tumor suppressor functions. The targeting of NMD could provide a new therapeutic opportunity, increasing the immunogenicity of tumors and potentially boosting the efficacy of the immunotherapy agents now available for cancer treatment.
Collapse
Affiliation(s)
- Roberta Bongiorno
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Mario Paolo Colombo
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Daniele Lecis
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
14
|
Reeves SR. Primary nasal epithelial cells from patients with cystic fibrosis hold promise for guiding precision medicine and expanding treatment. Eur Respir J 2021; 58:2102735. [PMID: 34857588 DOI: 10.1183/13993003.02735-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Stephen R Reeves
- Dept of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| |
Collapse
|
15
|
Bayat S, Lysak MA, Mandáková T. Genome structure and evolution in the cruciferous tribe Thlaspideae (Brassicaceae). THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 108:1768-1785. [PMID: 34661331 DOI: 10.1111/tpj.15542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
Whole-genome duplications (WGDs) and chromosome rearrangements (CRs) play the key role in driving the diversification and evolution of plant lineages. Although the direct link between WGDs and plant diversification is well documented, relatively few studies focus on the evolutionary significance of CRs. The cruciferous tribe Thlaspideae represents an ideal model system to address the role of large-scale chromosome alterations in genome evolution, as most Thlaspideae species share the same diploid chromosome number (2n = 2x = 14). Here we constructed the genome structure in 12 Thlaspideae species, including field pennycress (Thlaspi arvense) and garlic mustard (Alliaria petiolata). We detected and precisely characterized genus- and species-specific CRs, mostly pericentric inversions, as the main genome-diversifying drivers in the tribe. We reconstructed the structure of seven chromosomes of an ancestral Thlaspideae genome, identified evolutionary stable chromosomes versus chromosomes prone to CRs, estimated the rate of CRs, and uncovered an allohexaploid origin of garlic mustard from diploid taxa closely related to A. petiolata and Parlatoria cakiloidea. Furthermore, we performed detailed bioinformatic analysis of the Thlaspideae repeatomes, and identified repetitive elements applicable as unique species- and genus-specific barcodes and chromosome landmarks. This study deepens our general understanding of the evolutionary role of CRs, particularly pericentric inversions, in plant genome diversification, and provides a robust base for follow-up whole-genome sequencing efforts.
Collapse
Affiliation(s)
- Soheila Bayat
- CEITEC, Masaryk University, Brno, 62500, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Martin A Lysak
- CEITEC, Masaryk University, Brno, 62500, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Terezie Mandáková
- CEITEC, Masaryk University, Brno, 62500, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| |
Collapse
|
16
|
Integrity and Stability of PTC Bearing CFTR mRNA and Relevance to Future Modulator Therapies in Cystic Fibrosis. Genes (Basel) 2021; 12:genes12111810. [PMID: 34828417 PMCID: PMC8621375 DOI: 10.3390/genes12111810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/26/2022] Open
Abstract
Major advances have recently been made in the development and application of CFTR (cystic fibrosis transmembrane conductance regulator) mutation class-specific modulator therapies, but to date, there are no approved modulators for Class I mutations, i.e., those introducing a premature termination codon (PTC) into the CFTR mRNA. Such mutations induce nonsense-mediated decay (NMD), a cellular quality control mechanism that reduces the quantity of PTC bearing mRNAs, presumably to avoid translation of potentially deleterious truncated CFTR proteins. The NMD-mediated reduction of PTC-CFTR mRNA molecules reduces the efficacy of one of the most promising approaches to treatment of such mutations, namely, PTC readthrough therapy, using molecules that induce the incorporation of near-cognate amino acids at the PTC codon, thereby enabling translation of a full-length protein. In this study, we measure the effect of three different PTC mutations on the abundance, integrity, and stability of respective CFTR mRNAs, using CFTR specific RT-qPCR-based assays. Altogether, our data suggest that optimized rescue of PTC mutations has to take into account (1) the different steady-state levels of the CFTR mRNA associated with each specific PTC mutation; (2) differences in abundance between the 3' and 5' regions of CFTR mRNA, even following PTC readthrough or NMD inhibition; and (3) variable effects on CFTR mRNA stability for each specific PTC mutation.
Collapse
|
17
|
Therapeutic pipeline for individuals with cystic fibrosis with mutations nonresponsive to current cystic fibrosis transmembrane conductance regulator modulators. Curr Opin Pulm Med 2021; 27:567-574. [PMID: 34494979 DOI: 10.1097/mcp.0000000000000827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Cystic fibrosis is a severe autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator gene (CFTR) encoding the CFTR protein, a chloride channel expressed in many epithelial cells. New drugs called CFTR modulators aim at restoring the CFTR protein function and they will benefit most of the patients with cystic fibrosis in the near future. However, more than 10% of CFTR mutations do not produce any CFTR protein for CFTR modulators to act upon, and the purpose of this review is to provide an overview of different approaches pursued to treat patients bearing mutations nonresponsive to CFTR modulators. RECENT FINDINGS These different approaches constitute readthrough agents for nonsense mutations, nucleic acid-based therapies, RNA-based or DNA-based, and cell-based therapies. Some approaches using mRNA or cDNA combined with a delivery vehicle are mutation-agnostic therapies. Other approaches, such as the use of tRNA, antisense oligonucleotides, gene editing or cell-based therapies are mutation-specific therapies. SUMMARY Most of these approaches are in preclinical development or for some of them, early clinical phases. Many hurdles and challenges will have to be solved before they can be safely translated to patients.
Collapse
|
18
|
Therapeutic Approaches for Patients with Cystic Fibrosis Not Eligible for Current CFTR Modulators. Cells 2021; 10:cells10102793. [PMID: 34685773 PMCID: PMC8534516 DOI: 10.3390/cells10102793] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/26/2022] Open
Abstract
Cystic fibrosis is a severe autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene encoding the CFTR protein, a chloride channel expressed in many epithelial cells. New drugs called CFTR modulators aim at restoring the CFTR protein function, and they will benefit many patients with cystic fibrosis in the near future. However, some patients bear rare mutations that are not yet eligible for CFTR modulators, although they might be amenable to these new disease-modifying drugs. Moreover, more than 10% of CFTR mutations do not produce any CFTR protein for CFTR modulators to act upon. The purpose of this review is to provide an overview of different approaches pursued to treat patients bearing mutations ineligible for CFTR modulators. One approach is to broaden the numbers of mutations eligible for CFTR modulators. This requires developing strategies to evaluate drugs in populations bearing very rare genotypes. Other approaches aiming at correcting the CFTR defect develop new mutation-specific or mutation-agnostic therapies for mutations that do not produce a CFTR protein: readthrough agents for nonsense mutations, nucleic acid-based therapies, RNA- or DNA-based, and cell-based therapies. Most of these approaches are in pre-clinical development or, for some of them, early clinical phases. Many hurdles and challenges will have to be solved before they can be safely translated to patients.
Collapse
|
19
|
Meoli A, Fainardi V, Deolmi M, Chiopris G, Marinelli F, Caminiti C, Esposito S, Pisi G. State of the Art on Approved Cystic Fibrosis Transmembrane Conductance Regulator ( CFTR) Modulators and Triple-Combination Therapy. Pharmaceuticals (Basel) 2021; 14:ph14090928. [PMID: 34577628 PMCID: PMC8471029 DOI: 10.3390/ph14090928] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/23/2022] Open
Abstract
Cystic fibrosis (CF) is the most common life-limiting inherited disease in Caucasian populations, affecting approximately 80,000 people worldwide. CF is a complex multi-organ monogenic autosomal recessive disorder caused by a mutation in cystic fibrosis transmembrane conductance regulator (CFTR) gene. Since the discovery of the CFTR gene in 1989, more than 2000 mutations have been identified so far and about 240 can cause CF. Until recently, the treatment for CF was aimed to prevent and manage the manifestations of CFTR dysfunction, primarily recurrent pulmonary infections and pancreatic exocrine failure. Over the past few decades, the therapeutic approach to CF has been revolutionized by the development of a new class of small molecules called CFTR modulators that target specific defects caused by mutations in the CFTR gene. CFTR modulators have been shown to change profoundly the clinical course of the CF, leading to meaningful improvements in the lives of a large proportion of people of CF heterozygous for F508del, especially if started in young children. Further studies are needed to extend the use of triple CFTR modulation therapy also for young children in order to prevent the irreversible effects of the disease and for patients with very rare mutations with a personalized approach to treatment.
Collapse
Affiliation(s)
- Aniello Meoli
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Valentina Fainardi
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Michela Deolmi
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Giulia Chiopris
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Francesca Marinelli
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| | - Caterina Caminiti
- Research and Innovation Unit, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy;
| | - Susanna Esposito
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
- Correspondence:
| | - Giovanna Pisi
- Paediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy; (A.M.); (V.F.); (M.D.); (G.C.); (F.M.); (G.P.)
| |
Collapse
|
20
|
Cioni P, Gabellieri E, Campanini B, Bettati S, Raboni S. Use of Exogenous Enzymes in Human Therapy: Approved Drugs and Potential Applications. Curr Med Chem 2021; 29:411-452. [PMID: 34259137 DOI: 10.2174/0929867328666210713094722] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
The development of safe and efficacious enzyme-based human therapies has increased greatly in the last decades, thanks to remarkable advances in the understanding of the molecular mechanisms responsible for different diseases, and the characterization of the catalytic activity of relevant exogenous enzymes that may play a remedial effect in the treatment of such pathologies. Several enzyme-based biotherapeutics have been approved by FDA (the U.S. Food and Drug Administration) and EMA (the European Medicines Agency) and many are undergoing clinical trials. Apart from enzyme replacement therapy in human genetic diseases, which is not discussed in this review, approved enzymes for human therapy find applications in several fields, from cancer therapy to thrombolysis and the treatment, e.g., of clotting disorders, cystic fibrosis, lactose intolerance and collagen-based disorders. The majority of therapeutic enzymes are of microbial origin, the most convenient source due to fast, simple and cost-effective production and manipulation. The use of microbial recombinant enzymes has broadened prospects for human therapy but some hurdles such as high immunogenicity, protein instability, short half-life and low substrate affinity, still need to be tackled. Alternative sources of enzymes, with reduced side effects and improved activity, as well as genetic modification of the enzymes and novel delivery systems are constantly searched. Chemical modification strategies, targeted- and/or nanocarrier-mediated delivery, directed evolution and site-specific mutagenesis, fusion proteins generated by genetic manipulation are the most explored tools to reduce toxicity and improve bioavailability and cellular targeting. This review provides a description of exogenous enzymes that are presently employed for the therapeutic management of human diseases with their current FDA/EMA-approved status, along with those already experimented at the clinical level and potential promising candidates.
Collapse
Affiliation(s)
- Patrizia Cioni
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| | - Edi Gabellieri
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124 Parma. Italy
| | - Stefano Bettati
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| | - Samanta Raboni
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| |
Collapse
|
21
|
Dabrowski M, Bukowy-Bieryllo Z, Jackson CL, Zietkiewicz E. Properties of Non-Aminoglycoside Compounds Used to Stimulate Translational Readthrough of PTC Mutations in Primary Ciliary Dyskinesia. Int J Mol Sci 2021; 22:ijms22094923. [PMID: 34066907 PMCID: PMC8125088 DOI: 10.3390/ijms22094923] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 12/30/2022] Open
Abstract
Primary ciliary dyskinesia (PCD) is a rare disease with autosomal recessive inheritance, caused mostly by bi-allelic gene mutations that impair motile cilia structure and function. Currently, there are no causal treatments for PCD. In many disease models, translational readthrough of premature termination codons (PTC-readthrough) induced by aminoglycosides has been proposed as an effective way of restoring functional protein expression and reducing disease symptoms. However, variable outcomes of pre-clinical trials and toxicity associated with long-term use of aminoglycosides prompt the search for other compounds that might overcome these problems. Because a high proportion of PCD-causing variants are nonsense mutations, readthrough therapies are an attractive option. We tested a group of chemical compounds with known PTC-readthrough potential (ataluren, azithromycin, tylosin, amlexanox, and the experimental compound TC007), collectively referred to as non-aminoglycosides (NAGs). We investigated their PTC-readthrough efficiency in six PTC mutations found in Polish PCD patients, in the context of cell and cilia health, and in comparison to the previously tested aminoglycosides. The NAGs did not compromise the viability of the primary nasal respiratory epithelial cells, and the ciliary beat frequency was retained, similar to what was observed for gentamicin. In HEK293 cells transfected with six PTC-containing inserts, the tested compounds stimulated PTC-readthrough but with lower efficiency than aminoglycosides. The study allowed us to select compounds with minimal negative impact on cell viability and function but still the potential to induce PTC-readthrough.
Collapse
Affiliation(s)
- Maciej Dabrowski
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland; (M.D.); (Z.B.-B.)
| | - Zuzanna Bukowy-Bieryllo
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland; (M.D.); (Z.B.-B.)
| | - Claire L. Jackson
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Ewa Zietkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland; (M.D.); (Z.B.-B.)
- Correspondence:
| |
Collapse
|
22
|
Girón Moreno RM, García-Clemente M, Diab-Cáceres L, Martínez-Vergara A, Martínez-García MÁ, Gómez-Punter RM. Treatment of Pulmonary Disease of Cystic Fibrosis: A Comprehensive Review. Antibiotics (Basel) 2021; 10:486. [PMID: 33922413 PMCID: PMC8144952 DOI: 10.3390/antibiotics10050486] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease that causes absence or dysfunction of a protein named transmembrane conductance regulatory protein (CFTR) that works as an anion channel. As a result, the secretions of the organs where CFTR is expressed are very viscous, so their functionality is altered. The main cause of morbidity is due to the involvement of the respiratory system as a result of recurrent respiratory infections by different pathogens. In recent decades, survival has been increasing, rising by around age 50. This is due to the monitoring of patients in multidisciplinary units, early diagnosis with neonatal screening, and advances in treatments. In this chapter, we will approach the different therapies used in CF for the treatment of symptoms, obstruction, inflammation, and infection. Moreover, we will discuss specific and personalized treatments to correct the defective gene and repair the altered protein CFTR. The obstacle for personalized CF treatment is to predict the drug response of patients due to genetic complexity and heterogeneity of uncommon mutations.
Collapse
Affiliation(s)
- Rosa María Girón Moreno
- Servicio de Neumología, Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain; (R.M.G.M.); (R.M.G.-P.)
| | - Marta García-Clemente
- Servicio de Neumología, Hospital Universitario Central de Asturias, C/Avenida de Roma S/n, 33011 Oviedo, Spain
| | - Layla Diab-Cáceres
- Servicio de Neumología, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | | | | | - Rosa Mar Gómez-Punter
- Servicio de Neumología, Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain; (R.M.G.M.); (R.M.G.-P.)
| |
Collapse
|
23
|
Bandiera T, Galietta LJV. Pharmacological approaches to cystic fibrosis. Eur J Med Chem 2021; 216:113240. [PMID: 33691259 DOI: 10.1016/j.ejmech.2021.113240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Tiziano Bandiera
- D3 PharmaChemistry Line, Istituto Italiano di Tecnologia, via Morego 30, 16163, Genoa, Italy.
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Campi Flegrei 34, 80078, Pozzuoli, NA, Italy; Department of Translational Medical Sciences (DISMET), University of Naples, "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy
| |
Collapse
|
24
|
Sadeh TT, Black GC, Manson F. A Review of Genetic and Physiological Disease Mechanisms Associated With Cav1 Channels: Implications for Incomplete Congenital Stationary Night Blindness Treatment. Front Genet 2021; 12:637780. [PMID: 33584831 PMCID: PMC7876387 DOI: 10.3389/fgene.2021.637780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/12/2021] [Indexed: 11/13/2022] Open
Abstract
Calcium channels are crucial to a number of cellular functions. The high voltage-gated calcium channel family comprise four heteromeric channels (Cav1.1-1.4) that function in a similar manner, but that have distinct expression profiles. Three of the pore-forming α1 subunits are located on autosomes and the forth on the X chromosome, which has consequences for the type of pathogenic mutation and the disease mechanism associated with each gene. Mutations in this family of channels are associated with malignant hyperthermia (Cav1.1), various QT syndromes (Cav1.2), deafness (Cav1.3), and incomplete congenital stationary night blindness (iCSNB; Cav1.4). In this study we performed a bioinformatic analysis on reported mutations in all four Cav α1 subunits and correlated these with variant frequency in the general population, phenotype and the effect on channel conductance to produce a comprehensive composite Cav1 mutation analysis. We describe regions of mutation clustering, identify conserved residues that are mutated in multiple family members and regions likely to cause a loss- or gain-of-function in Cav1.4. Our research highlights that therapeutic treatments for each of the Cav1 channels will have to consider channel-specific mechanisms, especially for the treatment of X-linked iCSNB.
Collapse
Affiliation(s)
- Tal T Sadeh
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Graeme C Black
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, St Mary's Hospital, Manchester, United Kingdom
| | - Forbes Manson
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
25
|
Sharma J, Abbott J, Klaskala L, Zhao G, Birket SE, Rowe SM. A Novel G542X CFTR Rat Model of Cystic Fibrosis Is Sensitive to Nonsense Mediated Decay. Front Physiol 2020; 11:611294. [PMID: 33391025 PMCID: PMC7772197 DOI: 10.3389/fphys.2020.611294] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
Nonsense mutations that lead to the insertion of a premature termination codon (PTC) in the cystic fibrosis transmembrane conductance regulator (CFTR) transcript affect 11% of patients with cystic fibrosis (CF) worldwide and are associated with severe disease phenotype. While CF rat models have contributed significantly to our understanding of CF disease pathogenesis, there are currently no rat models available for studying CF nonsense mutations. Here we created and characterized the first homozygous CF rat model that bears the CFTR G542X nonsense mutation in the endogenous locus using CRISPR/Cas9 gene editing. In addition to displaying severe CF manifestations and developmental defects such as reduced growth, abnormal tooth enamel, and intestinal obstruction, CFTR G542X knockin rats demonstrated an absence of CFTR function in tracheal and intestinal sections as assessed by nasal potential difference and transepithelial short-circuit current measurements. Reduced CFTR mRNA levels in the model further suggested sensitivity to nonsense-mediated decay, a pathway elicited by the presence of PTCs that degrades the PTC-bearing transcripts and thus further diminishes the level of CFTR protein. Although functional restoration of CFTR was observed in G542X rat tracheal epithelial cells in response to single readthrough agent therapy, therapeutic efficacy was not observed in G542X knockin rats in vivo. The G542X rat model provides an invaluable tool for the identification and in vivo validation of potential therapies for CFTR nonsense mutations.
Collapse
Affiliation(s)
- Jyoti Sharma
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joseph Abbott
- Horizon Discovery Group, PLC, St. Louis, MO, United States
| | | | - Guojun Zhao
- Horizon Discovery Group, PLC, St. Louis, MO, United States
| | - Susan E. Birket
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Steven M. Rowe
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
Molecular Insights into Determinants of Translational Readthrough and Implications for Nonsense Suppression Approaches. Int J Mol Sci 2020; 21:ijms21249449. [PMID: 33322589 PMCID: PMC7764779 DOI: 10.3390/ijms21249449] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/27/2020] [Accepted: 12/05/2020] [Indexed: 02/07/2023] Open
Abstract
The fidelity of protein synthesis, a process shaped by several mechanisms involving specialized ribosome regions and external factors, ensures the precise reading of sense and stop codons. However, premature termination codons (PTCs) arising from mutations may, at low frequency, be misrecognized and result in PTC suppression, named ribosome readthrough, with production of full-length proteins through the insertion of a subset of amino acids. Since some drugs have been identified as readthrough inducers, this fidelity drawback has been explored as a therapeutic approach in several models of human diseases caused by nonsense mutations. Here, we focus on the mechanisms driving translation in normal and aberrant conditions, the potential fates of mRNA in the presence of a PTC, as well as on the results obtained in the research of efficient readthrough-inducing compounds. In particular, we describe the molecular determinants shaping the outcome of readthrough, namely the nucleotide and protein context, with the latter being pivotal to produce functional full-length proteins. Through the interpretation of experimental and mechanistic findings, mainly obtained in lysosomal and coagulation disorders, we also propose a scenario of potential readthrough-favorable features to achieve relevant rescue profiles, representing the main issue for the potential translatability of readthrough as a therapeutic strategy.
Collapse
|
27
|
Palma M, Lejeune F. Deciphering the molecular mechanism of stop codon readthrough. Biol Rev Camb Philos Soc 2020; 96:310-329. [PMID: 33089614 DOI: 10.1111/brv.12657] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/23/2020] [Accepted: 09/26/2020] [Indexed: 12/11/2022]
Abstract
Recognition of the stop codon by the translation machinery is essential to terminating translation at the right position and to synthesizing a protein of the correct size. Under certain conditions, the stop codon can be recognized as a coding codon promoting translation, which then terminates at a later stop codon. This event, called stop codon readthrough, occurs either by error, due to a dedicated regulatory environment leading to generation of different protein isoforms, or through the action of a readthrough compound. This review focuses on the mechanisms of stop codon readthrough, the nucleotide and protein environments that facilitate or inhibit it, and the therapeutic interest of stop codon readthrough in the treatment of genetic diseases caused by nonsense mutations.
Collapse
Affiliation(s)
- Martine Palma
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Fabrice Lejeune
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| |
Collapse
|