Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Hum Gene Ther ; 35(11-12): 374-387, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38717950

RESUMEN

The ongoing advancements in CRISPR-Cas technologies can significantly accelerate the preclinical development of both in vivo and ex vivo organ genome-editing therapeutics. One of the promising applications is to genetically modify donor organs prior to implantation. The implantation of optimized donor organs with long-lasting immunomodulatory capacity holds promise for reducing the need for lifelong potent whole-body immunosuppression in recipients. However, assessing genome-targeting interventions in a clinically relevant manner prior to clinical trials remains a major challenge owing to the limited modalities available. This study introduces a novel platform for testing genome editing in human lungs ex vivo, effectively simulating preimplantation genetic engineering of donor organs. We identified gene regulatory elements whose disruption via Cas nucleases led to the upregulation of the immunomodulatory gene interleukin 10 (IL-10). We combined this approach with adenoviral vector-mediated IL-10 delivery to create favorable kinetics for early (immediate postimplantation) graft immunomodulation. Using ex vivo organ machine perfusion and precision-cut tissue slice technology, we demonstrated the feasibility of evaluating CRISPR genome editing in human lungs. To overcome the assessment limitations in ex vivo perfused human organs, we conducted an in vivo rodent study and demonstrated both early gene induction and sustained editing of the lung. Collectively, our findings lay the groundwork for a first-in-human-organ study to overcome the current translational barriers of genome-targeting therapeutics.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Pulmón , Edición Génica/métodos , Humanos , Pulmón/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Animales , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación
2.
Nat Commun ; 15(1): 3182, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38609352

RESUMEN

Huntington's disease (HD) is a dominant neurological disorder caused by an expanded HTT exon 1 CAG repeat that lengthens huntingtin's polyglutamine tract. Lowering mutant huntingtin has been proposed for treating HD, but genetic modifiers implicate somatic CAG repeat expansion as the driver of onset. We find that branaplam and risdiplam, small molecule splice modulators that lower huntingtin by promoting HTT pseudoexon inclusion, also decrease expansion of an unstable HTT exon 1 CAG repeat in an engineered cell model. Targeted CRISPR-Cas9 editing shows this effect is not due to huntingtin lowering, pointing instead to pseudoexon inclusion in PMS1. Homozygous but not heterozygous inactivation of PMS1 also reduces CAG repeat expansion, supporting PMS1 as a genetic modifier of HD and a potential target for therapeutic intervention. Although splice modulation provides one strategy, genome-wide transcriptomics also emphasize consideration of cell-type specific effects and polymorphic variation at both target and off-target sites.


Asunto(s)
Enfermedad de Huntington , Humanos , Enfermedad de Huntington/genética , Exones/genética , Perfilación de la Expresión Génica , Heterocigoto , Homocigoto , Proteínas MutL , Proteínas de Neoplasias
3.
Nat Biomed Eng ; 8(2): 118-131, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38057426

RESUMEN

Spinal muscular atrophy (SMA) is caused by mutations in SMN1. SMN2 is a paralogous gene with a C•G-to-T•A transition in exon 7, which causes this exon to be skipped in most SMN2 transcripts, and results in low levels of the protein survival motor neuron (SMN). Here we show, in fibroblasts derived from patients with SMA and in a mouse model of SMA that, irrespective of the mutations in SMN1, adenosine base editors can be optimized to target the SMN2 exon-7 mutation or nearby regulatory elements to restore the normal expression of SMN. After optimizing and testing more than 100 guide RNAs and base editors, and leveraging Cas9 variants with high editing fidelity that are tolerant of different protospacer-adjacent motifs, we achieved the reversion of the exon-7 mutation via an A•T-to-G•C edit in up to 99% of fibroblasts, with concomitant increases in the levels of the SMN2 exon-7 transcript and of SMN. Targeting the SMN2 exon-7 mutation via base editing or other CRISPR-based methods may provide long-lasting outcomes to patients with SMA.


Asunto(s)
Atrofia Muscular Espinal , Proteínas de Unión al ARN , Ratones , Animales , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas del Complejo SMN/genética , ARN Guía de Sistemas CRISPR-Cas , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Exones/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética
4.
Mol Ther ; 31(8): 2439-2453, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37312453

RESUMEN

Usher syndrome type 1F (USH1F), characterized by congenital lack of hearing and balance and progressive loss of vision, is caused by mutations in the PCDH15 gene. In the Ashkenazi population, a recessive truncation mutation accounts for a large proportion of USH1F cases. The truncation is caused by a single C→T mutation, which converts an arginine codon to a stop (R245X). To test the potential for base editors to revert this mutation, we developed a humanized Pcdh15R245X mouse model for USH1F. Mice homozygous for the R245X mutation were deaf and exhibited profound balance deficits, while heterozygous mice were unaffected. Here we show that an adenine base editor (ABE) is capable of reversing the R245X mutation to restore the PCDH15 sequence and function. We packaged a split-intein ABE into dual adeno-associated virus (AAV) vectors and delivered them into cochleas of neonatal USH1F mice. Hearing was not restored in a Pcdh15 constitutive null mouse despite base editing, perhaps because of early disorganization of cochlear hair cells. However, injection of vectors encoding the split ABE into a late-deletion conditional Pcdh15 knockout rescued hearing. This study demonstrates the ability of an ABE to correct the PCDH15 R245X mutation in the cochlea and restore hearing.


Asunto(s)
Síndromes de Usher , Ratones , Animales , Síndromes de Usher/genética , Síndromes de Usher/terapia , Edición Génica , Mutación , Audición/genética , Cadherinas/genética
5.
J Heart Lung Transplant ; 42(10): 1363-1377, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37315746

RESUMEN

BACKGROUND: Inflammatory injury in the donor lung remains a persistent challenge in lung transplantation that limits donor organ usage and post-transplant outcomes. Inducing immunomodulatory capacity in donor organs could address this unsolved clinical problem. We sought to apply clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) technologies to the donor lung to fine-tune immunomodulatory gene expression, exploring for the first time the therapeutic use of CRISPR-mediated transcriptional activation in the whole donor lung. METHODS: We explored the feasibility of CRISPR-mediated transcriptional upregulation of interleukin 10 (IL-10), a key immunomodulatory cytokine, in vitro and in vivo. We first evaluated the potency, titratability, and multiplexibility of the gene activation in rat and human cell lines. Next, in vivo CRISPR-mediated IL-10 activation was characterized in rat lungs. Finally, the IL-10-activated donor lungs were transplanted into recipient rats to assess the feasibility in a transplant setting. RESULTS: The targeted transcriptional activation induced robust and titrable IL-10 upregulation in vitro. The combination of guide RNAs also facilitated multiplex gene modulation, that is, simultaneous activation of IL-10 and IL1 receptor antagonist. In vivo profiling demonstrated that adenoviral delivery of Cas9-based activators to the lung was feasible with the use of immunosuppression, which is routinely applied to organ transplant recipients. The transcriptionally modulated donor lungs retained IL-10 upregulation in isogeneic and allogeneic recipients. CONCLUSIONS: Our findings highlight the potential of CRISPR epigenome editing to improve lung transplant outcomes by creating a more favorable immunomodulatory environment in the donor organ, a paradigm that may be extendable to other organ transplants.


Asunto(s)
Edición Génica , Interleucina-10 , Humanos , Animales , Ratas , Interleucina-10/genética , Línea Celular , Pulmón , Inmunomodulación
6.
bioRxiv ; 2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36711797

RESUMEN

Spinal muscular atrophy (SMA) is a devastating neuromuscular disease caused by mutations in the SMN1 gene. Despite the development of various therapies, outcomes can remain suboptimal in SMA infants and the duration of such therapies are uncertain. SMN2 is a paralogous gene that mainly differs from SMN1 by a C•G-to-T•A transition in exon 7, resulting in the skipping of exon 7 in most SMN2 transcripts and production of only low levels of survival motor neuron (SMN) protein. Genome editing technologies targeted to the SMN2 exon 7 mutation could offer a therapeutic strategy to restore SMN protein expression to normal levels irrespective of the patient SMN1 mutation. Here, we optimized a base editing approach to precisely edit SMN2, reverting the exon 7 mutation via an A•T-to-G•C base edit. We tested a range of different adenosine base editors (ABEs) and Cas9 enzymes, resulting in up to 99% intended editing in SMA patient-derived fibroblasts with concomitant increases in SMN2 exon 7 transcript expression and SMN protein levels. We generated and characterized ABEs fused to high-fidelity Cas9 variants which reduced potential off-target editing. Delivery of these optimized ABEs via dual adeno-associated virus (AAV) vectors resulted in precise SMN2 editing in vivo in an SMA mouse model. This base editing approach to correct SMN2 should provide a long-lasting genetic treatment for SMA with advantages compared to current nucleic acid, small molecule, or exogenous gene replacement therapies. More broadly, our work highlights the potential of PAMless SpRY base editors to install edits efficiently and safely.

7.
Mol Ther Oncolytics ; 25: 121-136, 2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35572197

RESUMEN

Non-coding RNAs, including microRNAs (miRNAs), support the progression of glioma. miR-21 is a small, non-coding transcript involved in regulating gene expression in multiple cellular pathways, including the regulation of proliferation. High expression of miR-21 has been shown to be a major driver of glioma growth. Manipulating the expression of miRNAs is a novel strategy in the development of therapeutics in cancer. In this study we aimed to target miR-21. Using CRISPR genome-editing technology, we disrupted the miR-21 coding sequences in glioma cells. Depletion of this miRNA resulted in the upregulation of many downstream miR-21 target mRNAs involved in proliferation. Phenotypically, CRISPR-edited glioma cells showed reduced migration, invasion, and proliferation in vitro. In immunocompetent mouse models, miR-21 knockout tumors showed reduced growth resulting in an increased overall survival. In summary, we show that by knocking out a key miRNA in glioma, these cells have decreased proliferation capacity both in vitro and in vivo. Overall, we identified miR-21 as a potential target for CRISPR-based therapeutics in glioma.

8.
Front Immunol ; 13: 1067417, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36685559

RESUMEN

Introduction: Ex vivo gene therapy for treatment of Inborn errors of Immunity (IEIs) have demonstrated significant clinical benefit in multiple Phase I/II clinical trials. Current approaches rely on engineered retroviral vectors to randomly integrate copy(s) of gene-of-interest in autologous hematopoietic stem/progenitor cells (HSPCs) genome permanently to provide gene function in transduced HSPCs and their progenies. To circumvent concerns related to potential genotoxicities due to the random vector integrations in HSPCs, targeted correction with CRISPR-Cas9-based genome editing offers improved precision for functional correction of multiple IEIs. Methods: We compare the two approaches for integration of IL2RG transgene for functional correction of HSPCs from patients with X-linked Severe Combined Immunodeficiency (SCID-X1 or XSCID); delivery via current clinical lentivector (LV)-IL2RG versus targeted insertion (TI) of IL2RG via homology-directed repair (HDR) when using an adeno-associated virus (AAV)-IL2RG donor following double-strand DNA break at the endogenous IL2RG locus. Results and discussion: In vitro differentiation of LV- or TI-treated XSCID HSPCs similarly overcome differentiation block into Pre-T-I and Pre-T-II lymphocytes but we observed significantly superior development of NK cells when corrected by TI (40.7% versus 4.1%, p = 0.0099). Transplants into immunodeficient mice demonstrated robust engraftment (8.1% and 23.3% in bone marrow) for LV- and TI-IL2RG HSPCs with efficient T cell development following TI-IL2RG in all four patients' HSPCs. Extensive specificity analysis of CRISPR-Cas9 editing with rhAmpSeq covering 82 predicted off-target sites found no evidence of indels in edited cells before (in vitro) or following transplant, in stark contrast to LV's non-targeted vector integration sites. Together, the improved efficiency and safety of IL2RG correction via CRISPR-Cas9-based TI approach provides a strong rationale for a clinical trial for treatment of XSCID patients.


Asunto(s)
Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Animales , Ratones , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Dependovirus , Sistemas CRISPR-Cas , Ratones SCID , Células Madre Hematopoyéticas
9.
Cell ; 184(16): 4268-4283.e20, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34233163

RESUMEN

Ultraviolet (UV) light and incompletely understood genetic and epigenetic variations determine skin color. Here we describe an UV- and microphthalmia-associated transcription factor (MITF)-independent mechanism of skin pigmentation. Targeting the mitochondrial redox-regulating enzyme nicotinamide nucleotide transhydrogenase (NNT) resulted in cellular redox changes that affect tyrosinase degradation. These changes regulate melanosome maturation and, consequently, eumelanin levels and pigmentation. Topical application of small-molecule inhibitors yielded skin darkening in human skin, and mice with decreased NNT function displayed increased pigmentation. Additionally, genetic modification of NNT in zebrafish alters melanocytic pigmentation. Analysis of four diverse human cohorts revealed significant associations of skin color, tanning, and sun protection use with various single-nucleotide polymorphisms within NNT. NNT levels were independent of UVB irradiation and redox modulation. Individuals with postinflammatory hyperpigmentation or lentigines displayed decreased skin NNT levels, suggesting an NNT-driven, redox-dependent pigmentation mechanism that can be targeted with NNT-modifying topical drugs for medical and cosmetic purposes.


Asunto(s)
Factor de Transcripción Asociado a Microftalmía/metabolismo , NADP Transhidrogenasas/metabolismo , Pigmentación de la Piel/efectos de la radiación , Rayos Ultravioleta , Animales , Línea Celular , Estudios de Cohortes , AMP Cíclico/metabolismo , Daño del ADN , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Predisposición Genética a la Enfermedad , Humanos , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanosomas/efectos de los fármacos , Melanosomas/metabolismo , Melanosomas/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/metabolismo , NADP Transhidrogenasas/antagonistas & inhibidores , Oxidación-Reducción/efectos de los fármacos , Oxidación-Reducción/efectos de la radiación , Polimorfismo de Nucleótido Simple/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteolisis/efectos de la radiación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Pigmentación de la Piel/efectos de los fármacos , Pigmentación de la Piel/genética , Ubiquitina/metabolismo , Pez Cebra
10.
Blood ; 138(26): 2768-2780, 2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-34086870

RESUMEN

XMEN disease, defined as "X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus infection and N-linked glycosylation defect," is a recently described primary immunodeficiency marked by defective T cells and natural killer (NK) cells. Unfortunately, a potentially curative hematopoietic stem cell transplantation is associated with high mortality rates. We sought to develop an ex vivo targeted gene therapy approach for patients with XMEN using a CRISPR/Cas9 adeno-associated vector (AAV) to insert a therapeutic MAGT1 gene at the constitutive locus under the regulation of the endogenous promoter. Clinical translation of CRISPR/Cas9 AAV-targeted gene editing (GE) is hampered by low engraftable gene-edited hematopoietic stem and progenitor cells (HSPCs). Here, we optimized GE conditions by transient enhancement of homology-directed repair while suppressing AAV-associated DNA damage response to achieve highly efficient (>60%) genetic correction in engrafting XMEN HSPCs in transplanted mice. Restored MAGT1 glycosylation function in human NK and CD8+ T cells restored NK group 2 member D (NKG2D) expression and function in XMEN lymphocytes for potential treatment of infections, and it corrected HSPCs for long-term gene therapy, thus offering 2 efficient therapeutic options for XMEN poised for clinical translation.


Asunto(s)
Proteínas de Transporte de Catión/genética , Edición Génica , Células Madre Hematopoyéticas/metabolismo , Linfocitos/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Animales , Sistemas CRISPR-Cas , Proteínas de Transporte de Catión/deficiencia , Células Cultivadas , Femenino , Edición Génica/métodos , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Humanos , Linfocitos/patología , Masculino , Ratones Endogámicos NOD , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/patología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
11.
Blood ; 137(19): 2598-2608, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33623984

RESUMEN

Lentivector gene therapy for X-linked chronic granulomatous disease (X-CGD) has proven to be a viable approach, but random vector integration and subnormal protein production from exogenous promoters in transduced cells remain concerning for long-term safety and efficacy. A previous genome editing-based approach using Streptococcus pyogenes Cas9 mRNA and an oligodeoxynucleotide donor to repair genetic mutations showed the capability to restore physiological protein expression but lacked sufficient efficiency in quiescent CD34+ hematopoietic cells for clinical translation. Here, we report that transient inhibition of p53-binding protein 1 (53BP1) significantly increased (2.3-fold) long-term homology-directed repair to achieve highly efficient (80% gp91phox+ cells compared with healthy donor control subjects) long-term correction of X-CGD CD34+ cells.


Asunto(s)
Reparación del ADN , Edición Génica/métodos , Terapia Genética/métodos , Enfermedad Granulomatosa Crónica/terapia , Trasplante de Células Madre Hematopoyéticas , NADPH Oxidasa 2/genética , Proteína 1 de Unión al Supresor Tumoral P53/antagonistas & inhibidores , Animales , Proteínas Bacterianas , Caspasa 9 , Células Cultivadas , Reparación del ADN/genética , Dependovirus/genética , Exones/genética , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Enfermedad Granulomatosa Crónica/genética , Células Madre Hematopoyéticas/enzimología , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , NADPH Oxidasa 2/deficiencia , Fagocitos/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , Especies Reactivas de Oxígeno , Ribonucleoproteínas/genética , Eliminación de Secuencia , Streptococcus pyogenes/enzimología
12.
Nat Biotechnol ; 39(1): 94-104, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32661438

RESUMEN

Cas12a RNA-guided endonucleases are promising tools for multiplexed genetic perturbations because they can process multiple guide RNAs expressed as a single transcript, and subsequently cleave target DNA. However, their widespread adoption has lagged behind Cas9-based strategies due to low activity and the lack of a well-validated pooled screening toolkit. In the present study, we describe the optimization of enhanced Cas12a from Acidaminococcus (enAsCas12a) for pooled, combinatorial genetic screens in human cells. By assaying the activity of thousands of guides, we refine on-target design rules and develop a comprehensive set of off-target rules to predict and exclude promiscuous guides. We also identify 38 direct repeat variants that can substitute for the wild-type sequence. We validate our optimized AsCas12a toolkit by screening for synthetic lethalities in OVCAR8 and A375 cancer cells, discovering an interaction between MARCH5 and WSB2. Finally, we show that enAsCas12a delivers similar performance to Cas9 in genome-wide dropout screens but at greatly reduced library size, which will facilitate screens in challenging models.


Asunto(s)
Proteínas Bacterianas , Proteínas Asociadas a CRISPR , Sistemas CRISPR-Cas/genética , Endodesoxirribonucleasas , Edición Génica/métodos , ARN Guía de Kinetoplastida , Acidaminococcus/genética , Apoptosis/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Línea Celular Tumoral , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Biblioteca de Genes , Células HEK293 , Humanos , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo
13.
J Immunother Cancer ; 8(2)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32900862

RESUMEN

BACKGROUND: Adoptive cell therapy with chimeric antigen receptor T cells (CAR-T) has become a standard treatment for patients with certain aggressive B cell malignancies and holds promise to improve the care of patients suffering from numerous other cancers in the future. However, the high manufacturing cost of CAR-T cell therapies poses a major barrier to their broader clinical application. Among the key cost drivers of CAR-T production are single-use reagents for T cell activation and clinical-grade viral vector. The presence of variable amounts of contaminating monocytes in the starting material poses an additional challenge to CAR-T manufacturing, since they can impede T cell stimulation and transduction, resulting in manufacturing failure. METHODS: We created K562-based artificial antigen-presenting cells (aAPC) with genetically encoded T cell stimulation and costimulation that represent an inexhaustible source for T cell activation. We additionally disrupted endogenous expression of the low-density lipoprotein receptor (LDLR) on these aAPC (aAPC-ΔLDLR) using CRISPR-Cas9 gene editing nucleases to prevent inadvertent lentiviral transduction and avoid the sink effect on viral vector during transduction. Using various T cell sources, we produced CD19-directed CAR-T cells via aAPC-ΔLDLR-based activation and tested their in vitro and in vivo antitumor potency against B cell malignancies. RESULTS: We found that lack of LDLR expression on our aAPC-ΔLDLR conferred resistance to lentiviral transduction during CAR-T production. Using aAPC-ΔLDLR, we achieved efficient expansion of CAR-T cells even from unpurified starting material like peripheral blood mononuclear cells or unmanipulated leukapheresis product, containing substantial proportions of monocytes. CD19-directed CAR-T cells that we produced via aAPC-ΔLDLR-based expansion demonstrated potent antitumor responses in preclinical models of acute lymphoblastic leukemia and B-cell lymphoma. CONCLUSIONS: Our aAPC-ΔLDLR represent an attractive approach for manufacturing of lentivirally transduced T cells that may be simpler and more cost efficient than currently available methods.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Lentivirus/genética , Transducción Genética/métodos , Humanos
14.
Sci Rep ; 10(1): 4544, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32161326

RESUMEN

Ex-vivo gene therapy using stem cells or T cells transduced by retroviral or lentiviral vectors has shown remarkable efficacy in the treatment of immunodeficiencies and cancer. However, the process is expensive, technically challenging, and not readily scalable to large patient populations, particularly in underdeveloped parts of the world. Direct in vivo gene therapy would avoid these issues, and such approaches with adeno-associated virus (AAV) vectors have been shown to be safe and efficacious in clinical trials for diseases affecting differentiated tissues such as the liver and CNS. However, the ability to transduce lymphocytes with AAV in vivo after systemic delivery has not been carefully explored. Here, we show that both standard and exosome-associated preparations of AAV8 vectors can effectively transduce a variety of immune cell populations including CD4+ T cells, CD8+ T cells, B cells, macrophages, and dendritic cells after systemic delivery in mice. We provide direct evidence of T cell transduction through the detection of AAV genomes and transgene mRNA, and show that intracellular and transmembrane proteins can be expressed. These findings establish the feasibility of AAV-mediated in vivo gene delivery to immune cells which will facilitate both basic and applied research towards the goal of direct in vivo gene immunotherapies.


