Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.377
Filter
1.
Life Sci ; 348: 122685, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38710276

ABSTRACT

Gene therapy in pediatrics represents a cutting-edge therapeutic strategy for treating a range of genetic disorders that manifest in childhood. Gene therapy involves the modification or correction of a mutated gene or the introduction of a functional gene into a patient's cells. In general, it is implemented through two main modalities namely ex vivo gene therapy and in vivo gene therapy. Currently, a noteworthy array of gene therapy products has received valid market authorization, with several others in various stages of the approval process. Additionally, a multitude of clinical trials are actively underway, underscoring the dynamic progress within this field. Pediatric genetic disorders in the fields of hematology, oncology, vision and hearing loss, immunodeficiencies, neurological, and metabolic disorders are areas for gene therapy interventions. This review provides a comprehensive overview of the evolution and current progress of gene therapy-based treatments in the clinic for pediatric patients. It navigates the historical milestones of gene therapies, currently approved gene therapy products by the U.S. Food and Drug Administration (FDA) and/or European Medicines Agency (EMA) for children, and the promising future for genetic disorders. By providing a thorough compilation of approved gene therapy drugs and published results of completed or ongoing clinical trials, this review serves as a guide for pediatric clinicians to get a quick overview of the situation of clinical studies and approved gene therapy products as of 2023.


Subject(s)
Drug Approval , Genetic Therapy , Pediatrics , Humans , Genetic Therapy/methods , Child , Pediatrics/methods , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/drug therapy , Clinical Trials as Topic
2.
BMJ Open ; 14(5): e085237, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38760043

ABSTRACT

INTRODUCTION: Around 2000 children are born in the UK per year with a neurodevelopmental genetic syndrome with significantly increased morbidity and mortality. Often little is known about expected growth and phenotypes in these children. Parents have responded by setting up social media groups to generate data themselves. Given the significant clinical evidence gaps, this research will attempt to identify growth patterns, developmental profiles and phenotypes, providing data on long-term medical and educational outcomes. This will guide clinicians when to investigate, monitor or treat symptoms and when to search for additional or alternative diagnoses. METHODS AND ANALYSIS: This is an observational, multicentre cohort study recruiting between March 2023 and February 2026. Children aged 6 months up to 16 years with a pathogenic or likely pathogenic variant in a specified gene will be eligible. Children will be identified through the National Health Service and via self-recruitment. Parents or carers will complete a questionnaire at baseline and again 1 year after recruitment. The named clinician (in most cases a clinical geneticist) will complete a clinical proforma which will provide data from their most recent clinical assessment. Qualitative interviews will be undertaken with a subset of parents partway through the study. Growth and developmental milestone curves will be generated through the DECIPHER website (https://deciphergenomics.org) where 5 or more children have the same genetic syndrome (at least 10 groups expected). ETHICS AND DISSEMINATION: The results will be presented at national and international conferences concerning the care of children with genetic syndromes. Results will also be submitted for peer review and publication.


Subject(s)
Rare Diseases , Humans , Rare Diseases/genetics , Rare Diseases/therapy , Child , Child, Preschool , United Kingdom , Infant , Adolescent , Research Design , Female , Male , Observational Studies as Topic , Neurodevelopmental Disorders/genetics , Cohort Studies , Multicenter Studies as Topic , Genetic Diseases, Inborn/therapy , Quality Improvement , Parents
4.
J Control Release ; 369: 696-721, 2024 May.
Article in English | MEDLINE | ID: mdl-38580137

ABSTRACT

Rare genetic diseases, often referred to as orphan diseases due to their low prevalence and limited treatment options, have long posed significant challenges to our medical system. In recent years, Messenger RNA (mRNA) therapy has emerged as a highly promising treatment approach for various diseases caused by genetic mutations. Chemically modified mRNA is introduced into cells using carriers like lipid-based nanoparticles (LNPs), producing functional proteins that compensate for genetic deficiencies. Given the advantages of precise dosing, biocompatibility, transient expression, and minimal risk of genomic integration, mRNA therapies can safely and effectively correct genetic defects in rare diseases and improve symptoms. Currently, dozens of mRNA drugs targeting rare diseases are undergoing clinical trials. This comprehensive review summarizes the progress of mRNA therapy in treating rare genetic diseases. It introduces the development, molecular design, and delivery systems of mRNA therapy, highlighting their research progress in rare genetic diseases based on protein replacement and gene editing. The review also summarizes research progress in various rare disease models and clinical trials. Additionally, it discusses the challenges and future prospects of mRNA therapy. Researchers are encouraged to join this field and collaborate to advance the clinical translation of mRNA therapy, bringing hope to patients with rare genetic diseases.


Subject(s)
Genetic Therapy , RNA, Messenger , Rare Diseases , Humans , Rare Diseases/therapy , Rare Diseases/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Animals , Genetic Therapy/methods , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/genetics , Nanoparticles , Gene Editing/methods
5.
Mol Genet Metab ; 142(1): 108360, 2024 May.
Article in English | MEDLINE | ID: mdl-38428378

ABSTRACT

The Mendelian disorders of chromatin machinery (MDCMs) represent a distinct subgroup of disorders that present with neurodevelopmental disability. The chromatin machinery regulates gene expression by a range of mechanisms, including by post-translational modification of histones, responding to histone marks, and remodelling nucleosomes. Some of the MDCMs that impact on histone modification may have potential therapeutic interventions. Two potential treatment strategies are to enhance the intracellular pool of metabolites that can act as substrates for histone modifiers and the use of medications that may inhibit or promote the modification of histone residues to influence gene expression. In this article we discuss the influence and potential treatments of histone modifications involving histone acetylation and histone methylation. Genomic technologies are facilitating earlier diagnosis of many Mendelian disorders, providing potential opportunities for early treatment from infancy. This has parallels with how inborn errors of metabolism have been afforded early treatment with newborn screening. Before this promise can be fulfilled, we require greater understanding of the biochemical fingerprint of these conditions, which may provide opportunities to supplement metabolites that can act as substrates for chromatin modifying enzymes. Importantly, understanding the metabolomic profile of affected individuals may also provide disorder-specific biomarkers that will be critical for demonstrating efficacy of treatment, as treatment response may not be able to be accurately assessed by clinical measures.


Subject(s)
Chromatin , Metabolic Networks and Pathways , Humans , Chromatin/genetics , Chromatin/metabolism , Metabolic Networks and Pathways/genetics , Histones/metabolism , Histones/genetics , Protein Processing, Post-Translational , Acetylation , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/therapy , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/metabolism , Chromatin Assembly and Disassembly/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/metabolism , Infant, Newborn , Methylation
6.
Med Law Rev ; 32(2): 178-204, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38513296

ABSTRACT

Heritable human genome editing (HHGE) to correct a nuclear gene sequence that would result in a serious genetic condition in a future child is presented as 'treatment' in various ethics and policy materials, and as morally preferable to the 'selection' practice of preimplantation genetic testing (PGT), which is subject to the disability critique. However, whether HHGE is 'treatment' for a future child, or another form of 'selection', or whether HHGE instead 'treats' prospective parents, are now central questions in the debate regarding its possible legalisation. This article argues that the idea of 'treatment' for a future child is largely a proxy for 'seriousness of purpose', intended to distinguish HHGE to avoid serious genetic conditions from less obviously justifiable uses; that HHGE is best understood, and morally justified, as a form of 'treatment' for prospective parents who strongly desire an unaffected genetically related child and who have no, or poor, options to achieve this; that HHGE would be morally permissible if consistent with that child's welfare; that legalisation is supportable with reference to the right to respect for private and family life under Article 8 of the European Convention on Human Rights; and that HHGE is morally distinguishable from PGT.


