Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
Add more filters

Publication year range
1.
RNA Biol ; 19(1): 176-190, 2022.
Article in English | MEDLINE | ID: mdl-35067193

ABSTRACT

RNA-based therapeutics have entered the mainstream with seemingly limitless possibilities to treat all categories of neurological disease. Here, common RNA-based drug modalities such as antisense oligonucleotides, small interfering RNAs, RNA aptamers, RNA-based vaccines and mRNA drugs are reviewed highlighting their current and potential applications. Rapid progress has been made across rare genetic diseases and neurodegenerative disorders, but safe and effective delivery to the brain remains a significant challenge for many applications. The advent of individualized RNA-based therapies for ultra-rare diseases is discussed against the backdrop of the emergence of this field into more common conditions such as Alzheimer's disease and ischaemic stroke. There remains significant untapped potential in the use of RNA-based therapeutics for behavioural disorders and tumours of the central nervous system; coupled with the accelerated development expected over the next decade, the true potential of RNA-based therapeutics to transform the therapeutic landscape in neurology remains to be uncovered.


Subject(s)
Genetic Therapy , Nervous System Diseases/therapy , RNA/genetics , RNA/therapeutic use , Animals , Aptamers, Nucleotide , Disease Management , Disease Susceptibility , Gene Expression Regulation , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Nervous System Diseases/etiology , RNA/chemistry , RNA Interference , RNA, Small Interfering , RNAi Therapeutics , Targeted Gene Repair
2.
J Autoimmun ; 114: 102529, 2020 11.
Article in English | MEDLINE | ID: mdl-32782117

ABSTRACT

The identification of RNA interference (RNAi) has caused a growing interest in harnessing its potential in the treatment of different diseases. Modulation of dysregulated genes through targeting by RNAi represents a potential approach with which to alter the biological pathways at a post-transcriptional level, especially as it pertains to autoimmunity and malignancy. Short hairpin RNAs (shRNA), short interfering RNAs (siRNA), and microRNAs (miRNA) are mainly involved as effector mechanisms in the targeting of RNAi biological pathways. The manipulation and delivery of these molecules in an efficient way promotes the specificity and stability of RNAi-based systems, while minimizing the unwanted adverse reactions by the immune system and reducing cytotoxicity and off-target effects. Advances made to date in identifying the etiopathogenesis of autoimmune diseases has prompted the utilization of RNAi-based systems in vitro and in vivo. Future investigations aimed at deciphering the molecular basis of RNAi and optimizing the delivery of RNAi-based targeting systems will hopefully promote the applicability of such regulatory mechanisms and, ultimately, transfer the acquired knowledge from bench-to-bedside to ameliorate human diseases. In this review, we seek to clarify the potential of RNAi, with a focus on siRNAs, in designing therapeutics for potential treatment of human autoimmune disorders.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/therapy , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Animals , Gene Expression Regulation , Genetic Therapy/methods , Humans , MicroRNAs/genetics , Targeted Gene Repair , Translational Research, Biomedical
3.
Nature ; 510(7504): 235-240, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24870228

ABSTRACT

Targeted genome editing by artificial nucleases has brought the goal of site-specific transgene integration and gene correction within the reach of gene therapy. However, its application to long-term repopulating haematopoietic stem cells (HSCs) has remained elusive. Here we show that poor permissiveness to gene transfer and limited proficiency of the homology-directed DNA repair pathway constrain gene targeting in human HSCs. By tailoring delivery platforms and culture conditions we overcame these barriers and provide stringent evidence of targeted integration in human HSCs by long-term multilineage repopulation of transplanted mice. We demonstrate the therapeutic potential of our strategy by targeting a corrective complementary DNA into the IL2RG gene of HSCs from healthy donors and a subject with X-linked severe combined immunodeficiency (SCID-X1). Gene-edited HSCs sustained normal haematopoiesis and gave rise to functional lymphoid cells that possess a selective growth advantage over those carrying disruptive IL2RG mutations. These results open up new avenues for treating SCID-X1 and other diseases.