Asunto(s)
Dependovirus/genética , Exosomas/genética , Técnicas de Transferencia de Gen , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Linfocitos T/metabolismo , Animales , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/citología , Linfocitos T/virología , Transducción Genética , Transgenes
15.
Nat Commun ; 10(1): 4439, 2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31570731

RESUMEN

Adeno-associated virus (AAV) vectors have shown promising results in preclinical models, but the genomic consequences of transduction with AAV vectors encoding CRISPR-Cas nucleases is still being examined. In this study, we observe high levels of AAV integration (up to 47%) into Cas9-induced double-strand breaks (DSBs) in therapeutically relevant genes in cultured murine neurons, mouse brain, muscle and cochlea. Genome-wide AAV mapping in mouse brain shows no overall increase of AAV integration except at the CRISPR/Cas9 target site. To allow detailed characterization of integration events we engineer a miniature AAV encoding a 465 bp lambda bacteriophage DNA (AAV-λ465), enabling sequencing of the entire integrated vector genome. The integration profile of AAV-465λ in cultured cells display both full-length and fragmented AAV genomes at Cas9 on-target sites. Our data indicate that AAV integration should be recognized as a common outcome for applications that utilize AAV for genome editing.


Asunto(s)
Sistemas CRISPR-Cas , Roturas del ADN , Dependovirus/genética , Edición Génica/métodos , Vectores Genéticos , Integración Viral/genética , Animales , Bacteriófago lambda/genética , Encéfalo , Línea Celular , Mapeo Cromosómico , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Cóclea , Endonucleasas , Marcación de Gen/métodos , Terapia Genética/métodos , Genoma , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculos , Neuronas/virología , Reparación del Gen Blanco/métodos , Resultado del Tratamiento
16.
Nat Med ; 25(7): 1123-1130, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270503

RESUMEN

Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.


Asunto(s)
Alelos , Edición Génica , Pérdida Auditiva Sensorineural/prevención & control , Proteínas de la Membrana/genética , Animales , Proteína 9 Asociada a CRISPR/fisiología , Línea Celular , Células Cultivadas , Dependovirus/genética , Modelos Animales de Enfermedad , Pérdida Auditiva Sensorineural/genética , Humanos , Ratones , Ratones Endogámicos C57BL
17.
Mol Ther Nucleic Acids ; 11: 429-440, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29858078

RESUMEN

The APPswe (Swedish) mutation in the amyloid precursor protein (APP) gene causes dominantly inherited Alzheimer's disease (AD) as a result of increased ß-secretase cleavage of the amyloid-ß (Aß) precursor protein. This leads to abnormally high Aß levels, not only in brain but also in peripheral tissues of mutation carriers. Here, we selectively disrupted the human mutant APPSW allele using CRISPR. By applying CRISPR/Cas9 from Streptococcus pyogenes, we generated allele-specific deletions of either APPSW or APPWT. As measured by ELISA, conditioned media of targeted patient-derived fibroblasts displayed an approximate 60% reduction in secreted Aß. Next, coding sequences for the APPSW-specific guide RNA (gRNA) and Cas9 were packaged into separate adeno-associated viral (AAV) vectors. Site-specific indel formation was achieved both in primary neurons isolated from APPSW transgenic mouse embryos (Tg2576) and after co-injection of these vectors into hippocampus of adult mice. Taken together, we here present proof-of-concept data that CRISPR/Cas9 can selectively disrupt the APPSW allele both ex vivo and in vivo-and thereby decrease pathogenic Aß. Hence, this system may have the potential to be developed as a tool for gene therapy against AD caused by APPswe and other point mutations associated with increased Aß.

18.
Arterioscler Thromb Vasc Biol ; 38(7): 1562-1575, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29724820

RESUMEN

OBJECTIVE: Tie1 (tyrosine kinase containing immunoglobulin and epidermal growth factor homology 1), an endothelial and hematopoietic cell-specific receptor tyrosine kinase, is an important regulator of angiogenesis and critical for maintaining vascular integrity. The post-transcriptional regulation of tie1 mRNA expression is not understood, but it might partly explain Tie1's differential expression pattern in endothelium. Following up on our previous work that identified natural antisense transcripts from the tie1 locus-tie1 antisense (tie1AS), which regulates tie1 mRNA levels in zebrafish-we attempted to identify the mechanism of this regulation. APPROACH AND RESULTS: Through in vitro and in vivo ribonucleoprotein binding studies, we demonstrated that tie1AS long noncoding RNA interacts with an RNA binding protein-embryonic lethal and abnormal vision Drosophila-like 1 (Elavl1)-that regulates tie1 mRNA levels. When we disrupted the interaction between tie1AS and Elavl1 by using constitutively active antisense morpholino oligonucleotides or photoactivatable morpholino oligonucleotides, tie1 mRNA levels increased between 26 and 31 hours post-fertilization, particularly in the head. This increase correlated with dilation of primordial midbrain channels, smaller eyes, and reduced ventricular space. We also observed these phenotypes when we used CRISPR (clustered regularly interspaced short palindromic repeats)-mediated CRISPRi (CRISPR-mediated interference) to knock down tie1AS. Treatment of the morpholino oligonucleotide-injected embryos with a small molecule that decreased tie1 mRNA levels rescued all 3 abnormal phenotypes. CONCLUSIONS: We identified a novel mode of temporal and spatial post-transcriptional regulation of tie1 mRNA. It involves long noncoding RNA, tie1AS, and Elavl1 (an interactor of tie1AS).


Asunto(s)
Vasos Sanguíneos/enzimología , Encéfalo/irrigación sanguínea , Neovascularización Fisiológica/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/embriología , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , ARN sin Sentido/genética , ARN sin Sentido/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Receptor TIE-1/genética , Receptor TIE-1/metabolismo , Factores de Tiempo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
19.
Nat Methods ; 14(12): 1163-1166, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29083402

RESUMEN

Targeted and inducible regulation of mammalian gene expression is a broadly important capability. We engineered drug-inducible catalytically inactive Cpf1 nuclease fused to transcriptional activation domains to tune the expression of endogenous genes in human cells. Leveraging the multiplex capability of the Cpf1 platform, we demonstrate both synergistic and combinatorial gene expression in human cells. Our work should enable the development of multiplex gene perturbation library screens for understanding complex cellular phenotypes.


Asunto(s)
Proteínas Bacterianas/genética , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Endonucleasas/genética , Activación Transcripcional , Técnicas de Cultivo de Célula , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Proteína Vmw65 de Virus del Herpes Simple/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Plásmidos , Proteínas Recombinantes de Fusión/genética , Transactivadores/genética , Factor de Transcripción ReIA/genética , Transfección
20.
Oncotarget ; 8(27): 43733-43751, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28415827

RESUMEN

Proteasomal degradation of topoisomerase I (topoI) is one of the most remarkable cellular phenomena observed in response to camptothecin (CPT). Importantly, the rate of topoI degradation is linked to CPT resistance. Formation of the topoI-DNA-CPT cleavable complex inhibits DNA re-ligation resulting in DNA-double strand break (DSB). The degradation of topoI marks the first step in the ubiquitin proteasome pathway (UPP) dependent DNA damage response (DDR). Here, we show that the Ku70/Ku80 heterodimer binds with topoI, and that the DNA-dependent protein kinase (DNA-PKcs) phosphorylates topoI on serine 10 (topoI-pS10), which is subsequently ubiquitinated by BRCA1. A higher basal level of topoI-pS10 ensures rapid topoI degradation leading to CPT resistance. Importantly, PTEN regulates DNA-PKcs kinase activity in this pathway and PTEN deletion ensures DNA-PKcs dependent higher topoI-pS10, rapid topoI degradation and CPT resistance.


Asunto(s)
Camptotecina/farmacología , ADN-Topoisomerasas de Tipo I/metabolismo , Resistencia a Antineoplásicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Ubiquitina/metabolismo , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Edición Génica , Humanos , Autoantígeno Ku/metabolismo , Complejos Multiproteicos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Unión Proteica , Proteína Quinasa C/metabolismo , Proteolisis , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA