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











Intervalo de año de publicación
1.
Blood Adv ; 7(9): 1725-1738, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36453632

RESUMEN

We recently described a low-affinity second-generation CD19 chimeric antigen receptor (CAR) CAT that showed enhanced expansion, cytotoxicity, and antitumor efficacy compared with the high-affinity (FMC63-based) CAR used in tisagenlecleucel, in preclinical models. Furthermore, CAT demonstrated an excellent toxicity profile, enhanced in vivo expansion, and long-term persistence in a phase 1 clinical study. To understand the molecular mechanisms behind these properties of CAT CAR T cells, we performed a systematic in vitro characterization of the transcriptomic (RNA sequencing) and protein (cytometry by time of flight) changes occurring in T cells expressing low-affinity vs high-affinity CD19 CARs following stimulation with CD19-expressing cells. Our results show that CAT CAR T cells exhibit enhanced activation to CD19 stimulation and a distinct transcriptomic and protein profile, with increased activation and cytokine polyfunctionality compared with FMC63 CAR T cells. We demonstrate that the enhanced functionality of low-affinity CAT CAR T cells is a consequence of an antigen-dependent priming induced by residual CD19-expressing B cells present in the manufacture.


Asunto(s)
Citocinas , Receptores Quiméricos de Antígenos , Citocinas/metabolismo , Inmunoterapia Adoptiva/métodos , Linfocitos T , Receptores Quiméricos de Antígenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos CD19
2.
Lab Invest ; 102(2): 172-184, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34782726

RESUMEN

The phenotype of glioma-initiating cells (GIC) is modulated by cell-intrinsic and cell-extrinsic factors. Phenotypic heterogeneity and plasticity of GIC is an important limitation to therapeutic approaches targeting cancer stem cells. Plasticity also presents a challenge to the identification, isolation, and propagation of purified cancer stem cells. Here we use a barcode labelling approach of GIC to generate clonal populations over a number of passages, in combination with phenotyping using the established stem cell markers CD133, CD15, CD44, and A2B5. Using two cell lines derived from isocitrate dehydrogenase (IDH)-wildtype glioblastoma, we identify a remarkable heterogeneity of the phenotypes between the cell lines. During passaging, clonal expansion manifests as the emergence of a limited number of barcoded clones and a decrease in the overall number of clones. Dual-labelled GIC are capable of forming traceable clonal populations which emerge after as few as two passages from mixed cultures and through analyses of similarity of relative proportions of 16 surface markers we were able to pinpoint the fate of such populations. By generating tumour organoids we observed a remarkable persistence of dominant clones but also a significant plasticity of stemness marker expression. Our study presents an experimental approach to simultaneously barcode and phenotype glioma-initiating cells to assess their functional properties, for example to screen newly established GIC for tumour-specific therapeutic vulnerabilities.


Asunto(s)
Antígenos CD/inmunología , Neoplasias Encefálicas/inmunología , Glioma/inmunología , Células Madre Neoplásicas/inmunología , Microambiente Tumoral/inmunología , Antígeno AC133/inmunología , Antígeno AC133/metabolismo , Antígenos CD/metabolismo , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Células Cultivadas , Células Clonales/inmunología , Células Clonales/metabolismo , Citometría de Flujo , Glioma/metabolismo , Glioma/patología , Humanos , Receptores de Hialuranos/inmunología , Receptores de Hialuranos/metabolismo , Inmunofenotipificación , Antígeno Lewis X/inmunología , Antígeno Lewis X/metabolismo , Microscopía Confocal , Células Madre Neoplásicas/clasificación , Células Madre Neoplásicas/metabolismo
3.
Sci Rep ; 9(1): 11592, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31406195

RESUMEN

In utero gene therapy (IUGT) to the fetal hematopoietic compartment could be used to treat congenital blood disorders such as ß-thalassemia. A humanised mouse model of ß-thalassemia was used, in which heterozygous animals are anaemic with splenomegaly and extramedullary hematopoiesis. Intrahepatic in utero injections of a ß globin-expressing lentiviral vector (GLOBE), were performed in fetuses at E13.5 of gestation. We analysed animals at 12 and 32 weeks of age, for vector copy number in bone marrow, peripheral blood liver and spleen and we performed integration site analysis. Compared to noninjected heterozygous animals IUGT normalised blood haemoglobin levels and spleen weight. Integration site analysis showed polyclonality. The left ventricular ejection fraction measured using magnetic resonance imaging (MRI) in treated heterozygous animals was similar to that of normal non-ß-thalassemic mice but significantly higher than untreated heterozygous thalassemia mice suggesting that IUGT ameliorated poor cardiac function. GLOBE LV-mediated IUGT normalised the haematological and anatomical phenotype in a heterozygous humanised model of ß-thalassemia.


