Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
bioRxiv ; 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39071446

RESUMEN

Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) is a new approach for ex vivo genome editing of primary human cells. PERC uses a single amphiphilic peptide reagent to mediate intracellular delivery of the same pre-formed CRISPR ribonucleoprotein enzymes that are broadly used in research and therapeutics, resulting in high-efficiency editing of stimulated immune cells and cultured hematopoietic stem and progenitor cells (HSPCs). PERC facilitates nuclease-mediated gene knockout, precise transgene knock-in, and base editing. PERC involves mixing the CRISPR ribonucleoprotein enzyme with peptide and then incubating the formulation with cultured cells. For efficient transgene knock-in, adeno-associated virus (AAV) bearing homology-directed repair template DNA may be included. In contrast to electroporation, PERC is appealing as it requires no dedicated hardware and has less impact on cell phenotype and viability. Due to the gentle nature of PERC, delivery can be performed multiple times without substantial impact to cell health or phenotype. Here we report methods for improved PERC-mediated editing of T cells as well as novel methods for PERC-mediated editing of HSPCs, including knockout and precise knock-in. Editing efficiencies can surpass 90% using either Cas9 or Cas12a in primary T cells or HSPCs. Because PERC calls for only three readily available reagents - protein, RNA, and peptide - and does not require dedicated hardware for any step, PERC demands no special expertise and is exceptionally straightforward to adopt. The inherent compatibility of PERC with established cell engineering pipelines makes this approach appealing for rapid deployment in research and clinical settings.

2.
Nat Biomed Eng ; 7(5): 647-660, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37147433

RESUMEN

CRISPR-mediated genome editing of primary human lymphocytes is typically carried out via electroporation, which can be cytotoxic, cumbersome and costly. Here we show that the yields of edited primary human lymphocytes can be increased substantially by delivering a CRISPR ribonucleoprotein mixed with an amphiphilic peptide identified through screening. We evaluated the performance of this simple delivery method by knocking out genes in T cells, B cells and natural killer cells via the delivery of Cas9 or Cas12a ribonucleoproteins or an adenine base editor. We also show that peptide-mediated ribonucleoprotein delivery paired with an adeno-associated-virus-mediated homology-directed repair template can introduce a chimaeric antigen receptor gene at the T-cell receptor α constant locus, and that the engineered cells display antitumour potency in mice. The method is minimally perturbative, does not require dedicated hardware, and is compatible with multiplexed editing via sequential delivery, which minimizes the risk of genotoxicity. The peptide-mediated intracellular delivery of ribonucleoproteins may facilitate the manufacturing of engineered T cells.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Humanos , Ratones , Animales , Edición Génica/métodos , Linfocitos T/metabolismo , Péptidos/genética , Ribonucleoproteínas
3.
Sci Immunol ; 6(60)2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34088745

RESUMEN

P2RY8 promotes the confinement and growth regulation of germinal center (GC) B cells, and loss of human P2RY8 is associated with B cell lymphomagenesis. The metabolite S-geranylgeranyl-l-glutathione (GGG) is a P2RY8 ligand. The mechanisms controlling GGG distribution are poorly understood. Here, we show that gamma-glutamyltransferase-5 (Ggt5) expression in stromal cells was required for GGG catabolism and confinement of P2RY8-expressing cells to GCs. We identified the ATP-binding cassette subfamily C member 1 (Abcc1) as a GGG transporter and showed that Abcc1 expression by hematopoietic cells was necessary for P2RY8-mediated GC confinement. Furthermore, we discovered that P2RY8 and GGG negatively regulated trafficking of B and T cells to the bone marrow (BM). P2RY8 loss-of-function human T cells increased their BM homing. By defining how GGG distribution was determined and identifying sites of P2RY8 activity, this work helps establish how disruptions in P2RY8 function contribute to lymphomagenesis and other disease states.


Asunto(s)
Glutatión , Linfocitos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Receptores Purinérgicos P2Y , gamma-Glutamiltransferasa , Animales , Femenino , Humanos , Masculino , Ratones , gamma-Glutamiltransferasa/genética , gamma-Glutamiltransferasa/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Glutatión/metabolismo , Células HEK293 , Activación de Linfocitos , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Receptores Purinérgicos P2Y/genética , Receptores Purinérgicos P2Y/metabolismo
4.
Cell Rep ; 35(9): 109207, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34077734

RESUMEN

As genome engineering advances cell-based therapies, a versatile approach to introducing both CRISPR-Cas9 ribonucleoproteins (RNPs) and therapeutic transgenes into specific cells would be transformative. Autologous T cells expressing a chimeric antigen receptor (CAR) manufactured by viral transduction are approved to treat multiple blood cancers, but additional genetic modifications to alter cell programs will likely be required to treat solid tumors and for allogeneic cellular therapies. We have developed a one-step strategy using engineered lentiviral particles to introduce Cas9 RNPs and a CAR transgene into primary human T cells without electroporation. Furthermore, programming particle tropism allows us to target a specific cell type within a mixed cell population. As a proof-of-concept, we show that HIV-1 envelope targeted particles to edit CD4+ cells while sparing co-cultured CD8+ cells. This adaptable approach to immune cell engineering ex vivo provides a strategy applicable to the genetic modification of targeted somatic cells in vivo.


Asunto(s)
Sistemas CRISPR-Cas/genética , Ingeniería Celular , Técnicas de Transferencia de Gen , Transgenes , Células A549 , Linfocitos T CD4-Positivos/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Edición Génica , VIH-1/fisiología , Humanos , Células Jurkat , Lentivirus/genética , Receptores Quiméricos de Antígenos/metabolismo , Ribonucleoproteínas/metabolismo , Virión/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana
5.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33951726

RESUMEN

The pioneer transcription factor (TF) PU.1 controls hematopoietic cell fate by decompacting stem cell heterochromatin and allowing nonpioneer TFs to enter otherwise inaccessible genomic sites. PU.1 deficiency fatally arrests lymphopoiesis and myelopoiesis in mice, but human congenital PU.1 disorders have not previously been described. We studied six unrelated agammaglobulinemic patients, each harboring a heterozygous mutation (four de novo, two unphased) of SPI1, the gene encoding PU.1. Affected patients lacked circulating B cells and possessed few conventional dendritic cells. Introducing disease-similar SPI1 mutations into human hematopoietic stem and progenitor cells impaired early in vitro B cell and myeloid cell differentiation. Patient SPI1 mutations encoded destabilized PU.1 proteins unable to nuclear localize or bind target DNA. In PU.1-haploinsufficient pro-B cell lines, euchromatin was less accessible to nonpioneer TFs critical for B cell development, and gene expression patterns associated with the pro- to pre-B cell transition were undermined. Our findings molecularly describe a novel form of agammaglobulinemia and underscore PU.1's critical, dose-dependent role as a hematopoietic euchromatin gatekeeper.


Asunto(s)
Agammaglobulinemia/genética , Cromatina/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Adolescente , Adulto , Linfocitos B/fisiología , Diferenciación Celular/genética , Línea Celular , Niño , Preescolar , Células Dendríticas/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Humanos , Lactante , Linfopoyesis/genética , Masculino , Mutación/genética , Células Precursoras de Linfocitos B/fisiología , Células Madre/fisiología , Adulto Joven
6.
Cell ; 181(3): 728-744.e21, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32302591

RESUMEN

Adoptive transfer of genetically modified immune cells holds great promise for cancer immunotherapy. CRISPR knockin targeting can improve cell therapies, but more high-throughput methods are needed to test which knockin gene constructs most potently enhance primary cell functions in vivo. We developed a widely adaptable technology to barcode and track targeted integrations of large non-viral DNA templates and applied it to perform pooled knockin screens in primary human T cells. Pooled knockin of dozens of unique barcoded templates into the T cell receptor (TCR)-locus revealed gene constructs that enhanced fitness in vitro and in vivo. We further developed pooled knockin sequencing (PoKI-seq), combining single-cell transcriptome analysis and pooled knockin screening to measure cell abundance and cell state ex vivo and in vivo. This platform nominated a novel transforming growth factor ß (TGF-ß) R2-41BB chimeric receptor that improved solid tumor clearance. Pooled knockin screening enables parallelized re-writing of endogenous genetic sequences to accelerate discovery of knockin programs for cell therapies.


Asunto(s)
Técnicas de Sustitución del Gen/métodos , Ingeniería Genética/métodos , Inmunoterapia/métodos , Animales , Células Sanguíneas , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , ARN Guía de Kinetoplastida/genética , Análisis de la Célula Individual/métodos , Linfocitos T , Transcriptoma/genética
7.
Nat Biotechnol ; 38(1): 44-49, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819258

RESUMEN

Versatile and precise genome modifications are needed to create a wider range of adoptive cellular therapies1-5. Here we report two improvements that increase the efficiency of CRISPR-Cas9-based genome editing in clinically relevant primary cell types. Truncated Cas9 target sequences (tCTSs) added at the ends of the homology-directed repair (HDR) template interact with Cas9 ribonucleoproteins (RNPs) to shuttle the template to the nucleus, enhancing HDR efficiency approximately two- to fourfold. Furthermore, stabilizing Cas9 RNPs into nanoparticles with polyglutamic acid further improves editing efficiency by approximately twofold, reduces toxicity, and enables lyophilized storage without loss of activity. Combining the two improvements increases gene targeting efficiency even at reduced HDR template doses, yielding approximately two to six times as many viable edited cells across multiple genomic loci in diverse cell types, such as bulk (CD3+) T cells, CD8+ T cells, CD4+ T cells, regulatory T cells (Tregs), γδ T cells, B cells, natural killer cells, and primary and induced pluripotent stem cell-derived6 hematopoietic stem progenitor cells (HSPCs).


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Polímeros/química , Adulto , Edición Génica , Humanos , Nanopartículas/química , Estabilidad Proteica , ARN Guía de Kinetoplastida/metabolismo
8.
JCI Insight ; 4(24)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31852848

RESUMEN

Distinct subsets of Tregs reside in nonlymphoid tissues where they mediate unique functions. To interrogate the biology of tissue Tregs in human health and disease, we phenotypically and functionally compared healthy skin Tregs with those in peripheral blood, inflamed psoriatic skin, and metastatic melanoma. The mitochondrial enzyme, arginase 2 (ARG2), was preferentially expressed in Tregs in healthy skin, increased in Tregs in metastatic melanoma, and reduced in Tregs from psoriatic skin. ARG2 enhanced Treg suppressive capacity in vitro and conferred a selective advantage for accumulation in inflamed tissues in vivo. CRISPR-mediated deletion of this gene in primary human Tregs was sufficient to skew away from a tissue Treg transcriptional signature. Notably, the inhibition of ARG2 increased mTOR signaling, whereas the overexpression of this enzyme suppressed it. Taken together, our results suggest that Tregs express ARG2 in human tissues to both regulate inflammation and enhance their metabolic fitness.


Asunto(s)
Arginasa/metabolismo , Piel/patología , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Arginasa/genética , Células Cultivadas , Células Dendríticas , Técnicas de Inactivación de Genes , Humanos , Queratinocitos , Masculino , Melanoma/inmunología , Melanoma/patología , Ratones , Persona de Mediana Edad , Cultivo Primario de Células , Psoriasis/inmunología , Psoriasis/patología , RNA-Seq , Transducción de Señal/inmunología , Piel/citología , Piel/inmunología , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
9.
Nature ; 559(7714): 405-409, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29995861

RESUMEN

Decades of work have aimed to genetically reprogram T cells for therapeutic purposes1,2 using recombinant viral vectors, which do not target transgenes to specific genomic sites3,4. The need for viral vectors has slowed down research and clinical use as their manufacturing and testing is lengthy and expensive. Genome editing brought the promise of specific and efficient insertion of large transgenes into target cells using homology-directed repair5,6. Here we developed a CRISPR-Cas9 genome-targeting system that does not require viral vectors, allowing rapid and efficient insertion of large DNA sequences (greater than one kilobase) at specific sites in the genomes of primary human T cells, while preserving cell viability and function. This permits individual or multiplexed modification of endogenous genes. First, we applied this strategy to correct a pathogenic IL2RA mutation in cells from patients with monogenic autoimmune disease, and demonstrate improved signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR that redirected T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized tumour antigens and mounted productive anti-tumour cell responses in vitro and in vivo. Together, these studies provide preclinical evidence that non-viral genome targeting can enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.


Asunto(s)
Reprogramación Celular/genética , Edición Génica , Genoma Humano/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Autoinmunidad/genética , Sistemas CRISPR-Cas/genética , Células Cultivadas , Humanos , Subunidad alfa del Receptor de Interleucina-2/genética , Masculino , Ratones , Trasplante de Neoplasias , Ingeniería de Proteínas , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/citología
10.
Adv Mater ; 21(48): 4947-4951, 2009 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-25165411

RESUMEN

End-modified polymers are promising for the nonviral delivery of genes to cancer cells, immune cells, and human stem cells and point to polymer end-groups as regulators for cell-type specificity. A library of polymers has been synthesized and, although some polymers are strong transfection agents overall, for each cell type, a particular polymer is most effective.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA