Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Nature ; 610(7933): 737-743, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36071167

RESUMEN

The mutualistic relationship of gut-resident microbiota and the host immune system promotes homeostasis that ensures maintenance of the microbial community and of a largely non-aggressive immune cell compartment1,2. The consequences of disturbing this balance include proximal inflammatory conditions, such as Crohn's disease, and systemic illnesses. This equilibrium is achieved in part through the induction of both effector and suppressor arms of the adaptive immune system. Helicobacter species induce T regulatory (Treg) and T follicular helper (TFH) cells under homeostatic conditions, but induce inflammatory T helper 17 (TH17) cells when induced Treg (iTreg) cells are compromised3,4. How Helicobacter and other gut bacteria direct T cells to adopt distinct functions remains poorly understood. Here we investigated the cells and molecular components required for iTreg cell differentiation. We found that antigen presentation by cells expressing RORγt, rather than by classical dendritic cells, was required and sufficient for induction of Treg cells. These RORγt+ cells-probably type 3 innate lymphoid cells and/or Janus cells5-require the antigen-presentation machinery, the chemokine receptor CCR7 and the TGFß activator αv integrin. In the absence of any of these factors, there was expansion of pathogenic TH17 cells instead of iTreg cells, induced by CCR7-independent antigen-presenting cells. Thus, intestinal commensal microbes and their products target multiple antigen-presenting cells with pre-determined features suited to directing appropriate T cell differentiation programmes, rather than a common antigen-presenting cell that they endow with appropriate functions.


Asunto(s)
Diferenciación Celular , Microbioma Gastrointestinal , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Linfocitos T Reguladores , Células Dendríticas/inmunología , Microbioma Gastrointestinal/inmunología , Homeostasis , Inmunidad Innata , Integrina alfaV/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores CCR7/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología
4.
Cell Rep ; 36(8): 109608, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433045

RESUMEN

Differentiation of intestinal T helper 17 (Th17) cells, which contribute to mucosal barrier protection from invasive pathogens, is dependent on colonization with distinct commensal bacteria. Segmented filamentous bacteria (SFB) are sufficient to support Th17 cell differentiation in mouse, but the molecular and cellular requirements for this process remain incompletely characterized. Here, we show that intestine-draining mesenteric lymph nodes (MLNs), not intestine proper, are the dominant site of SFB-induced intestinal Th17 cell differentiation. Subsequent migration of these cells to the intestinal lamina propria is dependent on their upregulation of integrin ß7. Stat3-dependent induction of RORγt, the Th17 cell-specifying transcription factor, largely depends on IL-6, but signaling through the receptors for IL-21 and IL-23 can compensate for absence of IL-6 to promote SFB-directed Th17 cell differentiation. These results indicate that redundant cytokine signals guide commensal microbe-dependent Th17 cell differentiation in the MLNs and accumulation of the cells in the lamina propria.


Asunto(s)
Diferenciación Celular/inmunología , Citocinas/metabolismo , Intestinos/inmunología , Ganglios Linfáticos/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular/fisiología , Citocinas/inmunología , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Activación de Linfocitos/inmunología , Ratones
5.
Sci Adv ; 6(47)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208369

RESUMEN

Harnessing CRISPR-Cas9 technology for cancer therapeutics has been hampered by low editing efficiency in tumors and potential toxicity of existing delivery systems. Here, we describe a safe and efficient lipid nanoparticle (LNP) for the delivery of Cas9 mRNA and sgRNAs that use a novel amino-ionizable lipid. A single intracerebral injection of CRISPR-LNPs against PLK1 (sgPLK1-cLNPs) into aggressive orthotopic glioblastoma enabled up to ~70% gene editing in vivo, which caused tumor cell apoptosis, inhibited tumor growth by 50%, and improved survival by 30%. To reach disseminated tumors, cLNPs were also engineered for antibody-targeted delivery. Intraperitoneal injections of EGFR-targeted sgPLK1-cLNPs caused their selective uptake into disseminated ovarian tumors, enabled up to ~80% gene editing in vivo, inhibited tumor growth, and increased survival by 80%. The ability to disrupt gene expression in vivo in tumors opens new avenues for cancer treatment and research and potential applications for targeted gene editing of noncancerous tissues.


Asunto(s)
Nanopartículas , Neoplasias , Sistemas CRISPR-Cas , Edición Génica , Técnicas de Transferencia de Gen , Liposomas , Neoplasias/genética , Neoplasias/terapia
6.
Nat Commun ; 9(1): 4493, 2018 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-30374059

RESUMEN

Therapeutic alteration of gene expression in vivo can be achieved by delivering nucleic acids (e.g., mRNA, siRNA) using nanoparticles. Recent progress in modified messenger RNA (mmRNA) synthesis facilitated the development of lipid nanoparticles (LNPs) loaded with mmRNA as a promising tool for in vivo protein expression. Although progress have been made with mmRNA-LNPs based protein expression in hepatocytes, cell specificity is still a major challenge. Moreover, selective protein expression is essential for an improved therapeutic effect, due to the heterogeneous nature of diseases. Here, we present a precision protein expression strategy in Ly6c+ inflammatory leukocytes in inflammatory bowel disease (IBD) induced mice. We demonstrate a therapeutic effect in an IBD model by targeted expression of the interleukin 10 in Ly6c+ inflammatory leukocytes. A selective mmRNA expression strategy has tremendous therapeutic potential in IBD and can ultimately become a novel therapeutic modality in many other diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos , Interleucina-10/uso terapéutico , Leucocitos/metabolismo , Animales , Antígenos Ly/metabolismo , Colitis/tratamiento farmacológico , Colitis/patología , Modelos Animales de Enfermedad , Femenino , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Nanopartículas/ultraestructura , Células RAW 264.7 , ARN Mensajero/metabolismo
7.
Nat Nanotechnol ; 13(3): 214-219, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29379205

RESUMEN

Previous studies have identified relevant genes and signalling pathways that are hampered in human disorders as potential candidates for therapeutics. Developing nucleic acid-based tools to manipulate gene expression, such as short interfering RNAs1-3 (siRNAs), opens up opportunities for personalized medicine. Yet, although major progress has been made in developing siRNA targeted delivery carriers, mainly by utilizing monoclonal antibodies (mAbs) for targeting4-8, their clinical translation has not occurred. This is in part because of the massive development and production requirements and the high batch-to-batch variability of current technologies, which rely on chemical conjugation. Here we present a self-assembled modular platform that enables the construction of a theoretically unlimited repertoire of siRNA targeted carriers. The self-assembly of the platform is based on a membrane-anchored lipoprotein that is incorporated into siRNA-loaded lipid nanoparticles that interact with the antibody crystallizable fragment (Fc) domain. We show that a simple switch of eight different mAbs redirects the specific uptake of siRNAs by diverse leukocyte subsets in vivo. The therapeutic potential of the platform is demonstrated in an inflammatory bowel disease model by targeting colon macrophages to reduce inflammatory symptoms, and in a Mantle Cell Lymphoma xenograft model by targeting cancer cells to induce cell death and improve survival. This modular delivery platform represents a milestone in the development of precision medicine.


Asunto(s)
Colitis/terapia , Sistemas de Liberación de Medicamentos/métodos , Liposomas/química , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos , Animales , Anticuerpos Monoclonales/química , Colitis/genética , Femenino , Lipoproteínas/química , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacocinética , ARN Interferente Pequeño/uso terapéutico
8.
Sci Transl Med ; 8(345): 345fs11, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27358495

RESUMEN

Selectins are involved in leukocyte and cancer cell trafficking, which can be targeted with drugs and nanoparticles (Shamay et al., this issue).


Asunto(s)
Neoplasias/metabolismo , Selectinas/metabolismo , Animales , Humanos , Leucocitos/metabolismo , Modelos Biológicos , Neoplasias/genética , Selectinas/genética
9.
ACS Nano ; 9(7): 6706-16, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26042619

RESUMEN

Modulating T cell function by down-regulating specific genes using RNA interference (RNAi) holds tremendous potential in advancing targeted therapies in many immune-related disorders including cancer, inflammation, autoimmunity, and viral infections. Hematopoietic cells, in general, and primary T lymphocytes, in particular, are notoriously hard to transfect with small interfering RNAs (siRNAs). Herein, we describe a novel strategy to specifically deliver siRNAs to murine CD4(+) T cells using targeted lipid nanoparticles (tLNPs). To increase the efficacy of siRNA delivery, these tLNPs have been formulated with several lipids designed to improve the stability and efficacy of siRNA delivery. The tLNPs were surface-functionalized with anti-CD4 monoclonal antibody to permit delivery of the siRNAs specifically to CD4(+) T lymphocytes. Ex vivo, tLNPs demonstrated specificity by targeting only primary CD4(+) T lymphocytes and no other cell types. Systemic intravenous administration of these particles led to efficient binding and uptake into CD4(+) T lymphocytes in several anatomical sites including the spleen, inguinal lymph nodes, blood, and the bone marrow. Silencing by tLNPs occurs in a subset of circulating and resting CD4(+) T lymphocytes. Interestingly, we show that tLNP internalization and not endosome escape is a fundamental event that takes place as early as 1 h after systemic administration and determines tLNPs' efficacy. Taken together, these results suggest that tLNPs may open new avenues for the manipulation of T cell functionality and may help to establish RNAi as a therapeutic modality in leukocyte-associated diseases.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Silenciador del Gen , Nanopartículas/metabolismo , ARN Interferente Pequeño/administración & dosificación , Animales , Linfocitos T CD4-Positivos/trasplante , Células Cultivadas , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Tratamiento con ARN de Interferencia/métodos
10.
PLoS One ; 8(9): e72389, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24039756

RESUMEN

Live mRNA detection allows real time monitoring of specific transcripts and genetic alterations. The main challenge of live genetic detection is overcoming the high background generated by unbound probes and reaching high level of specificity with minimal off target effects. The use of Fluorescence Resonance Energy Transfer (FRET) probes allows differentiation between bound and unbound probes thus decreasing background. Probe specificity can be optimized by adjusting the length and through use of chemical modifications that alter binding affinity. Herein, we report the use of two oligonucleotide FRET probe system to detect a single nucleotide polymorphism (SNP) in murine Hras mRNA, which is associated with malignant transformations. The FRET oligonucleotides were modified with phosphorothioate (PS) bonds, 2'OMe RNA and LNA residues to enhance nuclease stability and improve SNP discrimination. Our results show that a point mutation in Hras can be detected in endogenous RNA of living cells. As determined by an Acceptor Photobleaching method, FRET levels were higher in cells transfected with perfect match FRET probes whereas a single mismatch showed decreased FRET signal. This approach promotes in vivo molecular imaging methods and could further be applied in cancer diagnosis and theranostic strategies.


Asunto(s)
Técnicas de Genotipaje , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , Alelos , Animales , Secuencia de Bases , Línea Celular Tumoral , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Genes ras , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos/química , Oligonucleótidos/genética , Oligonucleótidos Fosforotioatos/química , Oligonucleótidos Fosforotioatos/genética , Mutación Puntual , Sondas ARN/química , Sondas ARN/genética , Estabilidad del ARN , Sensibilidad y Especificidad , Análisis de la Célula Individual
11.
Genes Cells ; 18(3): 225-37, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23350932

RESUMEN

TAF4b is a cell type-specific subunit of the general transcription factor TFIID. Here, we show that TAF4b is highly expressed in embryonic stem cells (ESC) and is down-regulated upon differentiation. To examine the role of TAF4b in ESC, we applied a knockdown (KD) approach. TAF4b depletion is associated with morphological changes and reduced expression of the self-renewal marker alkaline phosphatase. In contrast, KD of TAF4, a ubiquitously expressed TAF4b paralog, retained and even stabilized ESC stemness. Retinoic acid-induced differentiation was facilitated in the absence of TAF4b but was significantly delayed by TAF4 KD. Furthermore, TAF4b supports, whereas TAF4 inhibits, ESC proliferation and cell cycle progression. We identified a subset of TAF4b target genes preferentially expressed in ESC and controlling the cell cycle. Among them are the germ cell-specific transcription factor Sohlh2 and the protein kinase Yes1, which was recently shown to regulate ESC self-renewal. Interestingly, Sohlh2 and Yes1 are also targets of the pluripotency factor Oct4, and their regulation by Oct4 is TAF4b-dependent. Consistent with that, TAF4b but not TAF4 interacts with Oct4. Our findings suggest that TAF4b cooperates with Oct4 to regulate a subset of genes in ESC, whereas TAF4 is required for later embryonic developmental stages.


Asunto(s)
Proliferación Celular , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ciclo Celular/genética , Línea Celular , Células Madre Embrionarias/citología , Ratones , Mutación , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-yes/genética , Proteínas Proto-Oncogénicas c-yes/metabolismo , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética , Transcripción Genética
12.
Methods Mol Biol ; 757: 497-507, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21909930

RESUMEN

Integrins are heterodimeric membrane glycoproteins composed of noncovalently associated α and ß subunits. Integrins support cell attachment and migration on the surrounding extracellular matrix as well as mediate cell-cell interaction in physiological and pathological settings. Constant recycling of integrins from the plasma membrane to the endosome makes integrins ideal receptors for the delivery of drugs to the cell cytoplasm. RNA interference (RNAi) has evolved not only as a powerful tool for studying gene expression and validating new drug targets, but also as a potential therapeutic intervention. However, the major challenge facing the translation of RNAi into clinical practice is the lack of efficient systemic delivery to specific cell types. Utilizing integrins as delivery target, we have recently devised a strategy to target leukocytes termed Integrin-targeted and stabilized NanoParticles (I-tsNPs) that entrap high RNAi payloads and deliver them in a leukocyte-specific manner to induce robust gene silencing.


Asunto(s)
Integrinas , Nanopartículas , ARN Interferente Pequeño , Animales , Línea Celular , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanopartículas/análisis , Nanopartículas/química , Interferencia de ARN
13.
Biomaterials ; 31(26): 6867-75, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20541799

RESUMEN

Delivery of nucleic acids with positively charged lipid nanoparticles ((+)NPs) is widely used as research reagents and potentially for therapeutics due to their ability to deliver nucleic acids into the cell cytoplasm. However, in most reports little attention has been made to their toxic effects. In the present study, we performed comprehensive analyses of the potential toxicity associated with (+)NPs. Mice treated with (+)NPs showed increased liver enzyme release and body weight loss compared to mice treated with neutral or negatively charged NPs ((-)NPs), suggesting hepatotoxicity. Intravenous administration of (+)NPs induced interferon type I response and elevated mRNA levels of interferon responsive genes 15-25-fold higher than neutral and (-)NPs in different subsets of leukocytes. Moreover, treatment with (+)NPs provoked a dramatic pro-inflammatory response by inducing Th1 cytokines expression (IL-2, IFN gamma and TNF alpha) 10-75-fold higher than treatment with control particles. Finally, we showed that activation of TLR4 might serve as the underlying mechanism for induction of an immune response when (+)NPs are used. These results suggest that a careful attention must be made when different types of (+)NPs are being developed as nanotherapeutics.


Asunto(s)
Inmunidad/efectos de los fármacos , Lípidos/toxicidad , Nanopartículas/química , Nanopartículas/toxicidad , Receptor Toll-Like 4/inmunología , Animales , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , ARN Interferente Pequeño/metabolismo , Propiedades de Superficie/efectos de los fármacos , Receptor Toll-Like 4/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...