Asunto(s)
Terapia Genética , Heterocigoto , Imagen por Resonancia Magnética/métodos , Animales , Femenino , Humanos , Ratones , Fenotipo , Embarazo , Talasemia beta/genética
4.
Sci Rep ; 7(1): 5160, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28698572

RESUMEN

Loss of skeletal muscle mass and function occurs with increasing age. Calorie restriction (CR) increases the lifespan of C57Bl/6 mice, but not in the shorter-lived DBA/2 strain. There is some evidence that calorie restriction reduces or delays many of the age-related defects that occur in rodent skeletal muscle. We therefore investigated the effect of short (2.5 month) and longer term (8.5 and 18.5 months) CR on skeletal muscle in male and female C57Bl/6 and DBA/2 mice. We found that short-term CR increased the satellite cell number and collagen VI content of muscle, but resulted in a delayed regenerative response to injury.Consistent with this, the in vitro proliferation of satellite cells derived from these muscles was reduced by CR. The percentage of stromal cells, macrophages, hematopoietic stem cells and fibroadipogenic cells in the mononucleated cell population derived from skeletal muscle was reduced by CR at various stages. But overall, these changes are neither consistent over time, nor between strain and sex. The fact that changes induced by CR do not persist with time and the dissimilarities between the two mouse strains, combined with sex differences, urge caution in applying CR to improve skeletal muscle function across the lifespan in humans.


Asunto(s)
Restricción Calórica/efectos adversos , Colágeno Tipo VI/metabolismo , Músculo Esquelético/citología , Animales , Peso Corporal , Restricción Calórica/métodos , Proliferación Celular , Células Cultivadas , Metabolismo Energético , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Caracteres Sexuales , Factores de Tiempo
5.
Artículo en Inglés | MEDLINE | ID: mdl-27458573

RESUMEN

Toll-like receptor (TLR)-1 and TLR2 have been shown to be receptors for Mycobacterium leprae (M. leprae), yet it is unclear whether M. leprae can signal through alternative TLRs. Other mycobacterial species possess ligands for TLR4 and genetic association studies in human populations suggest that people with TLR4 polymorphisms may be protected against leprosy. Using human embryonic kidney (HEK)-293 cells co-transfected with TLR4, we demonstrate that M. leprae activates TLR4. We used human macrophages to show that M. leprae stimulation of cytokine production is diminished if pre-treated with TLR4 neutralizing antibody. TLR4 protein expression was up-regulated on macrophages derived from non-bacillus Calmette-Guerin (BCG) vaccinated healthy volunteers after incubation with M. leprae, whereas it was down-regulated in macrophages derived from BCG-vaccinated donors. Finally, pre-treatment of macrophages derived from BCG-naive donors with BCG reversed the effect of M. leprae on TLR4 expression. This may be a newly described phenomenon by which BCG vaccination stimulates "non-specific" protection to the human immune system.


Asunto(s)
Vacuna BCG/inmunología , Macrófagos/metabolismo , Mycobacterium leprae/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Vacuna BCG/farmacología , Diferenciación Celular/inmunología , Citocinas/metabolismo , Células HEK293 , Humanos , Lepra/inmunología , Lepra/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Mycobacterium leprae/inmunología , Transducción de Señal , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/inmunología
6.
J Control Release ; 206: 75-90, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25758332

RESUMEN

Targeted nanocarriers undergo endocytosis upon binding to their membrane receptors and are transported into cellular compartments such as late endosomes and lysosomes. In gene delivery the genetic material has to escape from the cellular compartments into the cytosol. The process of endosomal escape is one of the most critical steps for successful gene delivery. For this reason synthetic lipids with fusogenic properties such as 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) are integrated into the nanocarriers. In this study we show that a natural, plant derived glycoside (SO1861) from Saponaria officinalis L. greatly improves the efficacy of lipid based as well as non-lipid based targeted nanoplexes consisting of a targeted K16 peptide with a nucleic acid binding domain and plasmid-DNA, minicircle-DNA or small interfering RNA (siRNA). By confocal live cell imaging and single cell analyses, we demonstrate that SO1861 augments the escape of the genetic cargo out of the intracellular compartments into the cytosol. Co-localisation experiments with fluorescence labelled dextran and transferrin indicate that SO1861 induces the release of the genetic cargo out of endosomes and lysosomes. However, the transduction efficacy of a lentivirus based gene delivery system was not augmented. In order to design receptor-targeted nanoplexes (LPD) with improved functional properties, SO1861 was integrated into the lipid matrix of the LPD. The SO1861 sensitized LPD (LPDS) were characterized by dynamic light scattering and transmission electron microscopy. Compared to their LPD counterparts the LPDS-nanoplexes showed a greatly improved gene delivery. As shown by differential scanning calorimetry SO1861 can be easily integrated into the lipid bilayer of glycerophospholipid model membranes. This underlines the great potential of SO1861 as a new transfection multiplier for non-viral gene delivery systems.


Asunto(s)
ADN/administración & dosificación , Técnicas de Transferencia de Gen , Glicósidos/química , Lípidos/química , Péptidos/química , Plásmidos/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Secuencia de Aminoácidos , Animales , Línea Celular , Endosomas/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Nanopartículas/química , Saponaria/química , Transfección
7.
PLoS One ; 8(7): e68958, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894383

RESUMEN

α-dystroglycan (α-DG) is a peripheral membrane protein that is an integral component of the dystrophin-glycoprotein complex. In an inherited subset of muscular dystrophies known as dystroglycanopathies, α-DG has reduced glycosylation which results in lower affinity binding to several extracellular matrix proteins including laminins. The glycosylation status of α-DG is normally assessed by the binding of the α-DG antibody IIH6 to a specific glycan epitope on α-DG involved in laminin binding. Immunocytochemistry and immunoblotting are two of the most widely used methods to detect the amount of α-DG glycosylation in muscle. While the interpretation of the presence or absence of the epitope on muscle using these techniques is straightforward, the assessment of a mild defect can be challenging. In this study, flow cytometry was used to compare the amount of IIH6-reactive glycans in fibroblasts from dystroglycanopathy patients with defects in genes known to cause α-DG hypoglycosylation to the amount in fibroblasts from healthy and pathological control subjects. A total of twenty one dystroglycanopathy patient fibroblasts were assessed, as well as fibroblasts from three healthy controls and seven pathological controls. Control fibroblasts have clearly detectable amounts of IIH6-reactive glycans, and there is a significant difference in the amount of this glycosylation, as measured by the mean fluorescence intensity of an antibody recognising the epitope and the percentage of cells positive for the epitope, between these controls and dystroglycanopathy patient fibroblasts (p<0.0001 for both). Our results indicate that the amount of α-DG glycosylation in patient fibroblasts is comparable to that in patient skeletal muscle. This method could complement existing immunohistochemical assays in skeletal muscle as it is quantitative and simple to perform, and could be used when a muscle biopsy is not available. This test could also be used to assess the pathogenicity of variants of unknown significance in genes involved in dystroglycanopathies.


Asunto(s)
Distroglicanos/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Distrofias Musculares/diagnóstico , Distrofias Musculares/patología , Niño , Glicosilación , Humanos , Distrofias Musculares/metabolismo
8.
Hum Gene Ther ; 23(11): 1176-85, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22909012

RESUMEN

TRIM5α (tripartite motif-containing protein-5, isoform α)-cyclophilin A fusion proteins are anti-human immunodeficiency virus (HIV) restriction factors that have evolved in certain nonhuman primates over millions of years and protect against HIV and related viruses. Restriction by TRIM5αCypA is potent and highly resistant to viral escape by mutation and, in combination with a suitable gene delivery platform, offers the possibility of novel therapeutic approaches against HIV. Here we report that lentiviral vector delivery of human mimics of TRIM5α-cyclophilin A (TRIM5CypA) fusion proteins afforded robust and durable protection against HIV-1, but resulted in downregulation of host cell antiviral responses mediated by endogenous TRIM5α. We found that substitution of TRIM5α RING, B-box, and coiled-coil domains with similar domains from a related TRIM protein, TRIM21, produced a novel and equally potent inhibitor of HIV-1. Both TRIM5CypA and TRIM21CypA inhibited transduction by HIV-1-derived viral vectors and prevented propagation of replication-competent HIV-1 in human cell lines and in primary human T cells. Restriction factor-modified T cells exhibited preferential survival in the presence of wild-type HIV. Restriction was dependent on proteasomal degradation and was reversed in the presence of the cyclophilin inhibitor cyclosporin. Importantly, TRIM21CypA did not disturb endogenous TRIM5α-mediated restriction of gammaretroviral infection. Furthermore, endogenous TRIM21 antiviral activity was assessed by measuring inhibition of adenovirus-antibody complexes and was found to be preserved in all TRIMCypA-modified groups. We conclude that lentivirus-mediated expression of the novel chimeric restriction factor TRIM21CypA provides highly potent protection against HIV-1 without loss of normal innate immune TRIM activity.


Asunto(s)
Ciclofilina A/genética , Vectores Genéticos/genética , Infecciones por VIH/genética , VIH-1/genética , Ribonucleoproteínas/genética , Factores de Restricción Antivirales , Proteínas Portadoras/metabolismo , Línea Celular , Supervivencia Celular , Expresión Génica , Orden Génico , Terapia Genética , Infecciones por VIH/terapia , Humanos , Transducción Genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Replicación Viral
9.
Small ; 7(22): 3230-8, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21919194

RESUMEN

The impact of nanomaterials such as carbon nanotubes on biological matter is a topic of increasing interest and concern and requires a multifaceted approach to be resolved. A modified cytotoxic (lactate dehydrogenase (LDH)) assay is developed in an attempt to offer a valid and reliable methodology for screening carbon nanotube toxicity in vitro. Two of the most widely used types of surface-modified multiwalled carbon nanotubes (MWNTs) are tested: ammonium-functionalized MWNTs (MWNT-NH3+ ) and Pluronic F127 coated MWNTs (MWNT:F127). Chemically functionalized MWNTs show significantly greater cellular uptake into lung epithelial A549 cells compared to the non-covalently Pluronic F127-coated MWNTs. In spite of this, MWNT:F127 exhibit enhanced cytotoxicity according to the modified LDH assay. The validity of the modified LDH assay is further validated by direct comparison with other less reliable or accurate cytotoxicity assays. These findings indicate the reliability of the modified LDH assay as a screening tool to assess carbon nanotube cytotoxicity and illustrate that high levels of carbon nanotube cellular internalization do not necessarily lead to adverse responses.


Asunto(s)
Endocitosis/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Polímeros/química , Anexina A5/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanotubos de Carbono/ultraestructura , Propidio/metabolismo , Propiedades de Superficie/efectos de los fármacos , Agua
10.
Analyst ; 135(10): 2600-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20694206

RESUMEN

Bio-electrospraying (BES) is a method for directly jetting living cells under conditions that allow their distribution in the x, y, and z axes. Previous work has been focused on achieving jetting in stable cone-jet mode, which is required for precision placement, and these studies have demonstrated that there are no significant effects of bio-electrospraying on cell morphology or viability. In this work, we examine the biological properties of bio-electrosprayed cells using assays of cellular function that range from the molecular level through to integrated cellular systems, and include proteomics, signal transduction, cell growth and proliferation, and the characterisation of apoptotic blebs. From these molecular methods, we have determined that bio-electrospraying, under the electric field conditions used to achieve stable cone-jet mode, causes no alterations to the biological properties and function of the cells being jetted. Bio-electrosprayed and control cells had similar viability, proliferation properties and virtually indistinguishable cell cycle profiles. The biophysical properties of large conducting (BK) potassium channels were unchanged, as were the pharmacological responses of the endogenous muscarinic and exogenous P2Y(11) receptors, both of which are cell surface receptors of the 7TM superfamily. Proteomic analyses revealed that although three proteins had subtle differences in expression level between bio-electrosprayed and control cells, none of these fold differences was above the 1.5-fold cut-off threshold required for further analyses. These findings support the further development of bio-electrosprays as a viable technology for a wide diversity of tissue engineering, regenerative biology, advanced cellular therapeutics and medicinal applications, having significance in the clinic.


Asunto(s)
Ingeniería de Tejidos/métodos , Astrocitoma , Neoplasias Encefálicas , Calcio/metabolismo , Supervivencia Celular , Electroforesis en Gel Bidimensional , Humanos , Indoles/farmacología , Maleimidas/farmacología , Canales de Potasio/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Transfección , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología
11.
FASEB J ; 24(11): 4354-65, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20647548

RESUMEN

One of the major obstacles to the clinical development of gene silencing by small interfering RNA (siRNA) is its effective cytoplasmic delivery. Carbon nanotubes have been proposed as novel nanomaterials that can offer significant advantages for the intracellular delivery of nucleic acids, such as siRNA. We recently demonstrated in a proof-of-principle study that amino-functionalized multiwalled carbon nanotubes (f-MWNT) can effectively deliver in vivo an siRNA sequence, triggering cell apoptosis that results in human lung xenograft eradication and prolonged survival. In the present study, we demonstrate how a newly synthesized series of polycationic dendron-MWNT constructs with a precisely tailored number of amino functions (dendron generations) can complex and effectively deliver double-stranded siRNA to achieve gene silencing in vitro. A systematic comparison between the f-MWNT series in terms of cellular uptake, cytotoxicity, and siRNA complexation is offered. Significant improvement in siRNA delivery with the dendron-MWNT conjugates is shown, and gene silencing was obtained in 2 human cell lines using 2 different siRNA sequences. The study reveals that through f-MWNT structure-biological function analysis novel nanotube-based siRNA transfer vectors can be designed with minimal cytotoxicity and effective delivery and gene-silencing capabilities.


Asunto(s)
Silenciador del Gen , Nanotubos de Carbono , ARN Interferente Pequeño/farmacología , Transporte Biológico/efectos de los fármacos , Cationes/química , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Silenciador del Gen/efectos de los fármacos , Células HeLa , Humanos , Modelos Moleculares , Nanotubos de Carbono/química , Transfección
12.
J Immunol ; 185(1): 134-43, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20498355

RESUMEN

The ectonucleotidase CD39 has recently been described as being highly expressed on regulatory Foxp3(+) CD4 T cells. Through hydrolysis of proinflammatory extracellular ATP, CD39 activity represents a newly described mechanism of regulatory T cell action. We report a novel population of human CD4 T cells that express CD39 yet are Foxp3 negative. These cells produce the proinflammatory cytokines IFN-gamma and IL-17 and fail to suppress proliferation; however, they still have high ATP hydrolysis activity. In the inflammatory site in human juvenile idiopathic arthritis, the CD39(+)Foxp3(-) population is greatly increased compared with peripheral blood of patients or healthy controls. We also show that cells expressing the AMPase CD73 are less frequent in the joint than in blood. To our knowledge, this is the first study to describe and characterize CD39 function on CD4 T cells from the target site in a human autoinflammatory condition. Our data suggest that in human CD4(+) T cells from the inflamed site, CD39 can be highly expressed on two populations, one regulatory and the other of a memory phenotype.


Asunto(s)
Antígenos CD/biosíntesis , Apirasa/biosíntesis , Artritis Juvenil/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Memoria Inmunológica , Mediadores de Inflamación/metabolismo , Subgrupos de Linfocitos T/enzimología , Adolescente , Artritis Juvenil/enzimología , Artritis Juvenil/patología , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/patología , Niño , Femenino , Humanos , Inmunofenotipificación , Inflamación/enzimología , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/fisiología , Masculino , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
13.
Cancer Res ; 69(16): 6598-606, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19654308

RESUMEN

The CD11c(int) B220(+) NK1.1(+) CD49(+) subset of cells has recently been described as IFN-producing killer dendritic cells (IKDC), which share phenotypic and functional properties with both dendritic cells and natural killer cells. We have previously shown that IKDCs within murine bone marrow-derived DC preparations are essential for the antitumor activity of unpulsed DCs. Here we show that bone marrow-derived IKDCs (BM-IKDC) migrate in vivo into tumors and thence to tumor draining lymph nodes, where they highly express MHC class II and costimulatory molecules. In vitro, freshly isolated BM-IKDCs, fluorescence-activated cell sorted to homogeneity, have no intrinsic antigen presentation function unless cocultured with tumor target cells. On killing of target cells, they can cross-present antigens to stimulate antigen-primed CD8 T cells and can also present antigens to antigen-primed CD4 cells. In vivo, in mice lacking class I-restricted antigen-presenting cell function, robust proliferation of antigen-specific T cells is achieved after adoptive transfer of BM-IKDCs at an injection site distant to the tumor site. Therefore, BM-IKDCs are capable of cytotoxic killing of tumor targets and also of potent antigen presentation after encountering antigen in the context of a viable target cell.


Asunto(s)
Presentación de Antígeno/fisiología , Movimiento Celular , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Interferones/metabolismo , Animales , Movimiento Celular/inmunología , Células Cultivadas , Citotoxicidad Inmunológica/fisiología , Células Dendríticas/metabolismo , Genes RAG-1 , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Células 3T3 NIH , Microglobulina beta-2/genética
14.
Cancer Res ; 69(10): 4134-42, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19435900

RESUMEN

Cancer is a leading cause of mortality throughout the world and new treatments are urgently needed. Recent studies suggest that bone marrow-derived mesenchymal stem cells (MSC) home to and incorporate within tumor tissue. We hypothesized that MSCs engineered to produce and deliver tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a transmembrane protein that causes selective apoptosis of tumor cells, would home to and kill cancer cells in a lung metastatic cancer model. Human MSCs were transduced with TRAIL and the IRES-eGFP reporter gene under the control of a tetracycline promoter using a lentiviral vector. Transduced and activated MSCs caused lung (A549), breast (MDAMB231), squamous (H357), and cervical (Hela) cancer cell apoptosis and death in coculture experiments. Subcutaneous xenograft experiments confirmed that directly delivered TRAIL-expressing MSCs were able to significantly reduce tumor growth [0.12 cm(3) (0.04-0.21) versus 0.66 cm(3) (0.21-1.11); P < 0.001]. We then found, using a pulmonary metastasis model, systemically delivered MSCs localized to lung metastases and the controlled local delivery of TRAIL completely cleared the metastatic disease in 38% of mice compared with 0% of controls (P < 0.05). This is the first study to show a significant reduction in metastatic tumor burden with frequent eradication of metastases using inducible TRAIL-expressing MSCs. This has a wide potential therapeutic role, which includes the treatment of both primary tumors and their metastases, possibly as an adjuvant therapy in clearing micrometastatic disease following primary tumor resection.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adipocitos/citología , Adipocitos/fisiología , Adulto , Apoptosis , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Muerte Celular , Diferenciación Celular , Línea Celular , Movimiento Celular , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Trasplante de Células Madre Mesenquimatosas , Metástasis de la Neoplasia/prevención & control , Neoplasias/patología , Osteoblastos/citología , Osteoblastos/fisiología , Plásmidos , Mapeo Restrictivo , Transfección
15.
J Immunol ; 182(6): 3492-3502, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265127

RESUMEN

We have previously reported that IL-10(+) regulatory B cells, known to play an important role in controlling autoimmunity and inflammatory disorders, are contained within the transitional 2 immature (T2) B cell pool (T2 Bregs). Therapeutic strategies facilitating their enrichment or enhancing their suppressive activity are highly attractive. In this study, we report that agonistic anti-CD40 specifically targets T2 B cells and enriches Bregs upon short-term in vitro culture. Although transfer of unmanipulated T2 B cells, isolated from mice with established lupus, failed to confer protection to diseased mice, transfer of in vitro anti-CD40-generated T2 B cells (T2-like-Bregs) significantly improved renal disease and survival by an IL-10-dependent mechanism. T2-like-Bregs readily accumulated in the spleen after transfer, suppressed Th1 responses, induced the differentiation of IL-10(+)CD4(+)T cells, and conveyed a regulatory effect to CD4(+)T cells. In addition, in vivo administration of agonistic anti-CD40, currently on trial for the treatment of cancer, halted and reversed established lupus. Taken together, our results suggest a novel cellular approach for the amelioration of experimental lupus.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Subgrupos de Linfocitos B/inmunología , Antígenos CD40/agonistas , Antígenos CD40/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Subgrupos de Linfocitos B/patología , Subgrupos de Linfocitos B/trasplante , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Lupus Eritematoso Sistémico/patología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Transgénicos , Células TH1/inmunología , Células TH1/patología
16.
Cell Res ; 19(3): 328-39, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18766172

RESUMEN

We have investigated the earliest events in commitment of human epidermal keratinocytes to terminal differentiation. Phosphorylated Akt and caspase activation were detected in cells exiting the basal layer of the epidermis. Activation of Akt by retroviral transduction of primary cultures of human keratinocytes resulted in an increase in abortive clones founded by transit amplifying cells, while inhibition of the upstream kinase, PI3-kinase, inhibited suspension-induced terminal differentiation. Caspase inhibition also blocked differentiation, the primary mediator being caspase 8. Caspase activation was initiated by 2 h in suspension, preceding the onset of expression of the terminal differentiation marker involucrin by several hours. Incubation of suspended cells with fibronectin or inhibition of PI3-kinase prevented caspase induction. At 2 h in suspension, keratinocytes that had become committed to terminal differentiation had increased side scatter, were 7-aminoactinomycin D (7-AAD) positive and annexin V negative; they exhibited loss of mitochondrial membrane potential and increased cardiolipin oxidation, but with no increase in reactive oxygen species. These properties indicate that the onset of terminal differentiation, while regulated by PI3-kinase and caspases, is not a classical apoptotic process.


Asunto(s)
Apoptosis , Diferenciación Celular , Células Epidérmicas , Queratinocitos/citología , Queratinocitos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Compartimento Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Fragmentación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Epidermis/enzimología , Humanos , Queratinocitos/efectos de los fármacos , MAP Quinasa Quinasa 1/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosfatidilserinas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Immunol ; 181(9): 6654-63, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941256

RESUMEN

The CD11c(int)B220(+)NK1.1(+)CD49(+) subset of cells has recently been described as IFN-producing killer dendritic cells (IKDC), which share phenotypic and functional properties of dendritic cells and NK cells. Herein we show that bone marrow-derived murine dendritic cell preparations contain abundant CD11c(int)B220(+)NK1.1(+)CD49(+) cells, the removal of which results in loss of tumoricidal activity of unpulsed dendritic cells in vivo. Moreover, following s.c. injection, as few as 5 x 10(3) highly pure bone marrow-derived IKDC cells are capable of shrinking small contralateral syngeneic tumors in C57BL/6 mice, but not in immunodeficient mice, implying the obligate involvement of host effector cells in tumor rejection. Our data suggest that bone marrow-derived IKDC represent a population that has powerful tumoricidal activity in vivo.


Asunto(s)
Células de la Médula Ósea/inmunología , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Interferón gamma/fisiología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Células de la Médula Ósea/metabolismo , Muerte Celular/inmunología , Línea Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Células Dendríticas/metabolismo , Inmunoterapia Adoptiva/métodos , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/patología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Ratas , Rabdomiosarcoma/inducido químicamente , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/terapia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
18.
J Immunol ; 178(12): 7868-78, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548625

RESUMEN

The immune system contains natural regulatory cells important in the maintenance of tolerance. Although this suppressive function is usually attributed to CD4 regulatory T cells, recent reports have revealed an immunoregulatory role for IL-10-producing B cells in the context of several autoimmune diseases including collagen-induced arthritis. In the present study, we attribute this suppressive function to a B cell subset expressing high levels of CD21, CD23, and IgM, previously identified as transitional 2-marginal zone precursor (T2-MZP) B cells. T2-MZP B cells are present in the spleens of naive mice and increase during the remission phase of arthritis. Following adoptive transfer to immunized DBA/1 mice, T2-MZP B cells significantly prevented new disease and ameliorated established disease. The suppressive effect on arthritis was paralleled by an inhibition of Ag-specific T cell activation and a reduction in cells exhibiting Th1-type functional responses. We also provide evidence that this regulatory subset mediates its suppression through the secretion of suppressive cytokines and not by cell-to-cell contact. The ability to regulate an established immune response by T2-MZP B cells endows this subset of B cells with a striking and previously unrecognized immunoregulatory potential.


Asunto(s)
Artritis Experimental/inmunología , Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Tolerancia Inmunológica , Traslado Adoptivo , Animales , Artritis Experimental/patología , Artritis Experimental/prevención & control , Subgrupos de Linfocitos B/trasplante , Linfocitos B/trasplante , Colágeno Tipo II/inmunología , Inmunoglobulina M/metabolismo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Activación de Linfocitos , Ratones , Fenotipo , Receptores de Complemento 3d/metabolismo , Receptores de IgE/metabolismo , Células TH1/inmunología
19.
Apoptosis ; 12(3): 549-60, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17195958

RESUMEN

Apoptotic elimination of UV-damaged cells from the epidermis is an important step in preventing both the emergence and expansion of cells with carcinogenic potential. A pivotal event in apoptosis is the release of apoptogenic factors from the mitochondria, although the mechanisms by which the different proteins are released are not fully understood. Here we demonstrate that UV radiation induced the mitochondrial to nuclear translocation of apoptosis inducing factor (AIF) in normal skin. The human papillomavirus (HPV) E6 protein prevented release of AIF and other apoptotic factors such as cytochrome c and Omi from mitochondria of UV-damaged primary epidermal keratinocytes and preserved mitochondrial integrity. shRNA silencing of Bak, a target for E6-mediated proteolysis, demonstrated the requirement of Bak for UV-induced AIF release and mitochondrial fragmentation. Furthermore, screening non-melanoma skin cancer biopsies revealed an inverse correlation between HPV status and AIF nuclear translocation. Our results indicate that the E6 activity towards Bak is a key factor that promotes survival of HPV-infected cells that facilitates tumor development.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis/fisiología , Proteínas de Unión al ADN/metabolismo , Mitocondrias , Proteínas Oncogénicas Virales/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Factor Inductor de la Apoptosis/genética , Línea Celular , Citocromos c/genética , Citocromos c/metabolismo , Proteínas de Unión al ADN/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Oncogénicas Virales/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Rayos Ultravioleta , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
20.
Stem Cells ; 25(4): 828-35, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17158238

RESUMEN

It is thought that, as we age, damage to our stem cells may lead to diminished stem cell pool function and, consequently, a reduced organ regeneration potential that contributes to somatic senescence. Stem cells have evolved many antitoxicity mechanisms, and certain mechanisms may be utilized to isolate hematopoietic stem cells. One method exploits the activity of the ATP-binding cassette/G2 transporter to efflux Hoechst 33,342 and results in a stem cell population known as the side population (SP). The SP subset represents a remarkable enrichment for hematopoietic stem cells and provides an opportunity to re-evaluate age-based changes in hematopoietic stem cells. We report here that the frequency of SP cells steadily increases with age, as does the proportion of Lin(-)/Sca-1(+)/c-kit(+) cells that is capable of Hoechst efflux. Phenotyping, progenitor, and long-term repopulation assays have indicated that SP cells in older mice are still stem cells, albeit with a lower homing efficiency than SP cells from younger mice. Analysis of apoptosis within SP cells has revealed an apoptosis-resistant population in SP cells from old mice. Gene expression analysis has determined that SP cells from old mice have a reduced expression of apoptosis-promoting genes than SP cells from young mice. This increase in SP cells with age seems to be an intrinsic property that may be independent of the age of the microenvironment (niche), and our data might provide some clues as to how this alteration in the proportion of stem/progenitor cells occurs. A possible selection-based mechanism of stem cell pool aging is discussed.


Asunto(s)
Envejecimiento/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Hematopoyesis/fisiología , Animales , Células de la Médula Ósea/efectos de la radiación , División Celular , Citometría de Flujo , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , ARN/genética , ARN/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA