Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 595(7866): 295-302, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34079130

RESUMEN

Sickle cell disease (SCD) is caused by a mutation in the ß-globin gene HBB1. We used a custom adenine base editor (ABE8e-NRCH)2,3 to convert the SCD allele (HBBS) into Makassar ß-globin (HBBG), a non-pathogenic variant4,5. Ex vivo delivery of mRNA encoding the base editor with a targeting guide RNA into haematopoietic stem and progenitor cells (HSPCs) from patients with SCD resulted in 80% conversion of HBBS to HBBG. Sixteen weeks after transplantation of edited human HSPCs into immunodeficient mice, the frequency of HBBG was 68% and hypoxia-induced sickling of bone marrow reticulocytes had decreased fivefold, indicating durable gene editing. To assess the physiological effects of HBBS base editing, we delivered ABE8e-NRCH and guide RNA into HSPCs from a humanized SCD mouse6 and then transplanted these cells into irradiated mice. After sixteen weeks, Makassar ß-globin represented 79% of ß-globin protein in blood, and hypoxia-induced sickling was reduced threefold. Mice that received base-edited HSPCs showed near-normal haematological parameters and reduced splenic pathology compared to mice that received unedited cells. Secondary transplantation of edited bone marrow confirmed that the gene editing was durable in long-term haematopoietic stem cells and showed that HBBS-to-HBBG editing of 20% or more is sufficient for phenotypic rescue. Base editing of human HSPCs avoided the p53 activation and larger deletions that have been observed following Cas9 nuclease treatment. These findings point towards a one-time autologous treatment for SCD that eliminates pathogenic HBBS, generates benign HBBG, and minimizes the undesired consequences of double-strand DNA breaks.


Asunto(s)
Adenina/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Edición Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Globinas beta/genética , Animales , Antígenos CD34/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Genoma Humano/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Humanos , Masculino , Ratones
2.
Cytotherapy ; 25(3): 261-269, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36123234

RESUMEN

Genome editing of hematopoietic stem and progenitor cells is being developed for the treatment of several inherited disorders of the hematopoietic system. The adaptation of CRISPR-Cas9-based technologies to make precise changes to the genome, and developments in altering the specificity and efficiency, and improving the delivery of nucleases to target cells have led to several breakthroughs. Many clinical trials are ongoing, and several pre-clinical models have been reported that would allow these genetic therapies to one day offer a potential cure to patients with diseases where limited options currently exist. However, there remain several challenges with respect to establishing safety, expanding accessibility and improving the manufacturing processes of these therapeutic products. This review focuses on some of the recent advances in the field of genome editing of hematopoietic stem and progenitor cells and illustrates the ongoing challenges.


Asunto(s)
Sistemas CRISPR-Cas , Células Madre Hematopoyéticas , Humanos , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Terapia Genética/métodos
3.
Nat Chem Biol ; 13(4): 415-424, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28192414

RESUMEN

Distinguishing cancer cells from normal cells through surface receptors is vital for cancer diagnosis and targeted therapy. Metabolic glycoengineering of unnatural sugars provides a powerful tool to manually introduce chemical receptors onto the cell surface; however, cancer-selective labeling still remains a great challenge. Herein we report the design of sugars that can selectively label cancer cells both in vitro and in vivo. Specifically, we inhibit the cell-labeling activity of tetraacetyl-N-azidoacetylmannosamine (Ac4ManAz) by converting its anomeric acetyl group to a caged ether bond that can be selectively cleaved by cancer-overexpressed enzymes and thus enables the overexpression of azido groups on the surface of cancer cells. Histone deacetylase and cathepsin L-responsive acetylated azidomannosamine, one such enzymatically activatable Ac4ManAz analog developed, mediated cancer-selective labeling in vivo, which enhanced tumor accumulation of a dibenzocyclooctyne-doxorubicin conjugate via click chemistry and enabled targeted therapy against LS174T colon cancer, MDA-MB-231 triple-negative breast cancer and 4T1 metastatic breast cancer in mice.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carbohidratos/análisis , Carbohidratos/química , Neoplasias del Colon/metabolismo , Sondas Moleculares/análisis , Sondas Moleculares/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias Experimentales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Carbohidratos/síntesis química , Línea Celular Tumoral , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Estructura Molecular , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/tratamiento farmacológico , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
Nat Nanotechnol ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783058

RESUMEN

Therapeutic genome editing of haematopoietic stem cells (HSCs) would provide long-lasting treatments for multiple diseases. However, the in vivo delivery of genetic medicines to HSCs remains challenging, especially in diseased and malignant settings. Here we report on a series of bone-marrow-homing lipid nanoparticles that deliver mRNA to a broad group of at least 14 unique cell types in the bone marrow, including healthy and diseased HSCs, leukaemic stem cells, B cells, T cells, macrophages and leukaemia cells. CRISPR/Cas and base editing is achieved in a mouse model expressing human sickle cell disease phenotypes for potential foetal haemoglobin reactivation and conversion from sickle to non-sickle alleles. Bone-marrow-homing lipid nanoparticles were also able to achieve Cre-recombinase-mediated genetic deletion in bone-marrow-engrafted leukaemic stem cells and leukaemia cells. We show evidence that diverse cell types in the bone marrow niche can be edited using bone-marrow-homing lipid nanoparticles.

5.
Hematol Oncol Clin North Am ; 37(2): 433-447, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36907613

RESUMEN

After many years of intensive research, emerging data from clinical trials indicate that gene therapy for transfusion-dependent ß-thalassemia is now possible. Strategies for therapeutic manipulation of patient hematopoietic stem cells include lentiviral transduction of a functional erythroid-expressed ß-globin gene and genome editing to activate fetal hemoglobin production in patient red blood cells. Gene therapy for ß-thalassemia and other blood disorders will invariably improve as experience accumulates over time. The best overall approaches are not known and perhaps not yet established. Gene therapy comes at a high cost, and collaboration between multiple stakeholders is required to ensure that these new medicines are administered equitably.


Asunto(s)
Talasemia beta , Humanos , Talasemia beta/genética , Edición Génica , Eritrocitos , Terapia Genética , Células Madre Hematopoyéticas
6.
JCI Insight ; 8(1)2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36413407

RESUMEN

Diamond-Blackfan anemia (DBA) is a genetic blood disease caused by heterozygous loss-of-function mutations in ribosomal protein (RP) genes, most commonly RPS19. The signature feature of DBA is hypoplastic anemia occurring in infants, although some older patients develop multilineage cytopenias with bone marrow hypocellularity. The mechanism of anemia in DBA is not fully understood and even less is known about the pancytopenia that occurs later in life, in part because patient hematopoietic stem and progenitor cells (HSPCs) are difficult to obtain, and the current experimental models are suboptimal. We modeled DBA by editing healthy human donor CD34+ HSPCs with CRISPR/Cas9 to create RPS19 haploinsufficiency. In vitro differentiation revealed normal myelopoiesis and impaired erythropoiesis, as observed in DBA. After transplantation into immunodeficient mice, bone marrow repopulation by RPS19+/- HSPCs was profoundly reduced, indicating hematopoietic stem cell (HSC) impairment. The erythroid and HSC defects resulting from RPS19 haploinsufficiency were partially corrected by transduction with an RPS19-expressing lentiviral vector or by Cas9 disruption of TP53. Our results define a tractable, biologically relevant experimental model of DBA based on genome editing of primary human HSPCs and they identify an associated HSC defect that emulates the pan-hematopoietic defect of DBA.


Asunto(s)
Anemia de Diamond-Blackfan , Humanos , Animales , Ratones , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Médula Ósea/metabolismo , Antígenos CD34/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
bioRxiv ; 2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36865281

RESUMEN

On-target toxicity to normal cells is a major safety concern with targeted immune and gene therapies. Here, we developed a base editing (BE) approach exploiting a naturally occurring CD33 single nucleotide polymorphism leading to removal of full-length CD33 surface expression on edited cells. CD33 editing in human and nonhuman primate (NHP) hematopoietic stem and progenitor cells (HSPCs) protects from CD33-targeted therapeutics without affecting normal hematopoiesis in vivo , thus demonstrating potential for novel immunotherapies with reduced off-leukemia toxicity. For broader applications to gene therapies, we demonstrated highly efficient (>70%) multiplexed adenine base editing of the CD33 and gamma globin genes, resulting in long-term persistence of dual gene-edited cells with HbF reactivation in NHPs. In vitro , dual gene-edited cells could be enriched via treatment with the CD33 antibody-drug conjugate, gemtuzumab ozogamicin (GO). Together, our results highlight the potential of adenine base editors for improved immune and gene therapies.

8.
Nat Biomed Eng ; 7(5): 616-628, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37069266

RESUMEN

Sickle-cell disease (SCD) is caused by an A·T-to-T·A transversion mutation in the ß-globin gene (HBB). Here we show that prime editing can correct the SCD allele (HBBS) to wild type (HBBA) at frequencies of 15%-41% in haematopoietic stem and progenitor cells (HSPCs) from patients with SCD. Seventeen weeks after transplantation into immunodeficient mice, prime-edited SCD HSPCs maintained HBBA levels and displayed engraftment frequencies, haematopoietic differentiation and lineage maturation similar to those of unedited HSPCs from healthy donors. An average of 42% of human erythroblasts and reticulocytes isolated 17 weeks after transplantation of prime-edited HSPCs from four SCD patient donors expressed HBBA, exceeding the levels predicted for therapeutic benefit. HSPC-derived erythrocytes carried less sickle haemoglobin, contained HBBA-derived adult haemoglobin at 28%-43% of normal levels and resisted hypoxia-induced sickling. Minimal off-target editing was detected at over 100 sites nominated experimentally via unbiased genome-wide analysis. Our findings support the feasibility of a one-time prime editing SCD treatment that corrects HBBS to HBBA, does not require any viral or non-viral DNA template and minimizes undesired consequences of DNA double-strand breaks.


Asunto(s)
Anemia de Células Falciformes , Edición Génica , Adulto , Humanos , Ratones , Animales , Sistemas CRISPR-Cas , Globinas beta/genética , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/genética , Células Madre Hematopoyéticas , Fenotipo , ADN
9.
Nat Genet ; 55(7): 1210-1220, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37400614

RESUMEN

Inducing fetal hemoglobin (HbF) in red blood cells can alleviate ß-thalassemia and sickle cell disease. We compared five strategies in CD34+ hematopoietic stem and progenitor cells, using either Cas9 nuclease or adenine base editors. The most potent modification was adenine base editor generation of γ-globin -175A>G. Homozygous -175A>G edited erythroid colonies expressed 81 ± 7% HbF versus 17 ± 11% in unedited controls, whereas HbF levels were lower and more variable for two Cas9 strategies targeting a BCL11A binding motif in the γ-globin promoter or a BCL11A erythroid enhancer. The -175A>G base edit also induced HbF more potently than a Cas9 approach in red blood cells generated after transplantation of CD34+ hematopoietic stem and progenitor cells into mice. Our data suggest a strategy for potent, uniform induction of HbF and provide insights into γ-globin gene regulation. More generally, we demonstrate that diverse indels generated by Cas9 can cause unexpected phenotypic variation that can be circumvented by base editing.


Asunto(s)
Anemia de Células Falciformes , Talasemia beta , Ratones , Animales , gamma-Globinas/genética , gamma-Globinas/metabolismo , Edición Génica , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Anemia de Células Falciformes/genética , Antígenos CD34/metabolismo , Talasemia beta/genética
10.
Dis Model Mech ; 15(6)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35793591

RESUMEN

We characterized the human ß-like globin transgenes in two mouse models of sickle cell disease (SCD) and tested a genome-editing strategy to induce red blood cell fetal hemoglobin (HbF; α2γ2). Berkeley SCD mice contain four to 22 randomly arranged, fragmented copies of three human transgenes (HBA1, HBG2-HBG1-HBD-HBBS and a mini-locus control region) integrated into a single site of mouse chromosome 1. Cas9 disruption of the BCL11A repressor binding motif in the γ-globin gene (HBG1 and HBG2; HBG) promoters of Berkeley mouse hematopoietic stem cells (HSCs) caused extensive death from multiple double-strand DNA breaks. Long-range sequencing of Townes SCD mice verified that the endogenous Hbb genes were replaced by single-copy segments of human HBG1 and HBBS including proximal but not some distal gene-regulatory elements. Townes mouse HSCs were viable after Cas9 disruption of the HBG1 BCL11A binding motif but failed to induce HbF to therapeutic levels, contrasting with human HSCs. Our findings provide practical information on the genomic structures of two common mouse SCD models, illustrate their limitations for analyzing therapies to induce HbF and confirm the importance of distal DNA elements in human globin regulation. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Anemia de Células Falciformes , Hemoglobina Fetal , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Modelos Animales de Enfermedad , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Edición Génica , Humanos , Ratones , Factores de Transcripción/genética , Transgenes , gamma-Globinas/genética
11.
Stem Cells ; 28(4): 713-20, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20201064

RESUMEN

We report here that butyrate, a naturally occurring fatty acid commonly used as a nutritional supplement and differentiation agent, greatly enhances the efficiency of induced pluripotent stem (iPS) cell derivation from human adult or fetal fibroblasts. After transient butyrate treatment, the iPS cell derivation efficiency is enhanced by 15- to 51-fold using either retroviral or piggyBac transposon vectors expressing 4 to 5 reprogramming genes. Butyrate stimulation is more remarkable (>100- to 200-fold) on reprogramming in the absence of either KLF4 or MYC transgene. Butyrate treatment did not negatively affect properties of iPS cell lines established by either 3 or 4 retroviral vectors or a single piggyBac DNA transposon vector. These characterized iPS cell lines, including those derived from an adult patient with sickle cell disease by either the piggyBac or retroviral vectors, show normal karyotypes and pluripotency. To gain insights into the underlying mechanisms of butyrate stimulation, we conducted genome-wide gene expression and promoter DNA methylation microarrays and other epigenetic analyses on established iPS cells and cells from intermediate stages of the reprogramming process. By days 6 to 12 during reprogramming, butyrate treatment enhanced histone H3 acetylation, promoter DNA demethylation, and the expression of endogenous pluripotency-associated genes, including DPPA2, whose overexpression partially substitutes for butyrate stimulation. Thus, butyrate as a cell permeable small molecule provides a simple tool to further investigate molecular mechanisms of cellular reprogramming. Moreover, butyrate stimulation provides an efficient method for reprogramming various human adult somatic cells, including cells from patients that are more refractory to reprogramming.


Asunto(s)
Butiratos/farmacología , Técnicas de Cultivo de Célula/métodos , Epigénesis Genética/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Secuencia de Bases , Células Cultivadas , Senescencia Celular , Elementos Transponibles de ADN , Vectores Genéticos/genética , Humanos , Factor 4 Similar a Kruppel , Alineación de Secuencia
12.
Nat Biotechnol ; 38(7): 892-900, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32284586

RESUMEN

The foundational adenine base editors (for example, ABE7.10) enable programmable A•T to G•C point mutations but editing efficiencies can be low at challenging loci in primary human cells. Here we further evolve ABE7.10 using a library of adenosine deaminase variants to create ABE8s. At NGG protospacer adjacent motif (PAM) sites, ABE8s result in ~1.5× higher editing at protospacer positions A5-A7 and ~3.2× higher editing at positions A3-A4 and A8-A10 compared with ABE7.10. Non-NGG PAM variants have a ~4.2-fold overall higher on-target editing efficiency than ABE7.10. In human CD34+ cells, ABE8 can recreate a natural allele at the promoter of the γ-globin genes HBG1 and HBG2 with up to 60% efficiency, causing persistence of fetal hemoglobin. In primary human T cells, ABE8s achieve 98-99% target modification, which is maintained when multiplexed across three loci. Delivered as messenger RNA, ABE8s induce no significant levels of single guide RNA (sgRNA)-independent off-target adenine deamination in genomic DNA and very low levels of adenine deamination in cellular mRNA.


Asunto(s)
Adenina/metabolismo , Sistemas CRISPR-Cas/genética , Citosina/metabolismo , ARN Guía de Kinetoplastida/genética , Adenosina Desaminasa , ADN/genética , Edición Génica/métodos , Células HEK293 , Humanos , Mutación/genética
13.
Sci Rep ; 8(1): 16304, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389991

RESUMEN

CRISPR/Cas9 mediated gene editing of patient-derived hematopoietic stem and progenitor cells (HSPCs) ex vivo followed by autologous transplantation of the edited HSPCs back to the patient can provide a potential cure for monogenic blood disorders such as ß-hemoglobinopathies. One challenge for this strategy is efficient delivery of the ribonucleoprotein (RNP) complex, consisting of purified Cas9 protein and guide RNA, into HSPCs. Because ß-hemoglobinopathies are most prevalent in developing countries, it is desirable to have a reliable, efficient, easy-to-use and cost effective delivery method. With this goal in mind, we developed TRansmembrane Internalization Assisted by Membrane Filtration (TRIAMF), a new method to quickly and effectively deliver RNPs into HSPCs by passing a RNP and cell mixture through a filter membrane. We achieved robust gene editing in HSPCs using TRIAMF and demonstrated that the multilineage colony forming capacities and the competence for engraftment in immunocompromised mice of HSPCs were preserved post TRIAMF treatment. TRIAMF is a custom designed system using inexpensive components and has the capacity to process HSPCs at clinical scale.


Asunto(s)
Hemoglobina Fetal/genética , Filtración/métodos , Edición Génica/métodos , Trasplante de Células Madre Hematopoyéticas , Ribonucleoproteínas/genética , Animales , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Células Cultivadas , Electroporación/métodos , Femenino , Hemoglobina Fetal/metabolismo , Filtración/economía , Filtración/instrumentación , Terapia Genética/economía , Terapia Genética/instrumentación , Terapia Genética/métodos , Células Madre Hematopoyéticas/metabolismo , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Humanos , Membranas Artificiales , Ratones , Modelos Animales , ARN Guía de Kinetoplastida/genética , Trasplante Autólogo
14.
Biomater Sci ; 5(12): 2398-2402, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29067374

RESUMEN

A hindered urea bond (HUB), recently reported as a new type of dynamic chemical bond, can be facilely constructed by mixing an isocyanate and a hindered amine. Here, we report the use of the HUB in the design of degradable hydrogel materials for applications of stem cell encapsulation and delivery. Polyethyleneglycol (PEG) diamine was end-capped with a HUB and an allyl group in a one-pot synthesis. The resulting polymer was cross-linked to form a hydrogel under UV with the addition of a 4-arm PEG thiol and a photoinitiator. The degradation properties of the hydrogels were confirmed with NMR, GPC, weight loss, and protein release studies. We found that the degradation kinetics is dependent on the size of the N-substituents, and the one with the tert-butyl group shows complete degradation within 2 days. The new hydrogel materials were also demonstrated to be biocompatible with hMSCs, and the cell release kinetics can be facilely tuned over 5 days.


Asunto(s)
Materiales Biocompatibles/química , Hidrogeles/química , Células Madre Mesenquimatosas/efectos de los fármacos , Urea/química , Aminas/química , Materiales Biocompatibles/uso terapéutico , Humanos , Hidrogeles/uso terapéutico , Hidrólisis , Isocianatos/química , Cinética , Polietilenglicoles/química , Polímeros/química , Proteínas/química , Compuestos de Sulfhidrilo/química
15.
ACS Biomater Sci Eng ; 2(3): 326-335, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-33429536

RESUMEN

Gene delivery is an important tool to study and manipulate human pluripotent stem cells for regenerative medicine purposes. Yet current methods of transient gene delivery to stem cells are still inefficient. Through the combination of biologically based concepts and material design, we aim to develop new methods to enhance the efficiency of gene delivery to stem cells. Specifically, we use poly(γ-4-(((2-(piperidin-1-yl)ethyl)amino)methyl)benzyl-l-glutamate) (PVBLG-8), a membrane-active helical, cationic polypeptide, to condense plasmid DNA to form stable nanocomplexes, which are further coated with hyaluronic acid (HA). HA not only shields the positive charges of PVBLG-8 to reduce toxicity, but also acts as a targeting moiety for cell surface receptor CD44, which binds HA and facilitates the internalization of the nanocomplexes. Upon entering cells, HA is degraded by hyaluronidase in endosomes and PVBLG-8 is exposed, facilitating the endosomal escape of DNA/polypeptide complex. Our studies show that the coating of HA significantly increases gene transfection efficiency of DNA/PVBLG-8 nanocomplexes from about 28 to 36% with largely reduced toxicity.

16.
IEEE Trans Image Process ; 14(12): 1977-89, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16370452

RESUMEN

Error diffusion halftoning is a popular method of producing frequency modulated (FM) halftones for printing and display. FM halftoning fixes the dot size (e.g., to one pixel in conventional error diffusion) and varies the dot frequency according to the intensity of the original grayscale image. We generalize error diffusion to produce FM halftones with user-controlled dot size and shape by using block quantization and block filtering. As a key application, we show how block-error diffusion may be applied to embed information in hardcopy using dot shape modulation. We enable the encoding and subsequent decoding of information embedded in the hardcopy version of continuous-tone base images. The encoding-decoding process is modeled by robust data transmission through a noisy print-scan channel that is explicitly modeled. We refer to the encoded printed version as an image barcode due to its high information capacity that differentiates it from common hardcopy watermarks. The encoding/halftoning strategy is based on a modified version of block-error diffusion. Encoder stability, image quality versus information capacity tradeoffs, and decoding issues with and without explicit knowledge of the base image are discussed.


Asunto(s)
Algoritmos , Gráficos por Computador , Seguridad Computacional , Interpretación de Imagen Asistida por Computador/métodos , Impresión/métodos , Etiquetado de Productos/métodos , Procesamiento de Señales Asistido por Computador , Artefactos , Colorimetría/métodos , Aumento de la Imagen/métodos
17.
Chem Sci ; 6(4): 2182-2186, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26146536

RESUMEN

Current cancer targeting relying on specific biological interaction between cell surface antigen and respective antibody or its analogue has proven to be effective in the treatment of different cancers; however, this strategy has its own limitations, such as heterogeneity of cancer cells and immunogenicity of the biomacromolecule binding ligands. Bioorthogonal chemical conjugation has emerged as an attractive alternative to biological interaction for in vivo cancer targeting. Here, we report an in vivo cancer targeting strategy mediated by bioorthogonal oxime ligation. Oxyamine group, the artificial target, is introduced onto 4T1 murine breast cancer cells through liposome delivery and fusion. Poly(ethylene glycol) -polylactide (PEG-PLA) nanoparticle (NP) is surface-functionalized with aldehyde groups as targeting ligands. The improved in vivo cancer targeting of PEG-PLA NPs is achieved through specific and efficient chemical reaction between the oxyamine and aldehyde groups.

18.
Biomater Sci ; 3(7): 1061-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26146551

RESUMEN

We report the design and development of redox-responsive chain-shattering polymeric therapeutics (CSPTs). CSPTs were synthesized by condensation polymerization and further modified with poly(ethylene glycol) (PEG) via "Click" reaction. Size-controlled CSPT nanoparticles (NPs) were formed through nanoprecipitation with high drug loading (up to 18%); the particle size increased in a concentration dependent manner. Drug release from particles was well controlled over 48 h upon redox triggering. The anticancer efficacy of the CSPT NPs was validated both in vitro and in vivo.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Doxorrubicina/toxicidad , Células MCF-7/efectos de los fármacos , Nanopartículas/química , Paclitaxel/uso terapéutico , Polietilenglicoles/química , Polímeros/síntesis química , Animales , Química Farmacéutica , Doxorrubicina/química , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Células MCF-7/química , Ratones , Oxidación-Reducción , Paclitaxel/química , Tamaño de la Partícula , Polimerizacion , Polímeros/química
19.
Chem Commun (Camb) ; 51(32): 6948-51, 2015 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-25798457

RESUMEN

We developed a real-time drug-reporting conjugate (CPT-SS-CyN) composed of a near-infrared (NIR) fluorescent cyanine-amine dye (CyN), a disulfide linker, and a model therapeutic drug (camptothecin, CPT). Treatment with dithiothreitol (DTT) induces cleavage of the disulfide bond, followed by two simultaneous intramolecular cyclization reactions with identical kinetics, one to cleave the urethane linkage to release the NIR dye and the other to cleave the carbonate linkage to release CPT. The released CyN has an emission wavelength (760 nm) that is significantly different from CPT-SS-CyN (820 nm), enabling easy detection and monitoring of drug release. A linear relationship between the NIR fluorescence intensity at 760 nm and the amount of CPT released was observed, substantiating the use of this drug-reporting conjugate to enable precise, real-time, and non-invasive quantitative monitoring of drug release in live cells and semi-quantitative monitoring in live animals.


Asunto(s)
Portadores de Fármacos/química , Liberación de Fármacos , Animales , Camptotecina/química , Carbocianinas/química , Femenino , Colorantes Fluorescentes/química , Células HeLa , Humanos , Rayos Infrarrojos , Ratones , Espectrometría de Fluorescencia , Factores de Tiempo
20.
J Mater Chem B ; 2(46): 8098-8105, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26005572

RESUMEN

Non-viral gene delivery into human embryonic stem cells (hESCs)is an important tool for controlling cell fate. However, the delivery efficiency remains low due in part to the tight colony structure of the cells which prevents effective exposure towards delivery vectors. We herein report a novel approach to enhance non-viral gene delivery to hESCs by transiently altering the cell and colony structure. (R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide (Y-27632), a small molecule that inhibits the rho-associated protein kinase pathway, is utilized to induce transient colony spreading which leads to increased transfection efficiency by 1.5 to 2 folds in a spectrum of non-viral transfection reagents including Lipofectamine 2000 and Fugene HD. After removal of Y-27632 post-transfection, cells can revert back to its normal state and do not show alteration of pluripotency. This approach provides a simple, effective tool to enhance non-viral gene delivery into adherent hESCs for genetic manipulation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA