Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 139(18): 2782-2796, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35231101

RESUMEN

We observed that the immune checkpoint protein B7-H3 is overexpressed in acute myeloid leukemia (AML) patients with poor treatment outcomes. Inhibition of B7-H3 expression or blocking of its activity using a novel monoclonal antibody (T-1A5) in AML cells significantly enhanced natural killer (NK) cell-mediated cytotoxicity in AML cells in vitro and in vivo. Moreover, a human-mouse chimera of this antibody (ChT-1A5) induced antibody-dependent cell-mediated cytotoxicity (ADCC) in B7-H3+ primary AML cells, but not in normal hematopoietic cells, suggesting the specify of this antibody for AML cells. Epitope mapping studies identified that both T-1A5 and ChT-1A5 antibodies bind to the FG-loop region of B7-H3, which is known to regulate the immunosuppressive function of B7-H3. Furthermore, treatment with ChT-1A5 in combination with human NK cells significantly prolonged survival in AML patient-derived xenograft (PDX) models. Our results suggest that the ChT-1A5 antibody can inhibit the immunosuppressive function of B7-H3 protein as well as induce ADCC in B7-H3+ AML.


Asunto(s)
Proteínas de Punto de Control Inmunitario , Leucemia Mieloide Aguda , Animales , Antígenos B7 , Línea Celular Tumoral , Humanos , Células Asesinas Naturales , Leucemia Mieloide Aguda/terapia , Ratones
2.
Eur J Immunol ; 45(10): 2821-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26138432

RESUMEN

DCs are professional APCs playing a crucial role in the initiation of T-cell responses to combat infection. However, systemic bacterial infection with various pathogens leads to DC-depletion in humans and mice. The mechanisms of pathogen-induced DC-depletion remain poorly understood. Previously, we showed that mice infected with Yersinia enterocolitica (Ye) had impaired de novo DC-development, one reason for DC-depletion. Here, we extend these studies to gain insight into the molecular mechanisms of DC-depletion and the impact of different bacteria on DC-development. We show that the number of bone marrow (BM) hematopoietic progenitors committed to the DC lineage is reduced following systemic infection with different Gram-positive and Gram-negative bacteria. This is associated with a TLR4- and IFN-γ-signaling dependent increase of committed monocyte progenitors in the BM and mature monocytes in the spleen upon Ye-infection. Adoptive transfer experiments revealed that infection-induced monopoiesis occurs at the expense of DC-development. Our data provide evidence for a general response of hematopoietic progenitors upon systemic bacterial infections to enhance monocyte production, thereby increasing the availability of innate immune cells for pathogen control, whereas impaired DC-development leads to DC-depletion, possibly driving transient immunosuppression in bacterial sepsis.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Mielopoyesis/inmunología , Yersiniosis/inmunología , Yersinia enterocolitica/inmunología , Animales , Células Dendríticas/patología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Yersiniosis/patología
3.
Blood ; 124(26): 3914-23, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25359989

RESUMEN

Therapeutic natural killer (NK)-cell-mediated alloreactivity toward acute myeloid leukemia has largely been attributed to mismatches between killer immunoglobulin-like receptors (KIRs) on NK cells and their ligands, HLA class I molecules, on target cells. While adult acute B-cell precursor leukemia (BCP-ALL) appears to be resistant to NK-cell-mediated lysis, recent data indicate that pediatric BCP-ALL might yet be a target of NK cells. In this study, we demonstrate in a donor-patient-specific NOD.Cg-Prkdc(scid) IL2rg(tmWjl)/Sz (NSG) xenotransplantation model that NK cells mediate considerable alloreactivity toward pediatric BCP-ALL in vivo. Notably, both adoptively transferred mature KIR(+) NK cells and immature KIR(-) NK cells arising early posttransplantation in humanized NSG mice exerted substantial antileukemic activity. Low-dose and long-term treatment of humanized NSG mice with the DNA-demethylating agent 5-aza-cytidine distinctly enhanced the antitumor response, interestingly without inducing common inhibitory KIR expression but rather by promoting the differentiation of various NK-cell precursor subsets. Collectively, these data indicate that the future design of innovative therapy protocols should consider further exploitation of NK-cell-mediated immune responses for poor prognosis pediatric BCP-ALL patients.