Subject(s)
Gene Editing , Preimplantation Diagnosis , Humans , Gene Editing/ethics , Gene Editing/legislation & jurisprudence , Preimplantation Diagnosis/ethics , Genome, Human , Genetic Testing/legislation & jurisprudence , Genetic Testing/ethics , Genetic Therapy/ethics , Genetic Therapy/legislation & jurisprudence , Genetic Diseases, Inborn/therapy
7.
Mutagenesis ; 39(3): 157-171, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38332115

ABSTRACT

The therapeutic potential of the human genome has been explored through the development of next-generation therapeutics, which have had a high impact on treating genetic disorders. Classical treatments have traditionally focused on common diseases that require repeated treatments. However, with the recent advancements in the development of nucleic acids, utilizing DNA and RNA to modify or correct gene expression in genetic disorders, there has been a paradigm shift in the treatment of rare diseases, offering more potential one-time cure options. Advanced technologies that use CRISPR-Cas 9, antisense oligonucleotides, siRNA, miRNA, and aptamers are promising tools that have achieved successful breakthroughs in the treatment of various genetic disorders. The advancement in the chemistry of these molecules has improved their efficacy, reduced toxicity, and expanded their clinical use across a wide range of tissues in various categories of human disorders. However, challenges persist regarding the safety and efficacy of these advanced technologies in translating into clinical practice. This review mainly focuses on the potential therapies for rare genetic diseases and considers how next-generation techniques enable drug development to achieve long-lasting curative effects through gene inhibition, replacement, and editing.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Genetic Diseases, Inborn , Genetic Therapy , Rare Diseases , Humans , Rare Diseases/genetics , Rare Diseases/therapy , Gene Editing/methods , Genetic Therapy/methods , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/genetics , Oligonucleotides, Antisense/therapeutic use , RNA, Small Interfering/therapeutic use , RNA, Small Interfering/genetics , MicroRNAs/genetics , Aptamers, Nucleotide/therapeutic use
8.
Rev. med. cine ; 19(4): 303-316, 11/14/2023. ilus
Article in Spanish | IBECS | ID: ibc-227599

ABSTRACT

Si bien la hemofilia era conocida como una enfermedad que perturbaba solo a las familias reales de Europa, actualmente afecta a uno de cada 5.000 y a uno de cada 30.000 varones recién nacidos vivos tanto para la de Tipo A como B, respectivamente. La hemofilia es un trastorno de la coagulación sanguínea que afecta principalmente a varones por su carácter de herencia recesiva ligada al X, siendo su manifestación principal las hemorragias que pueden llegar a ser mortales si no son tratadas correctamente. En Bombardier Blood (2020) de Patrick James Lynch, un documental enfocado al recorrido de Chris Bombardier, una persona hemofílica cuyo sueño es escalar las siete cumbres más altas del mundo, incluyendo en su paso el monte Everest en el Himalaya. El documental además de destacar los aspectos médicos sobre esta enfermedad hace ver los diferentes estilos de vida de acuerdo a su localización, las normativas y sistemas de salud que los rige y como esto incide en el diagnóstico, seguimiento y tratamiento. (AU)


While hemophilia was considered a disease that disturbed only the royal families of Europe, it currently affects one in 5.000 and one in 30.000 live newborn males for both Type A and B, respectively. Hemophilia is a blood clotting disorder that mainly affects men due to its character of recessive inheritance linked to X, its main manifestation hemorrhaging that can become fatal if they are not treated correctly. Bombardier Blood (2020) by Patrick James Lynch is a documentary focused on the journey of Chris Bombardier, a hemophilic person whose dream is to climb the seven highest summits in the world, including Mount Everest in the Himalayas. The documentary, in addition to highlighting the medical aspects of this disease, shows the different lifestyles according to their location, the regulations and health systems that govern them, and how these affect diagnosis, monitoring, and treatment. (AU)


Subject(s)
Humans , Hemophilia A/diagnosis , Hemophilia A/therapy , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/psychology , Genetic Diseases, Inborn/therapy , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/therapy , Heredity , Nepal
9.
Int J Mol Sci ; 24(7)2023 Mar 23.
Article in English | MEDLINE | ID: mdl-37047074

ABSTRACT

Nonsense mutations trigger premature translation termination and often give rise to prevalent and rare genetic diseases. Consequently, the pharmacological suppression of an unscheduled stop codon represents an attractive treatment option and is of high clinical relevance. At the molecular level, the ability of the ribosome to continue translation past a stop codon is designated stop codon readthrough (SCR). SCR of disease-causing premature termination codons (PTCs) is minimal but small molecule interventions, such as treatment with aminoglycoside antibiotics, can enhance its frequency. In this review, we summarize the current understanding of translation termination (both at PTCs and at cognate stop codons) and highlight recently discovered pathways that influence its fidelity. We describe the mechanisms involved in the recognition and readthrough of PTCs and report on SCR-inducing compounds currently explored in preclinical research and clinical trials. We conclude by reviewing the ongoing attempts of personalized nonsense suppression therapy in different disease contexts, including the genetic skin condition epidermolysis bullosa.


Subject(s)
Codon, Nonsense , Genetic Diseases, Inborn , Peptide Chain Elongation, Translational , Precision Medicine , Rare Diseases , Suppression, Genetic , Animals , Humans , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Codon, Nonsense/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Nephritis, Hereditary/genetics , Nephritis, Hereditary/therapy , Nonsense Mediated mRNA Decay , Peptide Chain Elongation, Translational/drug effects , Precision Medicine/methods , Precision Medicine/trends , Rare Diseases/genetics , Rare Diseases/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Shwachman-Diamond Syndrome/genetics , Shwachman-Diamond Syndrome/therapy , Suppression, Genetic/drug effects , Suppression, Genetic/genetics , Peptide Chain Termination, Translational/drug effects , Aminoglycosides/pharmacology
10.
Cell ; 186(7): 1302-1304, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001495

ABSTRACT

CRISPR-Cas9-based base editing allows precise base editing to achieve conversion of adenosine to guanine or cytosine to thymidine. In this issue of Cell, McAuley et al. use adenine base editing to correct a single base-pair mutation causing human CD3δ deficiency, demonstrating superior efficiency of genetic correction with reduced undesired genetic alterations compared with standard CRISPR-Cas9 editing.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Immune System Diseases , Humans , Adenine , CRISPR-Cas Systems/genetics , Genetic Therapy , Mutation , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Immune System Diseases/genetics , Immune System Diseases/therapy
11.
Singapore Med J ; 64(1): 7-16, 2023 01.
Article in English | MEDLINE | ID: mdl-36722512

ABSTRACT

There are more than 7,000 paediatric genetic diseases (PGDs) but less than 5% have treatment options. Treatment strategies targeting different levels of the biological process of the disease have led to optimal health outcomes in a subset of patients with PGDs, where treatment is available. In the past 3 decades, there has been rapid advancement in the development of novel therapies, including gene therapy, for many PGDs. The therapeutic success of treatment relies heavily on knowledge of the genetic basis and the disease mechanism. Specifically, gene therapy has been shown to be effective in various clinical trials, and indeed, these trials have led to regulatory approvals, paving the way for gene therapies for other types of PGDs. In this review, we provide an overview of the treatment strategies and focus on some of the recent advancements in therapeutics for PGDs.


Subject(s)
Genetic Diseases, Inborn , Child , Humans , Genetic Diseases, Inborn/therapy , Genetic Therapy
13.
Article in English | WPRIM (Western Pacific) | ID: wpr-969660

ABSTRACT

There are more than 7,000 paediatric genetic diseases (PGDs) but less than 5% have treatment options. Treatment strategies targeting different levels of the biological process of the disease have led to optimal health outcomes in a subset of patients with PGDs, where treatment is available. In the past 3 decades, there has been rapid advancement in the development of novel therapies, including gene therapy, for many PGDs. The therapeutic success of treatment relies heavily on knowledge of the genetic basis and the disease mechanism. Specifically, gene therapy has been shown to be effective in various clinical trials, and indeed, these trials have led to regulatory approvals, paving the way for gene therapies for other types of PGDs. In this review, we provide an overview of the treatment strategies and focus on some of the recent advancements in therapeutics for PGDs.


Subject(s)
Child , Humans , Genetic Diseases, Inborn/therapy , Genetic Therapy
15.
Nature ; 604(7905): 343-348, 2022 04.
Article in English | MEDLINE | ID: mdl-35322228

ABSTRACT

Gene therapy is a potentially curative medicine for many currently untreatable diseases, and recombinant adeno-associated virus (rAAV) is the most successful gene delivery vehicle for in vivo applications1-3. However, rAAV-based gene therapy suffers from several limitations, such as constrained DNA cargo size and toxicities caused by non-physiological expression of a transgene4-6. Here we show that rAAV delivery of a suppressor tRNA (rAAV.sup-tRNA) safely and efficiently rescued a genetic disease in a mouse model carrying a nonsense mutation, and effects lasted for more than 6 months after a single treatment. Mechanistically, this was achieved through a synergistic effect of premature stop codon readthrough and inhibition of nonsense-mediated mRNA decay. rAAV.sup-tRNA had a limited effect on global readthrough at normal stop codons and did not perturb endogenous tRNA homeostasis, as determined by ribosome profiling and tRNA sequencing, respectively. By optimizing the AAV capsid and the route of administration, therapeutic efficacy in various target tissues was achieved, including liver, heart, skeletal muscle and brain. This study demonstrates the feasibility of developing a toolbox of AAV-delivered nonsense suppressor tRNAs operating on premature termination codons (AAV-NoSTOP) to rescue pathogenic nonsense mutations and restore gene function under endogenous regulation. As nonsense mutations account for 11% of pathogenic mutations, AAV-NoSTOP can benefit a large number of patients. AAV-NoSTOP obviates the need to deliver a full-length protein-coding gene that may exceed the rAAV packaging limit, elicit adverse immune responses or cause transgene-related toxicities. It therefore represents a valuable addition to gene therapeutics.


Subject(s)
Codon, Nonsense , Dependovirus , Genetic Therapy , Adenoviridae , Animals , Codon, Nonsense/genetics , Codon, Terminator/genetics , Codon, Terminator/metabolism , Dependovirus/genetics , Genetic Diseases, Inborn/therapy , Genetic Vectors , Humans , Mice , Nonsense Mediated mRNA Decay/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism
16.
Life Sci ; 294: 120375, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35123997

ABSTRACT

Gene therapy is the product of man's quest to eliminate diseases. Gene therapy has three facets namely, gene silencing using siRNA, shRNA and miRNA, gene replacement where the desired gene in the form of plasmids and viral vectors, are directly administered and finally gene editing based therapy where mutations are modified using specific nucleases such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regulatory interspaced short tandem repeats (CRISPR)/CRISPR-associated protein (Cas)-associated nucleases. Transfer of gene is either through transformation where under specific conditions the gene is directly taken up by the bacterial cells, transduction where a bacteriophage is used to transfer the genetic material and lastly transfection that involves forceful delivery of gene using either viral or non-viral vectors. The non-viral transfection methods are subdivided into physical, chemical and biological. The physical methods include electroporation, biolistic, microinjection, laser, elevated temperature, ultrasound and hydrodynamic gene transfer. The chemical methods utilize calcium- phosphate, DAE-dextran, liposomes and nanoparticles for transfection. The biological methods are increasingly using viruses for gene transfer, these viruses could either integrate within the genome of the host cell conferring a stable gene expression, whereas few other non-integrating viruses are episomal and their expression is diluted proportional to the cell division. So far, gene therapy has been wielded in a plethora of diseases. However, coherent and innocuous delivery of genes is among the major hurdles in the use of this promising therapy. Hence this review aims to highlight the current options available for gene transfer along with the advantages and limitations of every method.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Gene Transfer Techniques , Genetic Diseases, Inborn/therapy , Genetic Therapy , Genetic Vectors/therapeutic use , Genetic Diseases, Inborn/genetics , Humans
17.
AAPS J ; 24(1): 31, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102450

ABSTRACT

Given the recent success of gene therapy modalities and the growing number of cell and gene-based therapies in clinical development across many different therapeutic areas, it is evident that this evolving field holds great promise for the unmet medical needs of patients. The recent approvals of Luxturna® and Zolgensma® prove that recombinant adeno-associated virus (rAAV)-based gene therapy is a transformative modality that enables curative treatment for genetic disorders. Over the last decade, Takeda has accumulated significant experience with rAAV-based gene therapies, especially in the early stage of development. In this review, based on the learnings from Takeda and publicly available information, we aim to provide a guiding perspective on Drug Metabolism and Pharmacokinetics (DMPK) substantial role in advancing therapeutic gene therapy modalities from nonclinical research to clinical development, in particular the characterization of gene therapy product biodistribution, elimination (shedding), immunogenicity assessment, multiple platform bioanalytical assays, and first-in-human (FIH) dose projection strategies. Graphical abstract.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Animals , Biological Products , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Humans , Recombinant Fusion Proteins/genetics
18.
Int J Mol Sci ; 23(2)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35054919

ABSTRACT

Inherited retinal diseases (IRDs) are a leading cause of blindness. To date, 260 disease-causing genes have been identified, but there is currently a lack of available and effective treatment options. Cone photoreceptors are responsible for daylight vision but are highly susceptible to disease progression, the loss of cone-mediated vision having the highest impact on the quality of life of IRD patients. Cone degeneration can occur either directly via mutations in cone-specific genes (primary cone death), or indirectly via the primary degeneration of rods followed by subsequent degeneration of cones (secondary cone death). How cones degenerate as a result of pathological mutations remains unclear, hindering the development of effective therapies for IRDs. This review aims to highlight similarities and differences between primary and secondary cone cell death in inherited retinal diseases in order to better define cone death mechanisms and further identify potential treatment options.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Predisposition to Disease , Retinal Cone Photoreceptor Cells/metabolism , Retinal Diseases/genetics , Retinal Diseases/metabolism , Animals , Apoptosis/genetics , Autophagy/genetics , Biomarkers , Cell Death , Endoplasmic Reticulum Stress , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/therapy , Humans , Oxidative Stress , Retinal Diseases/diagnosis , Retinal Diseases/therapy , Signal Transduction
19.
N Engl J Med ; 385(24): 2264-2270, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34881838

ABSTRACT

Inherited junctional epidermolysis bullosa is a severe genetic skin disease that leads to epidermal loss caused by structural and mechanical fragility of the integuments. There is no established cure for junctional epidermolysis bullosa. We previously reported that genetically corrected autologous epidermal cultures regenerated almost an entire, fully functional epidermis on a child who had a devastating form of junctional epidermolysis bullosa. We now report long-term clinical outcomes in this patient. (Funded by POR FESR 2014-2020 - Regione Emilia-Romagna and others.).


Subject(s)
Epidermis/transplantation , Epidermolysis Bullosa, Junctional/therapy , Keratinocytes/transplantation , Transduction, Genetic , Transgenes , Cell Self Renewal , Cells, Cultured/transplantation , Child , Clone Cells , Epidermis/pathology , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/pathology , Follow-Up Studies , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/therapy , Genetic Therapy , Genetic Vectors , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Male , Regeneration , Stem Cells/physiology , Transplantation, Autologous
20.
Int J Mol Sci ; 22(24)2021 Dec 12.
Article in English | MEDLINE | ID: mdl-34948153

ABSTRACT

Rare genetic diseases are a group of pathologies with often unmet clinical needs. Even if rare by a single genetic disease (from 1/2000 to 1/more than 1,000,000), the total number of patients concerned account for approximatively 400 million peoples worldwide. Finding treatments remains challenging due to the complexity of these diseases, the small number of patients and the challenge in conducting clinical trials. Therefore, innovative preclinical research strategies are required. The zebrafish has emerged as a powerful animal model for investigating rare diseases. Zebrafish combines conserved vertebrate characteristics with high rate of breeding, limited housing requirements and low costs. More than 84% of human genes responsible for diseases present an orthologue, suggesting that the majority of genetic diseases could be modelized in zebrafish. In this review, we emphasize the unique advantages of zebrafish models over other in vivo models, particularly underlining the high throughput phenotypic capacity for therapeutic screening. We briefly introduce how the generation of zebrafish transgenic lines by gene-modulating technologies can be used to model rare genetic diseases. Then, we describe how zebrafish could be phenotyped using state-of-the-art technologies. Two prototypic examples of rare diseases illustrate how zebrafish models could play a critical role in deciphering the underlying mechanisms of rare genetic diseases and their use to identify innovative therapeutic solutions.


Subject(s)
Genetic Diseases, Inborn , Models, Genetic , Rare Diseases , Zebrafish , Animals , Biomedical Research , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans , Rare Diseases/genetics , Rare Diseases/metabolism , Rare Diseases/therapy , Zebrafish/genetics , Zebrafish/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...