Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 7(1): 685, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834758

RESUMEN

Memory T cells demonstrate superior in vivo persistence and antitumor efficacy. However, methods for manufacturing less differentiated T cells are not yet well-established. Here, we show that producing chimeric antigen receptor (CAR)-T cells using berbamine (BBM), a natural compound found in the Chinese herbal medicine Berberis amurensis, enhances the antitumor efficacy of CAR-T cells. BBM is identified through cell-based screening of chemical compounds using induced pluripotent stem cell-derived T cells, leading to improved viability with a memory T cell phenotype. Transcriptomics and metabolomics using stem cell memory T cells reveal that BBM broadly enhances lipid metabolism. Furthermore, the addition of BBM downregulates the phosphorylation of p38 mitogen-activated protein kinase and enhanced mitochondrial respiration. CD19-CAR-T cells cultured with BBM also extend the survival of leukaemia mouse models due to their superior in vivo persistence. This technology offers a straightforward approach to enhancing the antitumor efficacy of CAR-T cells.


Asunto(s)
Bencilisoquinolinas , Receptores Quiméricos de Antígenos , Animales , Bencilisoquinolinas/farmacología , Ratones , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Inmunoterapia Adoptiva/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Técnicas de Cultivo de Célula/métodos
2.
Mol Ther Methods Clin Dev ; 31: 101109, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-37822720

RESUMEN

Allogeneic T cell platforms utilizing induced pluripotent stem cell (iPSC) technology exhibit significant promise for the facilitation of adoptive immunotherapies. While mature T cell receptor (TCR) signaling plays a crucial role in generating T cells from iPSCs, the introduction of exogenous mature TCR genes carries a potential risk of causing graft-versus-host disease (GvHD). In this study, we present the development of truncated TCRα and TCRß chains, termed mini-TCRs, which lack variable domains responsible for recognizing human leukocyte antigen (HLA)-peptide complexes. We successfully induced cytotoxic T lymphocytes (CTLs) from iPSCs by employing mini-TCRs. Combinations of TCRα and TCRß fragments were screened from mini-TCR libraries based on the surface localization of CD3 proteins and their ability to transduce T cell signaling. Consequently, mini-TCR-expressing iPSCs underwent physiological T cell development, progressing from the CD4 and CD8 double-positive stage to the CD8 single-positive stage. The resulting iPSC-derived CTLs exhibited comparable cytokine production and cytotoxicity in comparison to that of full-length TCR-expressing T lymphocytes when chimeric antigen receptors (CARs) were expressed. These findings demonstrate the potential of mini-TCR-carrying iPSCs as a versatile platform for CAR T cell therapy, offering a promising avenue for advancing adoptive immunotherapies.

3.
Mol Ther Nucleic Acids ; 23: 629-639, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33552683

RESUMEN

Genetic engineering of induced pluripotent stem cells (iPSCs) holds great promise for gene and cell therapy as well as drug discovery. However, there are potential concerns regarding the safety and control of gene expression using conventional vectors such as viruses and plasmids. Although human artificial chromosome (HAC) vectors have several advantages as a gene delivery vector, including stable episomal maintenance and the ability to carry large gene inserts, the full potential of HAC transfer into iPSCs still needs to be explored. Here, we provide evidence of a HAC transfer into human iPSCs by microcell-mediated chromosome transfer via measles virus envelope proteins for various applications, including gene and cell therapy, establishment of versatile human iPSCs capable of gene loading and differentiation into T cells, and disease modeling for aneuploidy syndrome. Thus, engineering of human iPSCs via desired HAC vectors is expected to be widely applied in biomedical research.

4.
J Pharmacol Sci ; 124(2): 201-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24451995

RESUMEN

The mouse embryonic stem cell test (mEST) is used to assess the embryotoxicity of drug candidates by evaluating the effects on the cardiac differentiation of stem cells. However, thalidomide embryotoxicity has not yet been reported using the mEST. To detect the effects of thalidomide, we used human induced pluripotent stem cells (hiPSCs) instead of mouse embryonic stem cells, and assessed three endpoints: the inhibition of cardiac differentiation, the cytotoxicity to hiPSCs, and the cytotoxicity to human dermal fibroblasts, according to the mEST. From these data (IC50 values), the embryotoxicity was classified into one of three different classes based on the mEST and our criteria. Valproate was used as a positive control and ascorbic acid was used as a negative control, and their effects were assessed. Similar to valproate, thalidomide was classified as a Class 2 agent, with weak embryotoxicity, by the mEST criteria, and was classified as Category 3 embryotoxic based on our criteria. Ascorbic acid was classified as a Class 1 / Category 1, non-embryotoxic agent, based on both criteria. Thalidomide embryotoxicity was detected in the embryonic stem cell test based on hiPSCs. This test system is thus considered to have a much greater predictive ability than the mEST.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Teratógenos/toxicidad , Talidomida/toxicidad , Pruebas de Toxicidad/métodos , Ácido Ascórbico/farmacología , Células Cultivadas , Células Madre Embrionarias/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Piel/citología , Ácido Valproico/farmacología
5.
Stem Cells Dev ; 20(1): 159-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20497033

RESUMEN

The use of induced pluripotent stem cells (iPSCs) is an exciting frontier in the study and treatment of human diseases through the generation of specific cell types. Here we show the derivation of iPSCs from human nonmobilized peripheral blood (PB) and bone marrow (BM) mononuclear cells (MNCs) by retroviral transduction of OCT3/4, SOX2, KLF4, and c-MYC. The PB- and BM-derived iPSCs were quite similar to human embryonic stem cells with regard to morphology, expression of surface antigens and pluripotency-associated transcription factors, global gene expression profiles, and differentiation potential in vitro and in vivo. Infected PB and BM MNCs gave rise to iPSCs in the presence of several cytokines, although transduction efficiencies were not high. We found that 5 × 10(5) PB MNCs, which corresponds to less than 1 mL of PB, was enough for the generation of several iPSC colonies. Generation of iPSCs from MNCs of nonmobilized PB, with its relative efficiency and ease of harvesting, could enable the therapeutic use of patient-specific pluripotent stem cells.


Asunto(s)
Células Sanguíneas/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes Inducidas/citología , Adulto , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular/genética , Células Clonales , Metilación de ADN/genética , Reordenamiento Génico/genética , Genoma Humano/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Masculino , Regiones Promotoras Genéticas/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Donantes de Tejidos
6.
Stem Cells Dev ; 19(2): 229-38, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19558219

RESUMEN

Reprogramming of somatic cells provides potential for the generation of specific cell types, which could be a key step in the study and treatment of human diseases. In vitro reprogramming of somatic cells into a pluripotent embryonic stem (ES) cell-like state has been reported by retroviral transduction of murine fibroblasts using four embryonic transcription factors or through cell fusion of somatic and pluripotent stem cells. Here we show that mouse adult bone marrow mononuclear cells (BM MNCs) are competent as donor cells and can be reprogrammed into pluripotent ES cell-like cells. We isolated BM MNCs and mouse embryonic fibroblasts (MEFs) from Oct4-GFP transgenic mice, fused them with ES cells, or infected them with retroviruses expressing Oct4, Sox2, Klf4, and c-Myc. Fused BM MNCs formed more ES-like colonies than did MEFs. Infected BM MNCs gave rise to induced pluripotent stem (iPS) cells, although transduction efficiencies were not high. It was more efficient to pick up iPS colonies as compared with MEFs. BM-derived iPS (BM iPS) cells expressed ES cell markers, formed teratomas, and contributed to chimera mice with germ line development. Clonal analysis revealed that BM iPS clones had diversity, although some clones were found to be genetically identical with different phenotypes. Our findings imply that BM MNCs have potential advantages to generate iPS cells for the clinical application.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , Animales , Células de la Médula Ósea/metabolismo , Fusión Celular , Trasplante de Células/métodos , Células Cultivadas , Embrión de Mamíferos/citología , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Transducción Genética
7.
J Hum Genet ; 51(2): 147-150, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16333524

RESUMEN

A number of gene delivery systems are currently being developed for potential use in gene therapy. Here, we demonstrate the feasibility of 21deltaqHAC, a newly developed human artificial chromosome (HAC), as a gene delivery system. We first introduced a 21deltaqHAC carrying an EGFP reporter gene and a geneticin-resistant gene (EGFP-21deltaqHAC) into hematopoietic cells by microcell-mediated chromosome transfer. These HAC-containing hematopoietic cells showed resistance to geneticin, expressed EGFP and retained the ability to differentiate into various lineages, and the EGFP-21deltaqHAC was successfully transduced into primary hematopoietic cells. Hematopoietic cells harboring the EGFP-21deltaqHAC could still be detected at two weeks post-transplantation in immunodeficient mice. We also showed effective expansion of hematopoietic cells by introducing the 21deltaqHAC containing ScFvg, a gp130-based chimeric receptor that transmits growth signals in response to specific-antigen of this receptor. All of these results demonstrate the usefulness of HAC in gene therapy.


Asunto(s)
Cromosomas Artificiales Humanos/genética , Cromosomas Humanos Par 21/genética , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Expresión Génica , Terapia Genética/métodos , Células Madre Hematopoyéticas/metabolismo , Animales , Cartilla de ADN , Citometría de Flujo , Vectores Genéticos/genética , Gentamicinas , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones
8.
Exp Hematol ; 33(12): 1459-69, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16338488

RESUMEN

OBJECTIVE: For the study of the function of genes of interest in hematopoietic stem cells (HSCs) and for successful gene therapy, it is fundamental to develop a method of efficient gene transfer into HSCs. In mice experiments, efforts have been made to raise the transduction efficiency by modifying the vectors, administrating 5-fluorouracil (5-FU) to donor mice, selecting cytokine cocktails to better sustain the long-term repopulating potential of the stem cells, and so on. The objective of this study is to examine whether the use of fibroblast growth factor-1 (FGF-1)-expanded bone marrow cells provide an improved source for retroviral gene delivery to HSCs. MATERIALS AND METHODS: Unfractionated bone marrow cells from one mouse were cultured in serum-free medium containing FGF-1. Both floating and attached cells were transferred to retronectin precoated dishes and infected with virus supernatant from MP34 cells stably transduced with pMY/GFP retrovirus. After 3-day infection, the green fluorescence protein-positive fraction was sorted and the cells were transplanted to lethally irradiated mice. RESULTS: The experiments illustrated that the number of bone marrow-derived competitive repopulation units (CRUs) was increased from 600 to 9300 per mouse after a 3-week culture period with FGF-1. Following retroviral transduction of the expanded cells, the absolute number of sorted retrovirus-transduced CRUs was 4200. Using these retrovirus-transduced cells in noncompetitive reconstitution assay, we achieved radiation protection and long-term bone marrow reconstitution in 100% of the recipients with average myeloid and lymphoid chimerisms of 70% and 50%, respectively, even if we transplanted 150 recipients with cells derived from a single donor mouse. CONCLUSION: In conclusion, FGF-1-expanded bone marrow cells constitute an excellent source of stem cells that could be used in a range of gene delivery protocols.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/farmacología , Células Madre Hematopoyéticas/citología , Retroviridae/genética , Transducción Genética/métodos , Animales , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL
9.
Blood ; 103(9): 3336-41, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14726394

RESUMEN

Stem cell leukemia (SCL) protein has been shown to be an essential transcription factor during hematopoietic development in the embryo. In adult hematopoiesis, however, the role for SCL has remained largely unknown, whereas it is expressed in bone marrow hematopoietic stem cells (HSCs). In this study, we performed HSC transplantation and an in vitro HSC differentiation assay using retrovirally transduced HSCs with wild-type (WT) and dominant-negative (DN) SCL. The transplantation experiments showed that SCL does not affect the long-term repopulating capacity of HSCs but that WT SCL and DN SCL increase the short-term contribution of the transduced HSCs in myeloid and lymphoid lineages, respectively. An in vitro single-cell assay using a fetal thymus organ culture system further demonstrated that WT SCL facilitates HSCs to differentiate into the myeloid lineage but that DN SCL facilitates HSCs to differentiate into the lymphoid lineage. We conclude that the up-regulation or down-regulation of SCL directs HSCs toward myeloid or lymphoid lineage, respectively, although SCL does not affect their long-term repopulating capacity.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Madre Hematopoyéticas/citología , Linfocitos/citología , Células Mieloides/citología , Proteínas Proto-Oncogénicas/fisiología , Factores de Transcripción/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Proteínas de Unión al ADN/genética , Feto , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Proteínas Proto-Oncogénicas/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Timo , Factores de Transcripción/genética , Transducción Genética
10.
Cancer Res ; 63(15): 4516-20, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12907625

RESUMEN

CD4+CD25+ regulatory T cells play an important role in peripheral tolerance. These cells have been reported to be capable of suppressing the response of CD4+CD25- T cells in vitro. The depletion of these cells evokes effective immune responses to tumor cells in vivo. In this study, we demonstrate that CD4+CD25+ T cells also suppress all subsets of Valpha24+NKT cells (Valpha24+CD4-CD8- double negative, Valpha24+CD4+, and Valpha24+CD8+) in both proliferation and cytokine production [IFN-gamma, interleukin-4 (IL-4), IL-13, and IL-10]. This suppression is mediated by cell-to-cell contact but not by a humoral factor or the inhibition of antigen-presenting cells. Moreover, the cytotoxic activity of Valpha24+NKT cells against some tumor cell lines is suppressed by CD4+CD25+ T cells. This finding is important in developing an effective immunotherapy for cancer.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Asesinas Naturales/inmunología , Receptores de Interleucina-2/inmunología , Comunicación Celular/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Galactosilceramidas/farmacología , Humanos , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Interleucinas/biosíntesis , Interleucinas/metabolismo , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Receptores de Interleucina-2/sangre , Subgrupos de Linfocitos T/inmunología
11.
Immunity ; 18(5): 675-85, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12753744

RESUMEN

The Notch genes play a key role in cellular differentiation. The significance of Notch1 during thymocyte development is well characterized, but the function of Notch2 is poorly understood. Here we demonstrate that Notch2 but no other Notch family member is preferentially expressed in mature B cells and that conditionally targeted deletion of Notch2 results in the defect of marginal zone B (MZB) cells and their presumed precursors, CD1d(hi) fraction of type 2 transitional B cells. Among Notch target genes, the expression level of Deltex1 is prominent in MZB cells and strictly dependent on that of Notch2, suggesting that Deltex1 may play a role in MZB cell differentiation.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Portadoras , Diferenciación Celular/genética , Linaje de la Célula/genética , Receptores de Superficie Celular/genética , Animales , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Heterocigoto , Ratones , Ratones Noqueados , Proteínas/metabolismo , Receptor Notch2 , Receptores de Superficie Celular/biosíntesis
12.
Immunity ; 18(5): 699-711, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12753746

RESUMEN

Hematopoietic stem cells (HSCs) are thought to arise in the aorta-gonad-mesonephros (AGM) region of embryo proper, although HSC activity can be detected in yolk sac (YS) and paraaortic splanchnopleura (P-Sp) when transplanted in newborn mice. We examined the role of Notch signaling in embryonic hematopoiesis. The activity of colony-forming cells in the YS from Notch1(-/-) embryos was comparable to that of wild-type embryos. However, in vitro and in vivo definitive hematopoietic activities from YS and P-Sp were severely impaired in Notch1(-/-) embryos. The population representing hemogenic endothelial cells, however, did not decrease. In contrast, Notch2(-/-) embryos showed no hematopoietic deficiency. These data indicate that Notch1, but not Notch2, is essential for generating hematopoietic stem cells from endothelial cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Factores de Transcripción , Animales , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Ratones , Receptor Notch1 , Receptor Notch2 , Saco Vitelino/citología , Saco Vitelino/metabolismo
13.
Blood ; 101(5): 1777-83, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12406868

RESUMEN

Mouse long-term hematopoietic reconstituting cells exist in the c-Kit+Sca-1+Lin- (KSL) cell population; among them, CD34(low/-) cells represent the most highly purified population of hematopoietic stem cells in the adult bone marrow. Here, we demonstrate that retrovirus-mediated transduction of CD34(low/-)c-Kit+Sca-1+Lin- (34-KSL) cells with the HES-1 gene, which encodes a basic helix-loop-helix transcription factor functioning downstream of the Notch receptor, and is a key molecule for the growth phase of neural stem cells in the embryo, preserves the long-term reconstituting activity of these cells in vitro. We also show that cells derived from the HES-1-transduced 34-KSL population produce progenies characterized by negative Hoechst dye staining, which defines the side population, and by CD34(low/-) profile in the bone marrow KSL population in each recipient mouse at ratios 3.5- and 7.8-fold those produced by nontransduced 34-KSL-derived competitor cells. We conclude that HES-1 preserves the long-term reconstituting hematopoietic activity of 34-KSL stem cells ex vivo. Up-regulation of HES-1 protein in the 34-KSL population before unnecessary cell division, that is, without retrovirus transduction, may represent a potent approach to absolute expansion of hematopoietic stem cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Linaje de la Célula , Supervivencia Celular , Células Cultivadas/citología , Genes Reporteros , Proteínas Fluorescentes Verdes , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/clasificación , Proteínas de Homeodominio/genética , Proteínas Luminiscentes/análisis , Ratones , Ratones Endogámicos C57BL , Fenotipo , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Factor de Transcripción HES-1 , Transducción Genética
14.
Nat Med ; 8(4): 403-9, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11927948

RESUMEN

Excessive accumulation of smooth-muscle cells (SMCs) has a key role in the pathogenesis of vascular diseases. It has been assumed that SMCs derived from the outer medial layer migrate, proliferate and synthesize extracellular matrix components on the luminal side of the vessel. Although much effort has been devoted to targeting migration and proliferation of medial SMCs, there is no effective therapy that prevents occlusive vascular remodeling. We show here that in models of post-angioplasty restenosis, graft vasculopathy and hyperlipidemia-induced atherosclerosis, bone-marrow cells give rise to most of the SMCs that contribute to arterial remodeling. Notably, purified hematopoietic stem cells differentiate into SMCs in vitro and in vivo. Our findings indicate that somatic stem cells contribute to pathological remodeling of remote organs, and may provide the basis for the development of new therapeutic strategies for vascular diseases through targeting mobilization, homing, differentiation and proliferation of bone marrow-derived vascular progenitor cells.


Asunto(s)
Arteriosclerosis/etiología , Arteriosclerosis/patología , Células Madre Hematopoyéticas/patología , Músculo Liso Vascular/patología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Diferenciación Celular , División Celular , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Oclusión de Injerto Vascular/etiología , Oclusión de Injerto Vascular/patología , Proteínas Fluorescentes Verdes , Técnicas In Vitro , Operón Lac , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas
15.
Immunol Lett ; 81(1): 59-64, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11841846

RESUMEN

Notch1 is indispensable for T cell development. It is anticipated that Notch1 and other Notch receptors expressed on the surface of thymic T cell precursors are activated by ligands present on environmental cells, including antigen presenting cells (APCs), and involved in positive and negative selections. Notch receptors on peripheral T cells may also be activated by ligands on APCs. Here, we examined the expression pattern of three Notch ligands, Jagged1, 2 and Delta1 in APCs by an immunofluorescence cell staining method and a reverse transcriptase-polymerase chain reaction (RT-PCR) method. Peritoneal macrophages were strongly positive for Jagged1 staining. In contrast, macrophages separated from spleen and dendritic cells (DCs) separated from spleen and thymus showed positive staining for all the three ligands at a similar intensity. An analysis by RT-PCR revealed that peritoneal and splenic macrophages and splenic and thymic DCs, show a distinct pattern in Notch ligand expression. These findings may represent that expression of various Notch ligands in APCs has a physiological relevance in each organ.


Asunto(s)
Proteínas Portadoras/biosíntesis , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Biosíntesis de Proteínas , Receptores de Citocinas/biosíntesis , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/inmunología , Separación Celular , Células Dendríticas/inmunología , Femenino , Inmunoglobulinas , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Proteína Jagged-2 , Ligandos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas/inmunología , Receptores de Citocinas/inmunología , Receptores Notch , Proteínas Serrate-Jagged
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...