Subject(s)
Gene Targeting/methods , Genome, Human/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Targeted Gene Repair/methods , X-Linked Combined Immunodeficiency Diseases/genetics , Animals , Antigens, CD34/metabolism , DNA, Complementary/genetics , Endonucleases/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Fetal Blood/transplantation , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Humans , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mutation/genetics , X-Linked Combined Immunodeficiency Diseases/therapy
4.
Int J Gynecol Cancer ; 30(10): 1608-1618, 2020 10.
Article in English | MEDLINE | ID: mdl-32928926

ABSTRACT

The clinical development of poly-(ADP)-ribose polymerase inhibitors (PARPi) began with the treatment of ovarian cancer patients harboring BRCA1/2 mutations and continues to be expanded to other gynecological cancers. Furthermore, The Cancer Genome Atlas (TCGA) analysis of endometrial and cervical cancers offered rationale that PARPi may be an option for treatment based on the molecular profiles of these cancer types. This review summarizes the current indications of PARPi, such as its role in the treatment and maintenance of recurrent ovarian cancer and for first-line maintenance therapy in advanced ovarian cancer. We also outline new concepts for PARPi therapy in other gynecological cancers such as endometrial and cervical cancers based on recent clinical data. Finally, we present potential future directions to continue exploring the world of PARPi resistance and combining PARPi with other therapies.


Subject(s)
Endometrial Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Uterine Cervical Neoplasms/drug therapy , DNA Damage/drug effects , Female , Humans , Neoplasm Recurrence, Local/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Progression-Free Survival , Targeted Gene Repair/methods
5.
Genomics ; 111(4): 560-566, 2019 07.
Article in English | MEDLINE | ID: mdl-29605634

ABSTRACT

The ability to direct the CRISPR/Cas9 nuclease to a unique target site within a genome would have broad use in targeted genome engineering. However, CRISPR RNA is reported to bind to other genomic locations that differ from the intended target site by a few nucleotides, demonstrating significant off-target activity. We have developed the CRISPcut tool that screens the off-targets using various parameters and predicts the ideal genomic target for -guide RNAs in human cell lines. sgRNAs for four different types of Cas9 nucleases can be designed with an option for the user to work with different PAM sequences. Direct experimental measurement of genome-wide DNA accessibility is incorporated that effectively restricts the prediction of CRISPR targets to open chromatin. An option to predict target sites for paired CRISPR nickases is also provided. The tool has been validated using a dataset of experimentally used sgRNA and their identified off-targets. URL: http://web.iitd.ac.in/crispcut.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , RNA, Guide, Kinetoplastida/genetics , Software , Targeted Gene Repair/methods , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Chromatin/chemistry , Humans , Nucleotide Motifs , RNA, Guide, Kinetoplastida/metabolism
6.
Int J Mol Sci ; 21(11)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471232

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease caused by the death of motor neurons in the spinal cord and brainstem. ALS has a diverse genetic origin; at least 20 genes have been shown to be related to ALS. Most familial and sporadic cases of ALS are caused by variants of the SOD1, C9orf72, FUS, and TARDBP genes. Genome editing using clustered regularly interspaced short palindromic repeats/CRISPR-associated system 9 (CRISPR/Cas9) can provide insights into the underlying genetics and pathophysiology of ALS. By correcting common mutations associated with ALS in animal models and patient-derived induced pluripotent stem cells (iPSCs), CRISPR/Cas9 has been used to verify the effects of ALS-associated mutations and observe phenotype differences between patient-derived and gene-corrected iPSCs. This technology has also been used to create mutations to investigate the pathophysiology of ALS. Here, we review recent studies that have used CRISPR/Cas9 to understand the genetic underpinnings of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , CRISPR-Cas Systems , Targeted Gene Repair/methods , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein/genetics , DNA-Binding Proteins/genetics , Humans , RNA-Binding Protein FUS/genetics , Superoxide Dismutase-1/genetics
7.
Int J Mol Sci ; 21(23)2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33266216

ABSTRACT

Using nanoparticles to carry and delivery anticancer drugs holds much promise in cancer therapy, but nanoparticles per se are lacking specificity. Active targeting, that is, using specific ligands to functionalize nanoparticles, is attracting much attention in recent years. Aptamers, with their several favorable features like high specificity and affinity, small size, very low immunogenicity, relatively low cost for production, and easiness to store, are one of the best candidates for the specific ligands of nanoparticle functionalization. This review discusses the benefits and challenges of using aptamers to functionalize nanoparticles for active targeting and especially presents nearly all of the published works that address the topic of using aptamers to functionalize nanoparticles for targeted drug delivery and cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/administration & dosage , Drug Carriers , Drug Delivery Systems , Nanoparticles , Animals , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Drug Carriers/chemistry , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Targeted Therapy , Nanoparticles/chemistry , Neoplasms/etiology , Neoplasms/pathology , Neoplasms/therapy , Targeted Gene Repair , Theranostic Nanomedicine
8.
Int J Mol Sci ; 21(8)2020 Apr 23.
Article in English | MEDLINE | ID: mdl-32340368

ABSTRACT

RNA-binding proteins (RBPs) are involved in regulating all aspects of RNA metabolism, including processing, transport, translation, and degradation. Dysregulation of RNA metabolism is linked to a plethora of diseases, such as cancer, neurodegenerative diseases, and neuromuscular disorders. Recent years have seen a dramatic shift in the knowledge base, with RNA increasingly being recognised as an attractive target for precision medicine therapies. In this article, we are going to review current RNA-targeted therapies. Furthermore, we will scrutinise a range of drug discoveries targeting protein-RNA interactions. In particular, we will focus on the interplay between Lin28 and let-7, splicing regulatory proteins and survival motor neuron (SMN) pre-mRNA, as well as HuR, Musashi, proteins and their RNA targets. We will highlight the mechanisms RBPs utilise to modulate RNA metabolism and discuss current high-throughput screening strategies. This review provides evidence that we are entering a new era of RNA-targeted medicine.


Subject(s)
Drug Discovery , Genetic Therapy , High-Throughput Screening Assays , Molecular Targeted Therapy , RNA/genetics , Animals , Biomarkers , Clinical Studies as Topic , Drug Evaluation, Preclinical , Humans , Molecular Targeted Therapy/methods , RNA/chemistry , RNA/metabolism , RNA Interference , RNA-Binding Proteins/metabolism , Targeted Gene Repair , Treatment Outcome
9.
RNA Biol ; 16(10): 1414-1423, 2019 10.
Article in English | MEDLINE | ID: mdl-31232219

ABSTRACT

The X-linked CDKL5 gene codes for a kinase whose mutations have been associated with a suite of neurodevelopmental disorders generally characterized by early-onset epileptic encephalopathy and severe intellectual disability. The impact of these mutations on CDKL5 functions and brain development remain mainly unknown, although the importance of maintaining the catalytic activity is generally recognized. Since no cure exists for CDKL5 disorders, the demand for innovative therapies is a real emergency. The recent discovery that CDKL5 is dosage sensitive poses concerns on conventional protein and gene augmentative therapies. Thus, RNA-based therapeutic approaches might be preferred. We studied the efficacy of read-through therapy on CDKL5 premature termination codons (PTCs) that correspond roughly to 15% of all mutations. Our results provide the first demonstration that all tested CDKL5 nonsense mutations are efficiently suppressed by aminoglycoside drugs. The functional characterization of the restored full-length CDKL5 reveals that read-through proteins fully recover their subcellular localization, but only partially rescue their catalytic activity. Since read-through can cause amino acid substitution, CDKL5 patients carrying the PTC outside the catalytic domain might benefit more from a nonsense suppression therapy. Eventually, we demonstrate that non-aminoglycoside drugs, such as Ataluren (PTC124) and GJ072, are unable to induce read-through activity on CDKL5 PTCs. Although these drugs might be more effective in vivo, these results question the validity of the Ataluren phase 2 clinical trial that is currently ongoing on CDKL5 patients.


Subject(s)
Aminoglycosides/pharmacology , Codon, Nonsense , Gene Expression Regulation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Disease Models, Animal , Enzyme Activation/drug effects , Epileptic Syndromes/genetics , Epileptic Syndromes/metabolism , Epileptic Syndromes/physiopathology , Epileptic Syndromes/therapy , Humans , Mice , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/physiopathology , Neurodevelopmental Disorders/therapy , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Spasms, Infantile/genetics , Spasms, Infantile/metabolism , Spasms, Infantile/physiopathology , Spasms, Infantile/therapy , Targeted Gene Repair
10.
Int J Mol Sci ; 20(15)2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31366028

ABSTRACT

The CRISPR/Cas9 system (clustered regularly interspaced short palindromic repeat-associated protein 9) is a powerful genome-editing tool in animals, plants, and humans. This system has some advantages, such as a high on-target mutation rate (targeting efficiency), less cost, simplicity, and high-efficiency multiplex loci editing, over conventional genome editing tools, including meganucleases, transcription activator-like effector nucleases (TALENs), and zinc finger nucleases (ZFNs). One of the crucial shortcomings of this system is unwanted mutations at off-target sites. We summarize and discuss different approaches, such as dCas9 and Cas9 paired nickase, to decrease the off-target effects in plants. According to studies, the most effective method to reduce unintended mutations is the use of ligand-dependent ribozymes called aptazymes. The single guide RNA (sgRNA)/ligand-dependent aptazyme strategy has helped researchers avoid unwanted mutations in human cells and can be used in plants as an alternative method to dramatically decrease the frequency of off-target mutations. We hope our concept provides a new, simple, and fast gene transformation and genome-editing approach, with advantages including reduced time and energy consumption, the avoidance of unwanted mutations, increased frequency of on-target changes, and no need for external forces or expensive equipment.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Plant Breeding/methods , Targeted Gene Repair/methods , Gene Editing/standards , Magnoliopsida/genetics , RNA, Guide, Kinetoplastida/genetics , Targeted Gene Repair/standards
11.
Int J Mol Sci ; 20(17)2019 Aug 24.
Article in English | MEDLINE | ID: mdl-31450582

ABSTRACT

Mutations in the CDKL5 gene lead to an incurable rare neurological condition characterized by the onset of seizures in the first weeks of life and severe intellectual disability. Replacement gene or protein therapies could represent intriguing options, however, their application may be inhibited by the recent demonstration that CDKL5 is dosage sensitive. Conversely, correction approaches acting on pre-mRNA splicing would preserve CDKL5 physiological regulation. Since ~15% of CDKL5 pathogenic mutations are candidates to affect splicing, we evaluated the capability of variants of the spliceosomal U1 small nuclear RNA (U1snRNA) to correct mutations affecting +1 and +5 nucleotides at the 5' donor splice site and predicted to cause exon skipping. Our results show that CDKL5 minigene variants expressed in mammalian cells are a valid approach to assess CDKL5 splicing pattern. The expression of engineered U1snRNA effectively rescued mutations at +5 but not at the +1 nucleotides. Importantly, we proved that U1snRNA-mediated splicing correction fully restores CDKL5 protein synthesis, subcellular distribution and kinase activity. Eventually, by correcting aberrant splicing of an exogenously expressed splicing-competent CDKL5 transgene, we provided insights on the morphological rescue of CDKL5 null neurons, reporting the first proof-of-concept of the therapeutic value of U1snRNA-mediated CDKL5 splicing correction.


Subject(s)
Mutation , Protein Serine-Threonine Kinases/genetics , RNA Splicing , RNA, Small Nuclear/genetics , Targeted Gene Repair , Alleles , Alternative Splicing , Cell Line , Epileptic Syndromes/genetics , Epileptic Syndromes/therapy , Exons , Gene Expression , Genetic Loci , Genetic Therapy , Genotype , Humans , Neurons/metabolism , Nonsense Mediated mRNA Decay , Protein Serine-Threonine Kinases/metabolism , Spasms, Infantile/genetics , Spasms, Infantile/therapy
12.
Medicina (Kaunas) ; 55(8)2019 Jul 28.
Article in English | MEDLINE | ID: mdl-31357735

ABSTRACT

The upgraded knowledge of tumor biology and microenviroment provides information on differences in neoplastic and normal cells. Thus, the need to target these differences led to the development of novel molecules (targeted therapy) active against the neoplastic cells' inner workings. There are several types of targeted agents, including Small Molecules Inhibitors (SMIs), monoclonal antibodies (mAbs), interfering RNA (iRNA) molecules and microRNA. In the clinical practice, these new medicines generate a multilayered step in pharmacokinetics (PK), which encompasses a broad individual PK variability, and unpredictable outcomes according to the pharmacogenetics (PG) profile of the patient (e.g., cytochrome P450 enzyme), and to patient characteristics such as adherence to treatment and environmental factors. This review focuses on the use of targeted agents in-human phase I/II/III clinical trials in cancer-hematology. Thus, it outlines the up-to-date anticancer drugs suitable for targeted therapies and the most recent finding in pharmacogenomics related to drug response. Besides, a summary assessment of the genotyping costs has been discussed. Targeted therapy seems to be an effective and less toxic therapeutic approach in onco-hematology. The identification of individual PG profile should be a new resource for oncologists to make treatment decisions for the patients to minimize the toxicity and or inefficacy of therapy. This could allow the clinicians to evaluate benefits and restrictions, regarding costs and applicability, of the most suitable pharmacological approach for performing a tailor-made therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Targeted Gene Repair/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Humans , MicroRNAs/pharmacology , MicroRNAs/therapeutic use , Oncolytic Viruses , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Review Literature as Topic , Targeted Gene Repair/statistics & numerical data
13.
Medicina (Kaunas) ; 55(2)2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30743998

ABSTRACT

Cholangiocarcinoma (CCA) is a highly-aggressive malignancy arising from the biliary tree, characterized by a steady increase in incidence globally and a high mortality rate. Most CCAs are diagnosed in the advanced and metastatic phases of the disease, due to the paucity of signs and symptoms in the early stages. This fact, along with the poor results of the local and systemic therapies currently employed, is responsible for the poor outcome of CCA patients and strongly supports the need for novel therapeutic agents and strategies. In recent years, the introduction of next-generation sequencing technologies has opened new horizons for a better understanding of the genetic pathophysiology of CCA and, consequently, for the identification and evaluation of new treatments tailored to the molecular features or alterations progressively elucidated. In this review article, we describe the potential targets under investigation and the current molecular therapies employed in biliary tract cancers. In addition, we summarize the main drugs against CCA under evaluation in ongoing trials and describe the preliminary data coming from these pioneering studies.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Enzyme Inhibitors/therapeutic use , Immunotherapy , Molecular Targeted Therapy , Clinical Trials as Topic , Feedback, Physiological , High-Throughput Nucleotide Sequencing , Humans , Signal Transduction/drug effects , Targeted Gene Repair
14.
Blood ; 127(21): 2553-60, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27053531

ABSTRACT

Gene editing is a rapidly developing area of biotechnology in which the nucleotide sequence of the genome of living cells is precisely changed. The use of genome-editing technologies to modify various types of blood cells, including hematopoietic stem cells, has emerged as an important field of therapeutic development for hematopoietic disease. Although these technologies offer the potential for generation of transformative therapies for patients suffering from myriad disorders of hematopoiesis, their application for therapeutic modification of primary human cells is still in its infancy. Consequently, development of ethical and regulatory frameworks that ensure their safe and effective use is an increasingly important consideration. Here, we review a number of issues that have the potential to impact the clinical implementation of genome-editing technologies, and suggest paths forward for resolving them such that new therapies can be safely and rapidly translated to the clinic.


Subject(s)
Bioethical Issues , Gene Editing , Animals , Gene Editing/ethics , Gene Editing/legislation & jurisprudence , Gene Editing/methods , Humans , Targeted Gene Repair/ethics , Targeted Gene Repair/legislation & jurisprudence , Targeted Gene Repair/methods
15.
Blood ; 127(21): 2523-4, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27053534

ABSTRACT

Our capacities to understand and manipulate mammalian genomes are accelerating at an astounding pace. In 2007, Capecchi, Evans, and Smithies were awarded the Nobel Prize in medicine for their work on gene targeting, which showed that embryonic stem cells could be modified by homologous recombination (HR) with engineered template DNA to alter virtually any gene and create mutant mice. This work revolutionized biology by allowing investigators to study the in vivo consequences of selected gene alteration. However, the efficiency of HR in embryonic stem cells is unpredictable, depending on the target gene and HR template. More importantly, spontaneous HR occurs at very low rates in most somatic cells, restricting the use of standard gene targeting for most laboratory and clinical applications. This limitation is being overcome by genome-editing technologies, which markedly enhance the capacity to alter cellular genes with laser-like precision. Four review articles in this edition of Blood summarize the field of genome editing, focusing on its potential for treating hematological disorders.


Subject(s)
Gene Editing/methods , Gene Targeting/methods , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Targeted Gene Repair/methods , Animals , Gene Editing/history , Gene Editing/trends , Gene Targeting/history , Gene Targeting/trends , History, 21st Century , Humans , Mice , Targeted Gene Repair/history , Targeted Gene Repair/trends
16.
Blood ; 127(21): 2525-35, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27053532

ABSTRACT

Gene editing enables the site-specific modification of the genome. These technologies have rapidly advanced such that they have entered common use in experimental hematology to investigate genetic function. In addition, genome editing is becoming increasingly plausible as a treatment modality to rectify genetic blood disorders and improve cellular therapies. Genome modification typically ensues from site-specific double-strand breaks and may result in a myriad of outcomes. Even single-strand nicks and targeted biochemical modifications that do not permanently alter the DNA sequence (epigenome editing) may be powerful instruments. In this review, we examine the various technologies, describe their advantages and shortcomings for engendering useful genetic alterations, and consider future prospects for genome editing to impact hematology.


Subject(s)
Gene Editing/methods , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Targeted Gene Repair/methods , Animals , Humans
17.
Blood ; 127(21): 2536-45, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27053533

ABSTRACT

Despite nearly complete understanding of the genetics of the ß-hemoglobinopathies for several decades, definitive treatment options have lagged behind. Recent developments in technologies for facile manipulation of the genome (zinc finger nucleases, transcription activator-like effector nucleases, or clustered regularly interspaced short palindromic repeats-based nucleases) raise prospects for their clinical application. The use of genome-editing technologies in autologous CD34(+) hematopoietic stem and progenitor cells represents a promising therapeutic avenue for the ß-globin disorders. Genetic correction strategies relying on the homology-directed repair pathway may repair genetic defects, whereas genetic disruption strategies relying on the nonhomologous end joining pathway may induce compensatory fetal hemoglobin expression. Harnessing the power of genome editing may usher in a second-generation form of gene therapy for the ß-globin disorders.


Subject(s)
Gene Editing/methods , Hematopoietic Stem Cell Transplantation , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Targeted Gene Repair/methods , Animals , Autografts , Humans
18.
Blood ; 127(21): 2546-52, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27053530

ABSTRACT

HIV/AIDS has long been at the forefront of the development of gene- and cell-based therapies. Although conventional gene therapy approaches typically involve the addition of anti-HIV genes to cells using semirandomly integrating viral vectors, newer genome editing technologies based on engineered nucleases are now allowing more precise genetic manipulations. The possible outcomes of genome editing include gene disruption, which has been most notably applied to the CCR5 coreceptor gene, or the introduction of small mutations or larger whole gene cassette insertions at a targeted locus. Disruption of CCR5 using zinc finger nucleases was the first-in-human application of genome editing and remains the most clinically advanced platform, with 7 completed or ongoing clinical trials in T cells and hematopoietic stem/progenitor cells (HSPCs). Here we review the laboratory and clinical findings of CCR5 editing in T cells and HSPCs for HIV therapy and summarize other promising genome editing approaches for future clinical development. In particular, recent advances in the delivery of genome editing reagents and the demonstration of highly efficient homology-directed editing in both T cells and HSPCs are expected to spur the development of even more sophisticated applications of this technology for HIV therapy.


Subject(s)
Acquired Immunodeficiency Syndrome , Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Mutagenesis, Insertional , Receptors, CCR5 , T-Lymphocytes/metabolism , Targeted Gene Repair/methods , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/metabolism , Acquired Immunodeficiency Syndrome/therapy , Deoxyribonucleases/genetics , Humans , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
19.
Blood ; 127(21): 2513-22, 2016 05 26.
Article in English | MEDLINE | ID: mdl-26903548

ABSTRACT

Loss of CD40 ligand (CD40L) expression or function results in X-linked hyper-immunoglobulin (Ig)M syndrome (X-HIGM), characterized by recurrent infections due to impaired immunoglobulin class-switching and somatic hypermutation. Previous attempts using retroviral gene transfer to correct murine CD40L expression restored immune function; however, treated mice developed lymphoproliferative disease, likely due to viral-promoter-dependent constitutive CD40L expression. These observations highlight the importance of preserving endogenous gene regulation in order to safely correct this disorder. Here, we report efficient, on-target, homology-directed repair (HDR) editing of the CD40LG locus in primary human T cells using a combination of a transcription activator-like effector nuclease-induced double-strand break and a donor template delivered by recombinant adeno-associated virus. HDR-mediated insertion of a coding sequence (green fluorescent protein or CD40L) upstream of the translation start site within exon 1 allowed transgene expression to be regulated by endogenous CD40LG promoter/enhancer elements. Additionally, inclusion of the CD40LG 3'-untranslated region in the transgene preserved posttranscriptional regulation. Expression kinetics of the transgene paralleled that of endogenous CD40L in unedited T cells, both at rest and in response to T-cell stimulation. The use of this method to edit X-HIGM patient T cells restored normal expression of CD40L and CD40-murine IgG Fc fusion protein (CD40-muIg) binding, and rescued IgG class switching of naive B cells in vitro. These results demonstrate the feasibility of engineered nuclease-directed gene repair to restore endogenously regulated CD40L, and the potential for its use in T-cell therapy for X-HIGM syndrome.


Subject(s)
B-Lymphocytes/immunology , CD40 Ligand , Gene Editing/methods , Gene Expression Regulation/immunology , Hyper-IgM Immunodeficiency Syndrome, Type 1 , T-Lymphocytes/immunology , Targeted Gene Repair/methods , 3' Untranslated Regions/immunology , Animals , CD40 Ligand/genetics , CD40 Ligand/immunology , Enhancer Elements, Genetic/immunology , Female , Humans , Hyper-IgM Immunodeficiency Syndrome, Type 1/genetics , Hyper-IgM Immunodeficiency Syndrome, Type 1/immunology , Hyper-IgM Immunodeficiency Syndrome, Type 1/therapy , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology
20.
Mol Ther ; 25(9): 2117-2128, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28629821

ABSTRACT

Glycogen storage disease type II (GSDII) is a lysosomal disorder caused by the deficient activity of acid alpha-glucosidase (GAA) enzyme, leading to the accumulation of glycogen within the lysosomes. The disease has been classified in infantile and late-onset forms. Most late-onset patients share a splicing mutation c.-32-13T > G in intron 1 of the GAA gene that prevents efficient recognition of exon 2 by the spliceosome. In this study, we have mapped the splicing silencers of GAA exon 2 and developed antisense morpholino oligonucleotides (AMOs) to inhibit those regions and rescue normal splicing in the presence of the c.-32-13T > G mutation. Using a minigene approach and patient fibroblasts, we successfully increased inclusion of exon 2 in the mRNA and GAA enzyme production by targeting a specific silencer with a combination of AMOs. Most importantly, the use of these AMOs in patient myotubes results in a decreased accumulation of glycogen. To our knowledge, this is the only therapeutic approach resulting in a decrease of glycogen accumulation in patient tissues beside enzyme replacement therapy (ERT) and TFEB overexpression. As a result, it may represent a highly novel and promising therapeutic line for GSDII.


Subject(s)
Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/metabolism , Glycogen/metabolism , Muscle Fibers, Skeletal/metabolism , Oligonucleotides, Antisense/genetics , Targeted Gene Repair , Alleles , Cell Line , Exons , Gene Order/genetics , Genetic Vectors/genetics , Glycogen Storage Disease Type II/therapy , Humans , Mutation , Oligonucleotides, Antisense/therapeutic use , Protein Binding , RNA Splicing , RNA Splicing Factors/metabolism , Silencer Elements, Transcriptional , Targeted Gene Repair/methods , alpha-Glucosidases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL