Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nature ; 539(7629): 384-389, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27820943

RESUMEN

The ß-haemoglobinopathies, such as sickle cell disease and ß-thalassaemia, are caused by mutations in the ß-globin (HBB) gene and affect millions of people worldwide. Ex vivo gene correction in patient-derived haematopoietic stem cells followed by autologous transplantation could be used to cure ß-haemoglobinopathies. Here we present a CRISPR/Cas9 gene-editing system that combines Cas9 ribonucleoproteins and adeno-associated viral vector delivery of a homologous donor to achieve homologous recombination at the HBB gene in haematopoietic stem cells. Notably, we devise an enrichment model to purify a population of haematopoietic stem and progenitor cells with more than 90% targeted integration. We also show efficient correction of the Glu6Val mutation responsible for sickle cell disease by using patient-derived stem and progenitor cells that, after differentiation into erythrocytes, express adult ß-globin (HbA) messenger RNA, which confirms intact transcriptional regulation of edited HBB alleles. Collectively, these preclinical studies outline a CRISPR-based methodology for targeting haematopoietic stem cells by homologous recombination at the HBB locus to advance the development of next-generation therapies for ß-haemoglobinopathies.


Asunto(s)
Anemia de Células Falciformes/genética , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Marcación de Gen , Terapia Genética/métodos , Células Madre Hematopoyéticas/metabolismo , Globinas beta/genética , Alelos , Anemia de Células Falciformes/patología , Anemia de Células Falciformes/terapia , Animales , Antígenos CD34/metabolismo , Proteínas Asociadas a CRISPR/metabolismo , Diferenciación Celular , Linaje de la Célula , Separación Celular , Dependovirus/genética , Eritrocitos , Femenino , Citometría de Flujo , Genes Reporteros , Recombinación Homóloga , Humanos , Imanes , Ratones Endogámicos NOD , Ratones SCID , Microesferas , Mutación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Talasemia beta/genética , Talasemia beta/terapia
2.
Curr Cardiol Rep ; 23(8): 103, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34196831

RESUMEN

PURPOSE OF REVIEW: Recent technological advances have led to an increased ability to define the gene expression profile of the cardiac conduction system (CCS). Here, we review the most salient studies to emerge in recent years and discuss existing gaps in our knowledge as well as future areas of investigation. RECENT FINDINGS: Molecular profiling of the CCS spans several decades. However, the advent of high-throughput sequencing strategies has allowed for the discovery of unique transcriptional programs of the many diverse CCS cell types. The CCS, a diverse structure with significant inter- and intra-component cellular heterogeneity, is essential to the normal function of the heart. Progress in transcriptomic profiling has improved the resolution and depth of characterization of these unique and clinically relevant CCS cell types. Future studies leveraging this big data will play a crucial role in improving our understanding of CCS development and function as well as translating these findings into tangible translational tools for the improved detection, prevention, and treatment of cardiac arrhythmias.


Asunto(s)
Arritmias Cardíacas , Sistema de Conducción Cardíaco , Arritmias Cardíacas/genética , Perfilación de la Expresión Génica , Corazón , Humanos , Transcriptoma
3.
J Clin Invest ; 132(20)2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-35951416

RESUMEN

Accidental injury to the cardiac conduction system (CCS), a network of specialized cells embedded within the heart and indistinguishable from the surrounding heart muscle tissue, is a major complication in cardiac surgeries. Here, we addressed this unmet need by engineering targeted antibody-dye conjugates directed against the CCS, allowing for the visualization of the CCS in vivo following a single intravenous injection in mice. These optical imaging tools showed high sensitivity, specificity, and resolution, with no adverse effects on CCS function. Further, with the goal of creating a viable prototype for human use, we generated a fully human monoclonal Fab that similarly targets the CCS with high specificity. We demonstrate that, when conjugated to an alternative cargo, this Fab can also be used to modulate CCS biology in vivo, providing a proof of principle for targeted cardiac therapeutics. Finally, in performing differential gene expression analyses of the entire murine CCS at single-cell resolution, we uncovered and validated a suite of additional cell surface markers that can be used to molecularly target the distinct subcomponents of the CCS, each prone to distinct life-threatening arrhythmias. These findings lay the foundation for translational approaches targeting the CCS for visualization and therapy in cardiothoracic surgery, cardiac imaging, and arrhythmia management.


Asunto(s)
Sistema de Conducción Cardíaco , Terapia Molecular Dirigida , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Corazón/fisiología , Sistema de Conducción Cardíaco/metabolismo , Humanos , Ratones , Miocardio
4.
Placenta ; 103: 50-52, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33075720

RESUMEN

Umbilical cord blood is an important graft source in the treatment of many genetic, hematologic, and immunologic disorders by hematopoietic stem cell transplantation. Millions of cord blood units have been collected and stored for clinical use since the inception of cord blood banking in 1989. However, the use of cord blood in biomedical research has been limited by access to viable samples. Here, we present a cost-effective, self-sustaining model for the procurement of fresh umbilical cord blood components for research purposes within hospital-affiliated academic institutions.


Asunto(s)
Investigación Biomédica/organización & administración , Bancos de Sangre/organización & administración , Sangre Fetal , Modelos Organizacionales , Academias e Institutos/economía , Academias e Institutos/organización & administración , Academias e Institutos/normas , Investigación Biomédica/economía , Investigación Biomédica/métodos , Investigación Biomédica/normas , Bancos de Sangre/economía , Bancos de Sangre/normas , Recolección de Muestras de Sangre/economía , Recolección de Muestras de Sangre/métodos , Recolección de Muestras de Sangre/normas , California , Análisis Costo-Beneficio , Femenino , Sangre Fetal/citología , Sangre Fetal/trasplante , Trasplante de Células Madre Hematopoyéticas/economía , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre Hematopoyéticas/normas , Humanos , Recién Nacido , Embarazo
5.
Sci Transl Med ; 13(598)2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135108

RESUMEN

Sickle cell disease (SCD) is the most common serious monogenic disease with 300,000 births annually worldwide. SCD is an autosomal recessive disease resulting from a single point mutation in codon six of the ß-globin gene (HBB). Ex vivo ß-globin gene correction in autologous patient-derived hematopoietic stem and progenitor cells (HSPCs) may potentially provide a curative treatment for SCD. We previously developed a CRISPR-Cas9 gene targeting strategy that uses high-fidelity Cas9 precomplexed with chemically modified guide RNAs to induce recombinant adeno-associated virus serotype 6 (rAAV6)-mediated HBB gene correction of the SCD-causing mutation in HSPCs. Here, we demonstrate the preclinical feasibility, efficacy, and toxicology of HBB gene correction in plerixafor-mobilized CD34+ cells from healthy and SCD patient donors (gcHBB-SCD). We achieved up to 60% HBB allelic correction in clinical-scale gcHBB-SCD manufacturing. After transplant into immunodeficient NSG mice, 20% gene correction was achieved with multilineage engraftment. The long-term safety, tumorigenicity, and toxicology study demonstrated no evidence of abnormal hematopoiesis, genotoxicity, or tumorigenicity from the engrafted gcHBB-SCD drug product. Together, these preclinical data support the safety, efficacy, and reproducibility of this gene correction strategy for initiation of a phase 1/2 clinical trial in patients with SCD.


Asunto(s)
Anemia de Células Falciformes , Compuestos Heterocíclicos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Sistemas CRISPR-Cas/genética , Edición Génica , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas , Humanos , Ratones , Reproducibilidad de los Resultados , Globinas beta/genética
6.
Nat Commun ; 11(1): 2470, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424320

RESUMEN

Human mesenchymal stromal cells (hMSCs) are a promising source for engineered cell-based therapies in which genetic engineering could enhance therapeutic efficacy and install novel cellular functions. Here, we describe an optimized Cas9-AAV6-based genome editing tool platform for site-specific mutagenesis and integration of up to more than 3 kilobases of exogenous DNA in the genome of hMSCs derived from the bone marrow, adipose tissue, and umbilical cord blood without altering their ex vivo characteristics. We generate safe harbor-integrated lines of engineered hMSCs and show that engineered luciferase-expressing hMSCs are transiently active in vivo in wound beds of db/db mice. Moreover, we generate PDGF-BB- and VEGFA-hypersecreting hMSC lines as short-term, local wound healing agents with superior therapeutic efficacy over wildtype hMSCs in the diabetic mouse model without replacing resident cells long-term. This study establishes a precise genetic engineering platform for genetic studies of hMSCs and development of engineered hMSC-based therapies.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Piel/patología , Cicatrización de Heridas , Animales , Proliferación Celular , Supervivencia Celular , Reactivos de Enlaces Cruzados/química , Dependovirus , Edición Génica , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hidrogeles/química , Cinética , Ratones , Proteínas Proto-Oncogénicas c-sis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nat Commun ; 10(1): 4045, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492863

RESUMEN

Lysosomal enzyme deficiencies comprise a large group of genetic disorders that generally lack effective treatments. A potential treatment approach is to engineer the patient's own hematopoietic system to express high levels of the deficient enzyme, thereby correcting the biochemical defect and halting disease progression. Here, we present an efficient ex vivo genome editing approach using CRISPR-Cas9 that targets the lysosomal enzyme iduronidase to the CCR5 safe harbor locus in human CD34+ hematopoietic stem and progenitor cells. The modified cells secrete supra-endogenous enzyme levels, maintain long-term repopulation and multi-lineage differentiation potential, and can improve biochemical and phenotypic abnormalities in an immunocompromised mouse model of Mucopolysaccharidosis type I. These studies provide support for the development of genome-edited CD34+ hematopoietic stem and progenitor cells as a potential treatment for Mucopolysaccharidosis type I. The safe harbor approach constitutes a flexible platform for the expression of lysosomal enzymes making it applicable to other lysosomal storage disorders.


Asunto(s)
Edición Génica/métodos , Genoma Humano , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Iduronidasa/metabolismo , Mucopolisacaridosis I/terapia , Animales , Antígenos CD34/genética , Antígenos CD34/metabolismo , Sistemas CRISPR-Cas , Terapia Genética/métodos , Humanos , Iduronidasa/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/patología , Células 3T3 NIH , Fenotipo , Receptores CCR5/genética , Receptores CCR5/metabolismo , Trasplante Heterólogo
9.
Nat Commun ; 10(1): 2021, 2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31028274

RESUMEN

The original version of this Article omitted the following from the Acknowledgements: "G.B. acknowledges the support from the Cancer Prevention and Research Institute of Texas (RR140081 and RR170721)."This has now been corrected in both the PDF and HTML versions of the Article.

10.
Nat Med ; 25(2): 249-254, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30692695

RESUMEN

The CRISPR-Cas9 system is a powerful tool for genome editing, which allows the precise modification of specific DNA sequences. Many efforts are underway to use the CRISPR-Cas9 system to therapeutically correct human genetic diseases1-6. The most widely used orthologs of Cas9 are derived from Staphylococcus aureus and Streptococcus pyogenes5,7. Given that these two bacterial species infect the human population at high frequencies8,9, we hypothesized that humans may harbor preexisting adaptive immune responses to the Cas9 orthologs derived from these bacterial species, SaCas9 (S. aureus) and SpCas9 (S. pyogenes). By probing human serum for the presence of anti-Cas9 antibodies using an enzyme-linked immunosorbent assay, we detected antibodies against both SaCas9 and SpCas9 in 78% and 58% of donors, respectively. We also found anti-SaCas9 T cells in 78% and anti-SpCas9 T cells in 67% of donors, which demonstrates a high prevalence of antigen-specific T cells against both orthologs. We confirmed that these T cells were Cas9-specific by demonstrating a Cas9-specific cytokine response following isolation, expansion, and antigen restimulation. Together, these data demonstrate that there are preexisting humoral and cell-mediated adaptive immune responses to Cas9 in humans, a finding that should be taken into account as the CRISPR-Cas9 system moves toward clinical trials.


Asunto(s)
Inmunidad Adaptativa , Proteína 9 Asociada a CRISPR/metabolismo , Adulto , Separación Celular , Femenino , Humanos , Inmunidad Humoral , Masculino , Linfocitos T/inmunología
11.
Nat Commun ; 10(1): 1634, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30967552

RESUMEN

Gene correction in human long-term hematopoietic stem cells (LT-HSCs) could be an effective therapy for monogenic diseases of the blood and immune system. Here we describe an approach for X-linked sSevere cCombined iImmunodeficiency (SCID-X1) using targeted integration of a cDNA into the endogenous start codon to functionally correct disease-causing mutations throughout the gene. Using a CRISPR-Cas9/AAV6 based strategy, we achieve up to 20% targeted integration frequencies in LT-HSCs. As measures of the lack of toxicity we observe no evidence of abnormal hematopoiesis following transplantation and no evidence of off-target mutations using a high-fidelity Cas9 as a ribonucleoprotein complex. We achieve high levels of targeting frequencies (median 45%) in CD34+ HSPCs from six SCID-X1 patients and demonstrate rescue of lymphopoietic defect in a patient derived HSPC population in vitro and in vivo. In sum, our study provides specificity, toxicity and efficacy data supportive of clinical development of genome editing to treat SCID-Xl.


Asunto(s)
ADN Complementario/genética , Edición Génica/métodos , Trasplante de Células Madre Hematopoyéticas , Subunidad gamma Común de Receptores de Interleucina/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Antígenos CD34/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular , Codón Iniciador/genética , Dependovirus , Exones/genética , Sangre Fetal/citología , Vectores Genéticos/genética , Voluntarios Sanos , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Mutación , Parvovirinae/genética , Cultivo Primario de Células , Factores de Tiempo , Transducción Genética/métodos , Quimera por Trasplante/genética , Trasplante Heterólogo/métodos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA