Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 13(1): 2349347, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38746870

RESUMEN

The innate lymphoid cell (ILC) family is composed of heterogeneous innate effector and helper immune cells that preferentially reside in tissues where they promote tissue homeostasis. In cancer, they have been implicated in driving both pro- and anti-tumor responses. This apparent dichotomy highlights the need to better understand differences in the ILC composition and phenotype within different tumor types that could drive seemingly opposite anti-tumor responses. Here, we characterized the frequency and phenotype of various ILC subsets in melanoma metastases and primary epithelial ovarian tumors. We observed high PD-1 expression on ILC subsets isolated from epithelial ovarian tumor samples, while ILC populations in melanoma samples express higher levels of LAG-3. In addition, we found that the frequency of cytotoxic ILCs and NKp46+ILC3 in tumors positively correlates with monocytic cells and conventional type 2 dendritic cells, revealing potentially new interconnected immune cell subsets in the tumor microenvironment. Consequently, these observations may have direct relevance to tumor microenvironment composition and how ILC subset may influence anti-tumor immunity.


Asunto(s)
Carcinoma Epitelial de Ovario , Inmunidad Innata , Linfocitos Infiltrantes de Tumor , Melanoma , Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Melanoma/inmunología , Melanoma/patología , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/patología , Linfocitos Infiltrantes de Tumor/inmunología , Microambiente Tumoral/inmunología , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Glandulares y Epiteliales/patología , Receptor de Muerte Celular Programada 1/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/patología , Células Dendríticas/metabolismo , Proteína del Gen 3 de Activación de Linfocitos , Antígenos CD/metabolismo
2.
Cancer Immunol Immunother ; 72(7): 2375-2392, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36943460

RESUMEN

Immunotherapeutic strategies aimed at enhancing tumor cell killing by tumor-specific T cells hold great potential for reducing tumor burden and prolonging survival of cancer patients. Although many potential tumor antigens have been described, identifying relevant targets when designing anti-cancer vaccines or targeted cell therapies remains a challenge. To identify novel, potentially immunogenic candidate tumor antigens, we performed integrated tumor transcriptomic, seromic, and proteomic analyses of high grade serous ovarian cancer (HGSC) patient tumor samples. We identified tumor neo-antigens and over-expressed antigens using whole exome and RNA sequencing and examined these in relation to patient-matched auto-antibody repertoires. Focusing on MHC class I epitopes recognized by CD8+ T cells, HLA-binding epitopes were identified or predicted from the highly expressed, mutated, or auto-antibody target antigen, or MHC-associated peptides (MAPs). Recognition of candidate antigenic peptides was assessed within the tumor-infiltrating T lymphocyte (TIL) population expanded from each patient. Known tumor-associated antigens (TAA) and cancer/testis antigens (CTA) were commonly found in the auto-antibody and MAP repertoires and CD8+ TILs recognizing epitopes from these antigens were detected, although neither expression level nor the presence of auto-antibodies correlated with TIL recognition. Auto-antibodies against tumor-mutated antigens were found in most patients, however, no TIL recognition of the highest predicted affinity neo-epitopes was detected. Using high expression level, auto-antibody recognition, and epitope prediction algorithms, we identified epitopes in 5 novel antigens (MOB1A, SOCS3, TUBB, PRKAR1A, CCDC6) recognized by HGSC patient TILs. Furthermore, selection of epitopes from the MAP repertoire identified 5 additional targets commonly recognized by multiple patient TILs. We find that the repertoire of TIL specificities includes recognition of highly expressed and immunogenic self-antigens that are processed and presented by tumors. These results indicate an ongoing autoimmune response against a range of self-antigens targeted by HGSC TILs.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Neoplasias Ováricas , Masculino , Humanos , Femenino , Epítopos/metabolismo , Linfocitos T CD8-positivos , Proteómica , Multiómica , Antígenos de Neoplasias , Péptidos , Autoantígenos , Epítopos de Linfocito T
3.
Blood ; 141(12): 1425-1441, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36179280

RESUMEN

Upregulation of the proto-oncogene T-cell leukemia/lymphoma 1A (TCL1A) is causally implicated in various B-cell and T-cell malignancies. High-level TCL1A correlates with aggressive disease features and inferior clinical outcomes. However, the molecular and cell biological consequences of, particularly nuclear, TCL1A are not fully elucidated. We observed here in mouse models of subcellular site-specific TCL1A-induced lymphomagenesis that TCL1A exerts a strong transforming impact via nuclear topography. In proteomic screens of TCL1A-bound molecules in chronic lymphocytic leukemia (CLL) cells and B-cell lymphoma lines, we identified regulators of cell cycle and DNA repair pathways as novel TCL1A interactors, particularly enriched under induced DNA damage and mitosis. By functional mapping and in silico modeling, we specifically identified the mitotic checkpoint protein, cell division cycle 20 (CDC20), as a direct TCL1A interactor. According to the regulatory impact of TCL1A on the activity of the CDC20-containing mitotic checkpoint and anaphase-promoting complexes during mitotic progression, TCL1A overexpression accelerated cell cycle transition in B-cell lymphoma lines, impaired apoptotic damage responses in association with pronounced chromosome missegregation, and caused cellular aneuploidy in Eµ-TCL1A mice. Among hematopoietic cancers, CDC20 levels seem particularly low in CLL. CDC20 expression negatively correlated with TCL1A and lower expression marked more aggressive and genomically instable disease and cellular phenotypes. Knockdown of Cdc20 in TCL1A-initiated murine CLL promoted aneuploidy and leukemic acceleration. Taken together, we discovered a novel cell cycle-associated effect of TCL1A abrogating controlled cell cycle transition. This adds to our concept of oncogenic TCL1A by targeting genome stability. Overall, we propose that TCL1A acts as a pleiotropic adapter molecule with a synergistic net effect of multiple hijacked pathways.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Linfoma de Células B , Ratones , Animales , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteómica , Linfoma de Células B/genética , Ciclo Celular/genética , Proto-Oncogenes , Proteínas de Ciclo Celular/genética , Mitosis
4.
Front Immunol ; 13: 948358, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36032129

RESUMEN

Innate and adaptive immune cells monitor, recognize, and eliminate transformed cells. Innate lymphoid cells (ILCs) are innate counterparts of T cells that play a key role in many facets of the immune response and have a profound impact on disease states, including cancer. ILCs regulate immune responses by responding and integrating a wide range of signals within the local microenvironment. As primarily tissue-resident cells, ILCs are ideally suited to sense malignant transformation and initiate anti-tumor immunity. However, as ILCs have been associated with anti-tumor and pro-tumor activities in established tumors, they could potentially have dual functions during carcinogenesis by promoting or suppressing the malignant outgrowth of premalignant lesions. Here we discuss emerging evidence that shows that ILCs can impact early tumor development by regulating immune responses against transformed cells, as well as the environmental cues that potentially induce ILC activation in premalignant lesions.


Asunto(s)
Inmunidad Innata , Neoplasias , Carcinogénesis , Humanos , Linfocitos , Microambiente Tumoral
5.
Cancers (Basel) ; 14(9)2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35565201

RESUMEN

Immune regulation is composed of a complex network of cellular and molecular pathways that regulate the immune system and prevent tissue damage. It is increasingly clear that innate lymphoid cells (ILCs) are also armed with immunosuppressive capacities similar to well-known immune regulatory cells (i.e., regulatory T cells). In cancer, immunoregulatory ILCs have been shown to inhibit anti-tumour immune response through various mechanisms including: (a) direct suppression of anti-tumour T cells or NK cells, (b) inhibiting T-cell priming, and (c) promoting other immunoregulatory cells. To provide a framework of understanding the role of immunosuppressive ILCs in the context of cancer, we first outline a brief history and challenges related to defining immunosuppressive ILCs. Furthermore, we focus on the mechanisms of ILCs in suppressing anti-tumour immunity and consequentially promoting tumour progression.

6.
Cancers (Basel) ; 13(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34885076

RESUMEN

Immune checkpoints (IC) are broadly characterized as inhibitory pathways that tightly regulate the activation of the immune system. These molecular "brakes" are centrally involved in the maintenance of immune self-tolerance and represent a key mechanism in avoiding autoimmunity and tissue destruction. Antibody-based therapies target these inhibitory molecules on T cells to improve their cytotoxic function, with unprecedented clinical efficacies for a number of malignancies. Many of these ICs are also expressed on innate lymphoid cells (ILC), drawing interest from the field to understand their function, impact for anti-tumor immunity and potential for immunotherapy. In this review, we highlight ILC specificities at different tissue sites and their migration potential upon inflammatory challenge. We further summarize the current understanding of IC molecules on ILC and discuss potential strategies for ILC modulation as part of a greater anti-cancer armamentarium.

7.
Semin Immunol ; 41: 101284, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-31383506

RESUMEN

The tumor microenvironment is shaped by interactions between tumor cells, stroma, and immune cells. T cells are a critical component of the immunological anti-tumor response and most immunotherapeutic approaches in cancer aim to augment T cell responses. Recently, innate lymphoid cells (ILCs) have been added to the list of cell populations that influence T cell responses within the tumor microenvironment. ILCs were shown to modulate T cell functions through a variety of mechanisms. However, the impact of ILC and T cell interactions on disease outcome appears to be contextual as their phenotype and function is strongly influenced by the local tissue microenvironment. Here we focus on recent advances in understanding the mechanisms of ILCs in regulating T cell responses in inflammatory diseases and cancer.


Asunto(s)
Inmunidad Innata , Inflamación/etiología , Inflamación/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación , Inflamación/patología , Activación de Linfocitos , Neoplasias/patología
8.
Eur J Cancer ; 95: 38-51, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29631102

RESUMEN

BACKGROUND: Clinical trials investigating immuno-oncology (IO) drug combinations are largely based on empiricism or limited non-clinical evaluations. This study identified the current combination IO drug clinical trials and investigated how tumour molecular profiling can help rationalise IO drug combinations. METHODS: IO targets were identified via PubMed search and expert opinion. IO drugs were compiled by searching the National Cancer Institute Drug Dictionary and pharmaceutical pipelines, August 2016. Combination IO trials were obtained by searching doublet IO drug combinations in www.clinicaltrials.gov from September to November 2016. IO target gene expressions were extracted from The Cancer Genome Atlas (TCGA) data set and compared with normal tissues from the Genotype-Tissue Expression database. Differentially expressed genes for each cancer were determined using the Wilcoxon rank-sum test, and p-values were corrected for multiple testing. RESULTS: In total, 178 IO targets were identified; 90 targets have either regulatory approved or investigational therapeutics. In total, 410 combination trials involving ≥2 IO drugs were identified: skin (n = 102) and genitourinary (n = 41) malignancies have the largest number of combination IO trials; 109 trials involved >2 disease sites. Summative patient accrual estimates among all trials are 71,345. Trials combining cytotoxic T lymphocyte antigen 4 (CTLA4) with programmed cell death protein 1 (n = 79) and CTLA4 with programmed cell death ligand 1 (n = 44) are the most common. Gene expression data from TCGA were mined to extract the 178 IO targets in 9089 tumours originating from 19 cancer types. IO target expression-clustered heatmap analysis identified several promising drug combinations. CONCLUSION: Our review highlights the great interest in combination IO clinical trials. Our analysis can enrich IO combination therapy selection.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ensayos Clínicos como Asunto/métodos , Oncología Médica/métodos , Neoplasias/tratamiento farmacológico , Terapia Combinada , Combinación de Medicamentos , Humanos , Inmunoterapia/métodos , Oncología Médica/normas , Racionalización , Proyectos de Investigación/normas
9.
Oncoimmunology ; 5(6): e1160186, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27471632

RESUMEN

Hodgkin lymphoma (HL) presents with a unique histologic pattern. Pathognomonic Hodgkin and Reed-Sternberg (HRS) cells usually account for less than 1% of the tumor and are embedded in a reactive infiltrate mainly comprised of CD4(+) T cells. HRS cells induce an immunosuppressive microenvironment and thereby escape antitumor immunity. To investigate the impact of interactions between HRS cells and T cells, we performed long-term co-culture studies that were further translated into a xenograft model. Surprisingly, we revealed a strong antitumor potential of allogeneic CD4(+) T cells against HL cell lines. HRS and CD4(+) T cells interact by adhesion complexes similar to immunological synapses. Tumor-cell killing was likely based on the recognition of allogeneic major histocompatibility complex class II (MHC-II) receptor, while CD4(+) T cells from MHC-II compatible donors did not develop any antitumor potential in case of HL cell line L428. However, gene expression profiling (GEP) of co-cultured HRS cells as well as tumor infiltration of matched CD4(+) T cells indicated cellular interactions. Moreover, matched CD4(+) T cells could be activated to kill CD30(+) HRS cells when redirected with a CD30-specific chimeric antigen receptor. Our work gives novel insights into the crosstalk between HRS and CD4(+) T cells, suggesting the latter as potent effector cells in the adoptive cell therapy of HL.

10.
Proc Natl Acad Sci U S A ; 110(51): 20729-34, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24302766

RESUMEN

Multinucleated Reed-Sternberg (RS) cells are pathognomonic for classical Hodgkin lymphoma (HL), and their presence is essential for diagnosis. How these giant tumor cells develop is controversial, however. It has been postulated that RS cells arise from mononucleated Hodgkin cells via endomitosis. Conversely, continuous single-cell tracking of HL cell lines by long-term time-lapse microscopy has identified cell fusion as the main route of RS cell formation. In contrast to growth-induced formation of giant Hodgkin cells, fusion of small mononuclear cells followed by a size increase gives rise to giant RS cells. Of note, fusion of cells originating from the same ancestor, termed re-fusion, is seen nearly exclusively. In the majority of cases, re-fusion of daughter cells is preceded by incomplete cytokinesis, as demonstrated by microtubule bonds among the cells. We confirm at the level of individual tracked cells that giant Hodgkin and RS cells have little proliferative capacity, further supporting small mononuclear Hodgkin cells as the proliferative compartment of the HL tumor clone. In addition, sister cells show a shared propensity for re-fusion, providing evidence of early RS cell fate commitment. Thus, RS cell generation is related neither to cell fusion of unrelated Hodgkin cells nor to endomitosis, but rather is mediated by re-fusion of daughter cells that underwent mitosis. This surprising finding supports the existence of a unique mechanism for the generation of multinuclear RS cells that may have implications beyond HL, given that RS-like cells are frequently observed in several other lymphoproliferative diseases as well.


Asunto(s)
Citocinesis , Células Gigantes , Enfermedad de Hodgkin , Leucocitos Mononucleares , Células de Reed-Sternberg , Fusión Celular , Línea Celular Tumoral , Células Gigantes/metabolismo , Células Gigantes/patología , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Células de Reed-Sternberg/metabolismo , Células de Reed-Sternberg/patología
11.
Crit Rev Oncol Hematol ; 88(3): 680-95, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23972664

RESUMEN

Mature T-cell lymphomas/leukemias (MTCL) have been understudied lymphoid neoplasms that currently receive growing attention. Our historically rudimentary molecular understanding and dissatisfactory interventional success in this complex and for the most part poor-prognostic group of tumors is only slightly improving. A major limiting aspect in further progress in these rare neoplasms is the lack of suitable model systems that would substantially facilitate pathogenic studies and pre-clinical drug evaluations. Such representations of MTCL have thus far not been systematically appraised. We therefore provide an overview on existing models and point out their particular advantages and limitations in the context of the specific scientific questions. After addressing issues of species-specific differences and classifications, we summarize data on MTCL cell lines of human as well as murine origin, on murine strain predispositions to MTCL, on available models of genetically engineered mice, and on transplant systems. From an in-silico meta-analysis of available primary data of gene expression profiles on human MTCL we cross-reference genes reported to transform T-cells in mice and reflect on their general vs entity-restricted relevance and on target-promoter influences. Overall, we identify the urgent need for new models of higher fidelity to human MTCL with respect to their increasingly recognized diversity and to predictions of drug response.


Asunto(s)
Linfoma de Células T/etiología , Linfoma de Células T/patología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Humanos , Ratones , Clasificación del Tumor
12.
Curr Hematol Malig Rep ; 8(3): 163-72, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23892905

RESUMEN

T-cell receptor (TCR) signaling is pivotal in T-cell development and function. In peripheral T-cell lymphomas/leukemias (PTCL/L), histogenesis, transforming events, epidemiology, and clinical presentation are also closely linked to TCR-mediated influences. After reviewing the physiology of normal TCR signaling and cellular responses, we describe here the association of subgroups of PTCL/L with specific patterns of TCR activation as relevant tumor-initiating and/or tumor-sustaining programs. We identify PTCL/L with a functionally intact TCR machinery in which stimulation is possibly incited by exogenous antigens or autoantigens. Distinct from these are tumors with autonomous oncogenic signaling by dysregulated TCR components uncoupled from extrinsic receptor input. A further subset is characterized by transforming events that activate molecules acting as substitutes for TCR signaling, but triggering similar downstream cascades. We finally discuss the consequences of such a functional model for TCR-targeted therapeutic strategies including those that are being tested in the clinic and those that still require further development.


Asunto(s)
Transformación Celular Neoplásica , Linfoma de Células T Periférico , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología , Humanos , Linfoma de Células T Periférico/metabolismo
13.
Mol Ther ; 21(6): 1160-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23609016

RESUMEN

Retroviral vectors (RVs) are powerful tools in clinical gene therapy. However, stable genomic integration of RVs can be oncogenic, as reported in several animal models and in clinical trials. Previously, we observed that T-cell receptor (TCR) polyclonal mature T cells are resistant to transformation after gammaretroviral transfer of (proto-)oncogenes, whereas TCR-oligoclonal T cells were transformable in the same setting. Here, we describe the induction of a mature T-cell lymphoma (MTCL) in TCR-oligoclonal OT-I transgenic T cells, transduced with an enhanced green fluorescent protein (EGFP)-encoding gammaretroviral vector. The tumor cells were of a mature T-cell phenotype and serially transplantable. Integration site analysis revealed a proviral hit in Janus kinase 1 (Jak1), which resulted in Jak1 overexpression and Jak/STAT-pathway activation, particularly via signal transducer and activator of transcription 3 (STAT3). Specific inhibition of Jak1 markedly delayed tumor growth. A systematic meta-analysis of available gene expression data on human mature T-cell lymphomas/leukemias confirmed the relevance of Jak/STAT overexpression in sporadic human T-cell tumorigenesis. To our knowledge, this is the first study to describe RV-associated insertional mutagenesis in mature T cells.


Asunto(s)
Linfoma de Células T/genética , Linfoma de Células T/terapia , Mutagénesis Insercional/métodos , Retroviridae/genética , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Exones , Femenino , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Linfoma de Células T/patología , Ratones , Fosforilación , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo
14.
Exp Hematol ; 40(5): 401-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22269116

RESUMEN

We have identified a population of normal mouse LN cells, termed LN lymphoid progenitor (LNLP), resembling common lymphoid progenitor (CLP) in the bone marrow. LNLPs lack lineage markers and express CD127, low levels of CD117 (c-Kit), and Sca-1, but lack fms-related tyrosine kinase 3. They efficiently differentiate in vitro into natural killer (NK) cells and T cells, but not mature B cells. LNLPs injected into nonirradiated lymphopenic mice that have no LN develop into mostly splenic T cells with low numbers of NK cells and B cells. When injected into irradiated mice, they generate NK cells and T cells, but not B cells, in the LN. By contrast, bone marrow CLPs develop into mostly B cells with very small numbers of T and NK cells in recipients' spleen and LN. LNLPs have NK and T-cell potentials, but little B-cell potential, and they can develop into NK cells within the LN of normal mice, but their contribution to the T-cell lineage is unknown.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Asesinas Naturales/citología , Ganglios Linfáticos/citología , Linfopoyesis/fisiología , Linfocitos T/citología , Animales , Antígenos Ly/análisis , Linfocitos B/citología , Médula Ósea , Linaje de la Célula , Células Cultivadas/citología , Trasplante de Células Madre Hematopoyéticas , Subunidad alfa del Receptor de Interleucina-7/análisis , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Proteínas Proto-Oncogénicas c-kit/análisis , Quimera por Radiación , Organismos Libres de Patógenos Específicos
15.
BMC Immunol ; 12: 57, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21978263

RESUMEN

BACKGROUND: Dendritic cells (DCs) are the highly specialized antigen presenting cells of the immune system that play a key role in regulating immune responses. DCs can efficiently initiate immune responses or induce tolerance. Due to this dual function, DCs are studied in the context of immunotherapy for both cancer and autoimmune diseases. Characterization of DC-specific genes, leading to better understanding of DC immunobiology, will help to guide their use in clinical settings. We previously identified DC-STAMP, a multi-membrane spanning protein preferentially expressed by DCs. DC-STAMP resides in the endoplasmic reticulum (ER) of immature DCs and translocates towards the Golgi compartment upon maturation. In this study we knocked down DC-STAMP in mouse bone marrow-derived DCs (mBMDCs) to determine its function. RESULTS: We demonstrate that DC-STAMP knock-down mBMDCs secrete less IL-6, IL-12, TNF-α and IL-10 while IL-1 production is enhanced. Moreover, LPS-matured DC-STAMP knock-down mBMDCs show impaired T cell activation potential and induction of Th1 responses in an alloreaction. CONCLUSIONS: We show that DC-STAMP plays an important role in cytokine production by mBMDCs following LPS exposure. Our results reveal a novel function of DC-STAMP in regulating DC-initiated immune responses.


Asunto(s)
Citocinas/metabolismo , Células Dendríticas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Linfocitos T/metabolismo , Animales , Médula Ósea/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Activación de Linfocitos/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , ARN Interferente Pequeño/genética , Linfocitos T/patología
16.
Blood ; 117(15): 4012-21, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21355093

RESUMEN

A subset of natural killer (NK) cells in normal mouse lymph node (LN) expresses CD127 (IL-7 receptor-α chain) and is thought to derive from the thymus. However, CD127(+) NK cells are found in the LN of athymic mice. Therefore, the origin of CD127(+) NK cells in the LN is unclear. Here, we have identified unique NK-cell progenitors (NKPs) in the LN that express the pan-NK cell marker CD49b and CD127 but lack CD122 and lineage markers. The LN NKPs develop in vitro into CD127(+) NK cells that display natural cytotoxicity and cytokine production capacity. They also become CD127(+) NK cells in lymphopenic mice that received a transplant. LN NKPs can be divided into stem cell antigen-1 (Sca-1)(hi) and Sca-1(lo) subsets. The latter comprise ∼ 60% of LN NKPs in normal mouse and < 10% of athymic mouse LN NKPs. Whereas both Sca-1(hi) and Sca-1(lo) NKPs develop into CD127(+) NK cells in vitro, only those derived from Sca-1(lo) LN NKPs have rearranged TCRγ genes. Thus, CD127(+) NK cells in the LN seem to be generated, at least in part, from both thymus-dependent Sca-1(lo) and thymus-independent Sca-1(hi) LN NKPs.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-7/metabolismo , Células Asesinas Naturales/citología , Ganglios Linfáticos/citología , Células Madre/citología , Timo/citología , Animales , Antígenos Ly/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Integrina alfa2/metabolismo , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ganglios Linfáticos/inmunología , Linfopenia/inmunología , Linfopenia/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Células Madre/inmunología , Timo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...