Asunto(s)
Antineoplásicos/química , Trasplante de Células Madre Hematopoyéticas , Células Asesinas Naturales/citología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores KIR/metabolismo , Animales , Azacitidina/química , Niño , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Metilación de ADN , Modelos Animales de Enfermedad , Genotipo , Efecto Injerto vs Leucemia , Humanos , Interleucina-2/genética , Ratones , Ratones Endogámicos NOD , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Pronóstico , Trasplante Heterólogo
4.
Mol Ther ; 23(4): 648-55, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25578618

RESUMEN

FLT3 is a receptor-tyrosine-kinase that is expressed on leukemic cells of the myeloid and lymphoid lineage rather specifically. We here report on the construction and selection of bispecific FLT3 X CD3 antibodies in a new recombinant format, termed Fabsc, that resembles the normal antibody structure more closely than the well-established bispecific single chain (bssc)-format. Our preferred antibody, which emerged from an initial selection procedure utilizing different FLT3- and CD3-antibodies, contains the FLT3-antibody 4G8 and the CD3-antibody UCHT1. The 4G8 X UCHT1 Fabsc-antibody was found to be superior to a bssc-antibody with identical specificities with respect to (i) affinity to the target antigen FLT3, (ii) production yield by transfected cells, and (iii) the diminished formation of aggregates. T-cell activation in the presence and absence of cultured leukemic cells and killing of these cells was comparable for both molecules. In addition, the 4G8 X UCHT1 Fabsc-antibody was found to induce T-cell activation and efficient killing of leukemic blasts in primary peripheral blood mononuclear cell (PBMC) cultures of acute myeloid leukemia (AML) patients. In these experiments, the bispecific molecule was clearly superior to an Fc-optimized monospecific FLT3-antibody described previously, indicating that within PBMC of AML patients the recruitment of T cells is more effective than that of natural killer cells.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3/inmunología , Leucemia Experimental/terapia , Tirosina Quinasa 3 Similar a fms/inmunología , Animales , Ratones , Ratones Endogámicos C57BL
5.
J Biol Chem ; 289(16): 11068-11082, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24567328

RESUMEN

Recruitment of mesenchymal stem cells (MSC) following cardiac injury, such as myocardial infarction, plays a critical role in tissue repair and may contribute to myocardial recovery. However, the mechanisms that regulate migration of MSC to the site of tissue damage remain elusive. Here, we demonstrate in vitro that activated platelets substantially inhibit recruitment of MSC toward apoptotic cardiac myocytes and fibroblasts. The alarmin high mobility group box 1 (HMGB1) was released by platelets upon activation and mediated inhibition of the cell death-dependent migratory response through Toll-like receptor (TLR)-4 expressed on the MSC. Migration of MSC to apoptotic cardiac myocytes and fibroblasts was driven by hepatocyte growth factor (HGF), and platelet activation was followed by HMGB1/TLR-4-dependent down-regulation of HGF receptor MET on MSC, thereby impairing HGF-driven MSC recruitment. We identify a novel mechanism by which platelets, upon activation, interfere with MSC recruitment to apoptotic cardiac cells, a process that may be of particular relevance for myocardial repair and regeneration.


Asunto(s)
Apoptosis/fisiología , Plaquetas/metabolismo , Movimiento Celular/fisiología , Regulación hacia Abajo/fisiología , Fibroblastos/metabolismo , Proteína HMGB1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/metabolismo , Activación Plaquetaria/fisiología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Receptor Toll-Like 4/metabolismo , Plaquetas/citología , Fibroblastos/citología , Proteína HMGB1/genética , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas c-met/genética , Regeneración/fisiología , Receptor Toll-Like 4/genética
6.
Glycobiology ; 25(8): 902-17, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25978997

RESUMEN

Stage-specific embryonic antigen-4 (SSEA-4) is a glycosphingolipid, which is overexpressed in some cancers and has been linked to disease progression. However, little is known about the functions of SSEA-4 and the characteristics of SSEA-4 expressing tumor cells. Our studies identified SSEA-4 expression on a subpopulation of cells in many solid tumor cell lines but not in leukemic cell lines. Fluorescence-activated cell sorting-sorted SSEA-4(+) prostate cancer cells formed fibroblast-like colonies with limited cell-cell contacts, whereas SSEA-4(-) cells formed cobblestone-like epithelial colonies. Only colonies derived from SSEA-4(+) cells were enriched for pluripotent embryonic stem cell markers. Moreover, major epithelial cell-associated markers Claudin-7, E-cadherin, ESRP1 and GRHL2 were down-regulated in the SSEA-4(+) fraction of DU145 and HCT-116 cells. Similar to cell lines, SSEA-4(+) primary prostate tumor cells also showed down-regulation of epithelial cell-associated markers. In addition, they showed up-regulation of epithelial-to-mesenchymal transition as well as mesenchymal markers. Furthermore, SSEA-4(+) cells escape from adhesive colonies spontaneously and form invadopodia-like migratory structures, in which SSEA-4, cortactin as well as active pPI3K, pAkt and pSrc are enriched and colocalized. Finally, SSEA-4(+) cells displayed strong tumorigenic ability and stable knockdown of SSEA-4 synthesis resulted in decreased cellular adhesion to different extracellular matrices. In conclusion, we introduce SSEA-4 as a novel marker to identify heterogeneous, invasive subpopulations of tumor cells. Moreover, increased cell-surface SSEA-4 expression is associated with the loss of cell-cell interactions and the gain of a migratory phenotype, suggesting an important role of SSEA-4 in cancer invasion by influencing cellular adhesion to the extracellular matrix.


Asunto(s)
Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Próstata/metabolismo , Antígenos Embrionarios Específico de Estadio/genética , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Claudinas/genética , Claudinas/metabolismo , Cortactina/genética , Cortactina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/patología , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Humanos , Masculino , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Próstata/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Antígenos Embrionarios Específico de Estadio/química , Antígenos Embrionarios Específico de Estadio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
7.
J Immunol ; 190(7): 3417-26, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23460735

RESUMEN

The major human pathogen Staphylococcus aureus has very efficient strategies to subvert the human immune system. Virulence of the emerging community-associated methicillin-resistant S. aureus depends on phenol-soluble modulin (PSM) peptide toxins, which are known to attract and lyse neutrophils. However, their influences on other immune cells remain elusive. In this study, we analyzed the impact of PSMs on dendritic cells (DCs) playing an essential role in linking innate and adaptive immunity. In human neutrophils, PSMs exert their function by binding to the formyl peptide receptor (FPR) 2. We show that mouse DCs express the FPR2 homolog mFPR2 as well as its paralog mFPR1 and that PSMs are chemoattractants for DCs at noncytotoxic concentrations. PSMs reduced clathrin-mediated endocytosis and inhibited TLR2 ligand-induced secretion of the proinflammatory cytokines TNF, IL-12, and IL-6, while inducing IL-10 secretion by DCs. As a consequence, treatment with PSMs impaired the capacity of DCs to induce activation and proliferation of CD4(+) T cells, characterized by reduced Th1 but increased frequency of FOXP3(+) regulatory T cells. These regulatory T cells secreted high amounts of IL-10, and their suppression capacity was dependent on IL-10 and TGF-ß. Interestingly, the induction of tolerogenic DCs by PSMs appeared to be independent of mFPRs, as shown by experiments with mice lacking mFPR2 (mFPR2(-/-)) and the cognate G protein (p110γ(-/-)). Thus, PSMs from highly virulent pathogens affect DC functions, thereby modulating the adaptive immune response and probably increasing the tolerance toward the pathogen.


Asunto(s)
Toxinas Bacterianas/inmunología , Células Dendríticas/inmunología , Péptidos/inmunología , Staphylococcus aureus/inmunología , Linfocitos T Reguladores/inmunología , Animales , Toxinas Bacterianas/química , Quimiotaxis/inmunología , Clatrina/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Endocitosis/inmunología , Femenino , Activación de Linfocitos/inmunología , Ratones , Receptores de Formil Péptido/metabolismo , Staphylococcus aureus/química , Receptor Toll-Like 2/metabolismo
8.
Nature ; 456(7220): 344-9, 2008 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-18849962

RESUMEN

Human primordial germ cells and mouse neonatal and adult germline stem cells are pluripotent and show similar properties to embryonic stem cells. Here we report the successful establishment of human adult germline stem cells derived from spermatogonial cells of adult human testis. Cellular and molecular characterization of these cells revealed many similarities to human embryonic stem cells, and the germline stem cells produced teratomas after transplantation into immunodeficient mice. The human adult germline stem cells differentiated into various types of somatic cells of all three germ layers when grown under conditions used to induce the differentiation of human embryonic stem cells. We conclude that the generation of human adult germline stem cells from testicular biopsies may provide simple and non-controversial access to individual cell-based therapy without the ethical and immunological problems associated with human embryonic stem cells.


Asunto(s)
Células Madre Pluripotentes/citología , Testículo/citología , Adulto , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Pluripotentes/metabolismo , Espermatogonias/citología , Espermatogonias/ultraestructura , Teratoma/patología
9.
Prostate ; 73(14): 1576-90, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836489

RESUMEN

BACKGROUND: Benign prostatic hyperplasia (BPH) and prostate cancer (PCa) are common abnormalities in elderly men. It is considered that epithelial stem cells are involved in the etiology and development of both diseases. To distinguish aberrant from normal cells, the knowledge about primary epithelial stem/progenitor cells (ES/P) is essential. The aim of this study was to examine the role of surface markers to distinguish between different subsets of prostate basal epithelium. METHODS: The expression pattern of prostate tissue single cell suspensions was analyzed by flow cytometry using different markers. Sorted cell populations were examined for their clonogenic capacity and the resulted colonies were analyzed with flow cytometry, Western blot, and qPCR for stem cell, basal, and luminal epithelium markers. Additionally, the histological localization of the examined markers was determined using immunofluorescence. RESULTS: Using the combination of CD49f, Trop-2, and surface CD24, basal cell subsets with distinct differentiation capacities were dissected (CD49f(+) Trop-2(+) CD24(-) and CD49f(+) Trop-2(+) CD24(+) ). Although cells from the two subsets gave rise to similar basal colonies, qPCR of primary tissue revealed that higher levels of basal marker expression were detected in the CD49f(+) Trop-2(+) CD24(-) subset. Immunofluorescence analysis showed a prominent expression of CD24 by luminal and basal cells. CONCLUSIONS: Subsets with distinct differentiation capacities within the basal epithelium (CD49f(+) Trop-2(+) CD24(-) and CD49f(+) Trop-2(+) CD24(+) ) can be distinguished in human prostate. CD24 is a marker expressed on the basal transit-amplifying cells (transition cells) and may play a role in the differentiation and migration of ES/P cells to the luminal layer. The knowledge of this mechanism is of relevance for treatment of both diseases.


Asunto(s)
Antígeno CD24 , Próstata/patología , Hiperplasia Prostática , Neoplasias de la Próstata , Anciano , Antígenos de Diferenciación , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Antígeno CD24/genética , Antígeno CD24/metabolismo , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Expresión Génica , Humanos , Integrina alfa6/metabolismo , Masculino , Próstata/metabolismo , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Células Madre/metabolismo , Células Madre/patología
10.
Prostate ; 73(2): 162-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22715006

RESUMEN

OBJECTIVES: The receptor activator of the NF-kB ligand (RANKL) pathway is a key mediator of prostate cancer (PC)-induced bone disease. However, little is known about this pathway in patients with non-metastatic PC. We aimed to investigate whether changes of RANKL, its inhibitor osteoprotegerin (OPG) and bone marrow-mesenchymal stromal cells (BM-MSCs) occur in PC patients without manifest bone metastases. PATIENTS AND METHODS: We determined OPG and soluble RANKL (sRANKL) in serum and corresponding bone marrow (BM) samples of 140 patients before radical prostatectomy by enzyme-linked immunosorbent assay (ELISA). As control serum samples of 50 patients with benign prostate hyperplasia were analyzed. BM mononuclear cells (BMNCs) of 16 PC patients were analyzed for expression of RANKL and CD271 (as marker for MSCs) by flow cytometry. RESULTS: PC patients had significantly lower serum levels of OPG compared to BPH patients (P = 0.007), whereas no differences were observed for serum sRANKL (P = 0.74). Both OPG and sRANKL concentrations of serum and corresponding BM samples correlated significantly (P < 0.0001 each). Interestingly, in PC patients, lower serum and BM OPG levels were associated with a higher proportion of BM-MSCs (P = 0.04 and 0.0016, respectively). No correlations were observed for sRANKL, OPG, BM-MSCs, and established risk parameters of PC. DISCUSSION: The results of the study indicate that localized PC is associated with early specific changes of the RANKL pathway in serum and bone marrow (BM). These changes might be part of the pre-metastatic niche of PC and implicate a potential benefit of RANKL inhibition in patients with localized PC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Médula Ósea/metabolismo , Neoplasias Óseas , Neoplasias de la Próstata/metabolismo , Ligando RANK/metabolismo , Transducción de Señal/fisiología , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Médula Ósea/patología , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/diagnóstico , Ligando RANK/sangre
11.
Cancer Immunol Immunother ; 62(9): 1485-97, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23817722

RESUMEN

INTRODUCTION: Lung cancer is the most common cancer worldwide. Every year, as many people die of lung cancer as of breast, colon and rectum cancers combined. Because most patients are being diagnosed in advanced, not resectable stages and therefore have a poor prognosis, there is an urgent need for alternative therapies. Since it has been demonstrated that a high number of tumor- and stromal-infiltrating cytotoxic T cells (CTLs) is associated with an increased disease-specific survival in lung cancer patients, it can be assumed that immunotherapy, e.g. peptide vaccines that are able to induce a CTL response against the tumor, might be a promising approach. METHODS: We analyzed surgically resected lung cancer tissues with respect to HLA class I- and II-presented peptides and gene expression profiles, aiming at the identification of (novel) tumor antigens. In addition, we tested the ability of HLA ligands derived from such antigens to generate a CTL response in healthy donors. RESULTS: Among 170 HLA ligands characterized, we were able to identify several potential targets for specific CTL recognition and to generate CD8+ T cells which were specific for peptides derived from cyclin D1 or protein-kinase, DNA-activated, catalytic polypeptide and lysed tumor cells loaded with peptide. CONCLUSIONS: This is the first molecular analysis of HLA class I and II ligands ex vivo from human lung cancer tissues which reveals known and novel tumor antigens able to elicit a CTL response.


Asunto(s)
Epítopos de Linfocito T/inmunología , Antígenos HLA/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Ciclina D1/inmunología , Proteína Quinasa Activada por ADN/inmunología , Células Dendríticas/inmunología , Expresión Génica , Humanos , Inmunohistoquímica , Inmunoterapia , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Datos de Secuencia Molecular , Proteínas Nucleares/inmunología , Péptidos/inmunología
12.
Mov Disord ; 28(3): 392-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23401086

RESUMEN

BACKGROUND: Emerging evidence has highlighted the pivotal role of the immune system in neurodegenerative diseases. This study investigated the impact of progressive neurodegeneration on the differentiation and development of hematopoietic stem cells in the peripheral blood of Parkinson's patients. METHODS: A colony-forming cell assay was established to study hematopoietic stem cells from venous blood of Parkinson's patients, and flow cytometry was used to analyze the expression of chemokine receptors on monocytes. RESULTS: We demonstrate that there is strong upregulation in the percentage of monocyte precursors in the peripheral blood of Parkinson's patients and asymptomatic high-risk individuals. We identify the receptor CCR2 as undergoing strong upregulation on the surface of classical monocytes in Parkinson's patients. CONCLUSIONS: The association between blood cell development and progressive cell death in the brain of Parkinson's patients should be further investigated as a potential dynamic biomarker and indicator of disease progression.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Monocitos/fisiología , Enfermedad de Parkinson/patología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/inmunología , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Enfermedad de Parkinson/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores CCR2/metabolismo , Estadísticas no Paramétricas
13.
PLoS Pathog ; 6(11): e1001212, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21124820

RESUMEN

CD4(+) T cells are essential for the control of Yersinia enterocolitica (Ye) infection in mice. Ye can inhibit dendritic cell (DC) antigen uptake and degradation, maturation and subsequently T-cell activation in vitro. Here we investigated the effects of Ye infection on splenic DCs and T-cell proliferation in an experimental mouse infection model. We found that OVA-specific CD4(+) T cells had a reduced potential to proliferate when stimulated with OVA after infection with Ye compared to control mice. Additionally, proliferation of OVA-specific CD4(+) T cells was markedly reduced when cultured with splenic CD8α(+) DCs from Ye infected mice in the presence of OVA. In contrast, T-cell proliferation was not impaired in cultures with CD4(+) or CD4(-)CD8α(-) DCs isolated from Ye infected mice. However, OVA uptake and degradation as well as cytokine production were impaired in CD8α(+) DCs, but not in CD4(+) and CD4(-)CD8α(-) DCs after Ye infection. Pathogenicity factors (Yops) from Ye were most frequently injected into CD8α(+) DCs, resulting in less MHC class II and CD86 expression than on non-injected CD8α(+) DCs. Three days post infection with Ye the number of splenic CD8α(+) and CD4(+) DCs was reduced by 50% and 90%, respectively. The decreased number of DC subsets, which was dependent on TLR4 and TRIF signaling, was the result of a faster proliferation and suppressed de novo DC generation. Together, we show that Ye infection negatively regulates the stimulatory capacity of some but not all splenic DC subpopulations in vivo. This leads to differential antigen uptake and degradation, cytokine production, cell loss, and cell death rates in various DC subpopulations. The data suggest that these effects might be caused directly by injection of Yops into DCs and indirectly by affecting the homeostasis of CD4(+) and CD8α(+) DCs. These events may contribute to reduced T-cell proliferation and immune evasion of Ye.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Evasión Inmune/inmunología , Factores de Virulencia/metabolismo , Yersiniosis/inmunología , Yersinia enterocolitica/inmunología , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Animales , Presentación de Antígeno , Western Blotting , Linfocitos T CD4-Positivos/microbiología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas para Inmunoenzimas , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Bazo/inmunología , Bazo/microbiología , Receptor Toll-Like 4/fisiología , Factores de Virulencia/genética , Yersiniosis/metabolismo , Yersiniosis/microbiología
14.
Acta Neuropathol ; 124(1): 83-97, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22249620

RESUMEN

Glioblastoma (GBM), the most common malignant brain tumor, is among the most lethal neoplasms, with a median survival of approximately 1 year. Prognosis is poor since GBMs possess a strong migratory and highly invasive potential, making complete surgical resection impossible. Reduced expression of carboxypeptidase E (CPE), a neuropeptide-processing enzyme, in a cell death-resistant glioma cell line and lower CPE expression levels in the cohort of GBM samples of The Cancer Genome Atlas compared to normal brain control specimens prompted us to analyze the function of CPE as a putative tumor suppressor gene. In our samples, CPE was also reduced in GBM compared to normal brain with the strongest loss in cells surrounding hypoxic tumor areas as well as in most glioma cell lines and primary glioma cells. In our cohort of glioma patients, loss of CPE predominantly occurred in glioblastomas and was associated with worse prognosis. In glioma cells, CPE overexpression was significantly reduced, whereas knockdown or inhibition enhanced glioma cell migration and invasion. The decreased migratory potential following CPE overexpression was paralleled by altered cellular morphology, promoting a transition to focal adhesions and associated stress fibers. In contrast to the decreased migration, high CPE levels were associated with higher proliferative rates. As microenvironmental regulation cues, we identified CPE as being downregulated upon hypoxia or glucose deprivation. Our findings indicate an oxygen- and nutrition-dependent anti-migratory, but pro-proliferative role of CPE in gliomas with prognostic impact for patient survival, thereby contributing to the understanding of the "go or grow" hypothesis in gliomas.


Asunto(s)
Neoplasias Encefálicas/enzimología , Carboxipeptidasa H/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Glioma/enzimología , Estrés Fisiológico/fisiología , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Carbocianinas/metabolismo , Carboxipeptidasa H/genética , Adhesión Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioma/mortalidad , Glioma/patología , Glucosa/deficiencia , Humanos , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Estrés Fisiológico/efectos de los fármacos , Succinatos/farmacología , Análisis de Supervivencia , Factores de Tiempo
15.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35288466

RESUMEN

BACKGROUND: In lymphoid malignancies, the introduction of chimeric antigen receptor T (CAR-T) cells and bispecific antibodies (bsAbs) has achieved remarkable clinical success. However, such immunotherapeutic strategies are not yet established for acute myeloid leukemia (AML), the most common form of acute leukemia in adults. Common targets in AML such as CD33, CD123, and CLEC12A are highly expressed on both AML blasts and on normal myeloid cells and hematopoietic stem cells (HSCs), thereby raising toxicity concerns. In B-cell acute lymphoblastic leukemia (B-ALL), bsAbs and CAR-T therapy targeting CD19 and CD22 have demonstrated clinical success, but resistance via antigen loss is common, motivating the development of agents focused on alternative targets. An attractive emerging target is FLT3, a proto-oncogene expressed in both AML and B-ALL, with low and limited expression on myeloid dendritic cells and HSCs. METHODS: We developed and characterized CLN-049, a T cell-activating bsAb targeting CD3 and FLT3, constructed as an IgG heavy chain/scFv fusion. CLN-049 binds the membrane proximal extracellular domain of the FLT3 protein tyrosine kinase, which facilitates the targeting of leukemic blasts regardless of FLT3 mutational status. CLN-049 was evaluated for preclinical safety and efficacy in vitro and in vivo. RESULTS: CLN-049 induced target-restricted activation of CD4+ and CD8+ T cells. AML cell lines expressing a broad range of surface levels of FLT3 were efficiently lysed on treatment with subnanomolar concentrations of CLN-049, whereas FLT3-expressing hematopoietic progenitor cells and dendritic cells were not sensitive to CLN-049 killing. Treatment with CLN-049 also induced lysis of AML and B-ALL patient blasts by autologous T cells at the low effector-to-target ratios typically observed in patients with overt disease. Lysis of leukemic cells was not affected by supraphysiological levels of soluble FLT3 or FLT3 ligand. In mouse xenograft models, CLN-049 was highly active against human leukemic cell lines and patient-derived AML and B-ALL blasts. CONCLUSIONS: CLN-049 has a favorable efficacy and safety profile in preclinical models, warranting evaluation of its antileukemic activity in the clinic.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Humanos , Inmunoglobulina G/uso terapéutico , Inmunoterapia Adoptiva , Subunidad alfa del Receptor de Interleucina-3 , Lectinas Tipo C , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Receptores Mitogénicos
16.
NPJ Regen Med ; 6(1): 33, 2021 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103536

RESUMEN

Accurately defining hierarchical relationships between human stem cells and their progeny, and using this knowledge for new cellular therapies, will undoubtedly lead to further successful treatments for life threatening and chronic diseases, which represent substantial burdens on patient quality of life and to healthcare systems globally. Clinical translation relies in part on appropriate biomarker, in vitro manipulation and transplantation strategies. CD164 has recently been cited as an important biomarker for enriching both human haematopoietic and skeletal stem cells, yet a thorough description of extant human CD164 monoclonal antibody (Mab) characteristics, which are critical for identifying and purifying these stem cells, was not discussed in these articles. Here, we highlight earlier but crucial research describing these relevant characteristics, including the differing human CD164 Mab avidities and their binding sites on the human CD164 sialomucin, which importantly may affect subsequent stem cell function and fate.

17.
Bone Marrow Transplant ; 56(10): 2336-2354, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33976380

RESUMEN

Detrimental graft-versus-host disease (GVHD) still remains a major cause of death in hematopoietic stem cell transplantation (HSCT). The recently explored depletion of naive cells from mobilized grafts (CD45RA depletion) has shown considerable promise, yet is unable to eliminate the incidence of GVHD. Analysis of CD45RA-depleted haploidentical mixed lymphocytes culture (haplo-MLC) revealed insufficient suppression of alloresponses in the CD4+ compartment and identified CD276 as a marker for alloreactive memory Th1 T cells. Conclusively, depleting CD276+ cells from CD45RA-depleted haplo-MLC significantly attenuated alloreactivity to recipient cells while increasing antiviral reactivity and maintaining anti-third party reactivity in vitro. To evaluate these findings in vivo, bulk, CD45RA-depleted, or CD45RA/CD276-depleted CD4+ T cells from HLA-DR4negative healthy humans were transplanted into NSG-Ab°DR4 mice, a sensitive human allo-GVHD model. Compellingly, CD45RA/CD276-depleted grafts from HLA-DR4negative donors or in vivo depletion of CD276+ cells after transplant of HLA-DR4negative memory CD4 T cells significantly delay the onset of GVHD symptoms and significantly alleviate its severity in NSG-Ab°DR4 mice. The clinical courses correlated with diminished Th1-cytokine secretion and downregulated CXCR6 expression of engrafted peripheral T cells. Collectively, mismatched HLA-mediated GVHD can be controlled by depleting recipient-specific CD276+ alloreacting T cells from the graft, highlighting its application in haplo-HSCT.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Memoria Inmunológica , Depleción Linfocítica , Ratones , Linfocitos T
18.
J Cell Physiol ; 225(1): 123-31, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20458727

RESUMEN

Mesenchymal stem cells (MSCs) are self-renewing cells with the ability to differentiate into various mesodermal-derived tissues. Recently, we have identified in adult human periodontal ligament (PDL) a population of stem cells (PDL-MSCs) with the ability to differentiate into osteoblasts and adipocytes. The aim of the present work was to further characterize this population and the expression profile of its cells. To achieve our objective we have used flow cytometry, magnetic cell sorting, cytokine antibody array, and light and electron microscope immunostaining. Our results show that the PDL-MSCs contain a subpopulation of frizzled-9 (CD349) positive cells expressing a panel of key mesenchymal and embryonic markers including CD10, CD26, CD29, CD44, CD73, CD90, CD105, CD166, SSEA-1, and SSEA-4. They are additionally positive for nanog and Oct-4; two critical transcription factors directing self-renewal and pluripotency of embryonic stem cells, and they also express the cytokines EGF and IP-10. The presence of nanog, Oct-4, SSEA-1, and SSEA-4 suggests that PDL-MSCs are less differentiated than bone marrow-derived MSCs. Taken together, these data indicate the presence of immature MSCs in PDL and suggest that the frizzled-9/Wnt pathway plays an important role in regulating proliferation and differentiation of these cells.


Asunto(s)
Receptores Frizzled/metabolismo , Proteínas de Homeodominio/metabolismo , Antígeno Lewis X/metabolismo , Células Madre Mesenquimatosas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ligamento Periodontal/citología , Receptores Acoplados a Proteínas G/metabolismo , Antígenos Embrionarios Específico de Estadio/metabolismo , Adulto , Biomarcadores/metabolismo , Proliferación Celular , Separación Celular/métodos , Forma de la Célula , Células Cultivadas , Citocinas/metabolismo , Receptores Frizzled/genética , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Antígeno Lewis X/genética , Células Madre Mesenquimatosas/citología , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Receptores Acoplados a Proteínas G/genética , Antígenos Embrionarios Específico de Estadio/genética
19.
Cell Physiol Biochem ; 26(6): 1073-80, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21220938

RESUMEN

Human jaw periosteum-derived cells (JPCs) represent an alternative cell source to bone marrow-derived mesenchymal stem cells for tissue engineering applications in the oral and maxillofacial surgery. In this study we investigated how far the presence or expression of human mesenchymal stem cell antigen-1/tissue non-specific alkaline phosphatase (MSCA-1/TNAP) and LNGFR (CD271) can be utilized to select and enrich the osteogenic progenitor cell fraction from the entire JPC population. Depending on their mineralization capacity, we classified the human isolated JPCs into mineralizing (mJPCs) and non-mineralizing JPCs (nmJPCs). Flow cytometric analyses revealed that undifferentiated mJPCs expressed MSCA-1/TNAP at significant higher levels than nmJPCs at day 5 and 10 of osteogenesis. Western blot analyses showed increased MSCA-1/TNAP expression levels in mJPCs during osteogenesis, whereas in nmJPCs MSCA-1/TNAP expression remained undetectable. Using the MSCA-1 and LNGFR specific antibodies, we separated the positive and negative fractions from the entire mJPC population. In order to analyse the mineralization capacity of the MSCA-1(+) and LNGFR(+) cell subsets, we quantified the calcium deposition in both subpopulations in comparison to the respective negative subpopulations. The MSCA-1(+)/TNAP(+) cell fraction showed a significant higher osteogenic capacity compared to the MSCA-1-/TNAP- cell fraction whereas the LNGFR(+/-) cell fractions did not differ in their osteogenic potential. Our findings suggest that MSCA-1 may represent a promising osteogenic marker for mJPC.


Asunto(s)
Antígenos de Superficie/metabolismo , Proteínas Portadoras/metabolismo , Maxilares/metabolismo , Periostio/metabolismo , Adapaleno , Fosfatasa Alcalina , Biomarcadores/metabolismo , Calcificación Fisiológica/fisiología , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Humanos , Maxilares/citología , Naftalenos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Osteogénesis/fisiología , Periostio/citología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Ingeniería de Tejidos
20.
Sci Rep ; 10(1): 11145, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32636407

RESUMEN

Osteoarthritis (OA), the most common joint disorder, is characterised by progressive structural changes in both the cartilage and the underlying subchondral bone. In late disease stages, subchondral bone sclerosis has been linked to heightened osteogenic commitment of bone marrow stromal cells (BMSCs). This study utilised cell sorting and immunohistochemistry to identify a phenotypically-distinct, osteogenically-committed BMSC subset in human OA trabecular bone. Femoral head trabecular bone tissue digests were sorted into CD45-CD271+CD56+CD146-, CD45-CD271+CD56-CD146+ and CD45-CD271+CD56-CD146-(termed double-negative, DN) subsets, and CD45+CD271-hematopoietic-lineage cells served as control. Compared to the CD146+ subset, the CD56+ subset possessed a lower-level expression of adipocyte-associated genes and significantly over 100-fold higher-level expression of many osteoblast-related genes including osteopontin and osteocalcin, whilst the DN subset presented a transcriptionally 'intermediate' BMSC population. All subsets were tri-potential following culture-expansion and were present in control non-OA trabecular bone. However, while in non-OA bone CD56+ cells only localised on the bone surface, in OA bone they were additionally present in the areas of new bone formation rich in osteoblasts and newly-embedded osteocytes. In summary, this study reveals a distinct osteogenically-committed CD271+CD56+ BMSC subset and implicates it in subchondral bone sclerosis in hip OA. CD271+CD56+ subset may represent a future therapeutic target for OA and other bone-associated pathologies.


Asunto(s)
Antígeno CD56/metabolismo , Cabeza Femoral/metabolismo , Células Madre Mesenquimatosas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Osteoartritis/metabolismo , Osteogénesis , Receptores de Factor de Crecimiento Nervioso/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígeno CD56/fisiología , Hueso Esponjoso/metabolismo , Hueso Esponjoso/patología , Estudios de Casos y Controles , Femenino , Cabeza Femoral/patología , Citometría de Flujo , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Proteínas del Tejido Nervioso/fisiología , Osteoartritis/patología , Osteogénesis/fisiología , Receptores de Factor de Crecimiento Nervioso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA