Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci Res ; 99(9): 2228-2249, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34060113

RESUMEN

The formation of the cerebellum is highly coordinated to obtain its characteristic morphology and all cerebellar cell types. During mouse postnatal development, cerebellar progenitors with astroglial-like characteristics generate mainly astrocytes and oligodendrocytes. However, a subset of astroglial-like progenitors found in the prospective white matter (PWM) produces astroglia and interneurons. Characterizing these cerebellar astroglia-like progenitors and distinguishing their developmental fates is still elusive. Here, we reveal that astrocyte cell surface antigen-2 (ACSA-2), lately identified as ATPase, Na+/K+ transporting, beta 2 polypeptide, is expressed by glial precursors throughout postnatal cerebellar development. In contrast to common astrocyte markers, ACSA-2 appears on PWM cells but is absent on Bergmann glia (BG) precursors. In the adult cerebellum, ACSA-2 is broadly expressed extending to velate astrocytes in the granular layer, white matter astrocytes, and to a lesser extent to BG. Cell transplantation and transcriptomic analysis revealed that marker staining discriminates two postnatal progenitor pools. One subset is defined by the co-expression of ACSA-2 and GLAST and the expression of markers typical of parenchymal astrocytes. These are PWM precursors that are exclusively gliogenic. They produce predominantly white matter and granular layer astrocytes. Another subset is constituted by GLAST positive/ACSA-2 negative precursors that express neurogenic and BG-like progenitor genes. This population displays multipotency and gives rise to interneurons besides all glial types, including BG. In conclusion, this work reports about ACSA-2, a marker that in combination with GLAST enables for the discrimination and isolation of multipotent and glia-committed progenitors, which generate different types of cerebellar astrocytes.


Asunto(s)
Antígenos de Superficie/análisis , Cerebelo/química , Cerebelo/citología , Transportador 1 de Aminoácidos Excitadores/análisis , Células Madre Multipotentes/química , Neuroglía/química , Animales , Animales Recién Nacidos , Femenino , Separación Inmunomagnética/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroglía/clasificación , Análisis de Secuencia de ARN/métodos
2.
Breast Cancer Res ; 17(1): 146, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26607327

RESUMEN

INTRODUCTION: Chemotherapy resistance resulting in incomplete pathologic response is associated with high risk of metastasis and early relapse in breast cancer. The aim of this study was to identify and evaluate biomarkers of treatment-resistant tumor cells. METHODS: We performed a cell surface marker screen in triple-negative breast cancer patient-derived xenograft models treated with standard care genotoxic chemotherapy. Global expression profiling was used to further characterize the identified treatment-resistant subpopulations. RESULTS: High expression of sialyl-glycolipid stage-specific embryonic antigen 4 (SSEA4) was found in residual tumor cells surviving chemotherapy and in samples from metastatic patients who relapsed after neoadjuvant chemotherapy. Gene and microRNA (miRNA) expression profiling linked SSEA4 positivity with a mesenchymal phenotype and a deregulation of drug resistance pathways. Functional assays demonstrated a direct link between epithelial-mesenchymal transition (EMT) and SSEA4 expression. Interestingly, SSEA4 expression, EMT, and drug resistance seemed to be regulated posttranscriptionally. Finally, high expression of CMP-N-acetylneuraminate-ß-galactosamide-α-2,3-sialyltransferase 2 (ST3GAL2), the rate-limiting enzyme of SSEA4 synthesis, was found to be associated with poor clinical outcome in breast and ovarian cancer patients treated with chemotherapy. CONCLUSIONS: In this study, we identified SSEA4 as highly expressed in a subpopulation of tumor cells resistant to multiple commonly used chemotherapy drugs, as well as ST3GAL2, the rate-limiting enzyme of SSEA4 synthesis, as a predictive marker of poor outcome for breast and ovarian cancer patients undergoing chemotherapy. Both biomarkers and additionally identified regulatory miRNAs may be used to further understand chemoresistance, to stratify patient groups in order to avoid ineffective and painful therapies, and to develop alternative treatment regimens for breast cancer patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Antígenos Embrionarios Específico de Estadio/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Ratones , Trasplante de Neoplasias
4.
Eur J Immunol ; 43(11): 2993-3005, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922217

RESUMEN

Plasmacytoid dendritic cells (pDCs) play an important role in innate and adaptive immunity and were shown to be identical to previously described natural interferon (IFN)-α-producing cells. Here, we describe two functionally distinct pDC subpopulations that are characterized by the differential expression of stem cell antigen-1 (Sca-1; Ly-6A/E). Sca-1(-) pDCs are mainly found in the BM, appear first during development, show a higher proliferative activity, and represent the more precursor phenotype. Sca-1(+) pDCs are mostly located in secondary lymphoid organs and represent a later developmental stage. Sca-1(-) pDCs give rise to an Sca-1(+) subset upon activation or in response to endogenous type I IFN. Interestingly, in contrast to Sca-1(-) pDCs, Sca-1(+) pDCs are defective in IFN-α production upon endosomal TLR9 stimulation, whereas lysosomal signaling via TLR9 is functional in both subsets. Gene expression analysis revealed that osteopontin is strongly upregulated in Sca-1(-) pDCs. These data provide evidence for the molecular basis of the observed functional heterogeneity, as the intracellular isoform of osteopontin couples TLR9 signaling to IFN-α expression. Taken together, our results indicate that Sca-1(-) pDCs are an early developmental stage of pDCs with distinct innate functions representing the true murine natural IFN-α-producing cells.


Asunto(s)
Antígenos Ly/genética , Células Dendríticas/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Receptor Toll-Like 9/biosíntesis , Animales , Antígenos Ly/biosíntesis , Proliferación Celular , Células Dendríticas/inmunología , Femenino , Expresión Génica , Interferón-alfa/biosíntesis , Activación de Linfocitos/inmunología , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Osteopontina/biosíntesis , Transducción de Señal/inmunología , Regulación hacia Arriba
5.
Mol Ther ; 21(2): 409-22, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22929659

RESUMEN

Mouse monocytes exposed to macrophage colony-stimulating factor (M-CSF) and interferon-γ (IFN-γ) were driven to a novel suppressor phenotype. These regulatory macrophages (M regs) expressed markers distinguishing them from M0-, M1-, and M2-polarized macrophages and monocyte-derived dendritic cells (DCs). M regs completely suppressed polyclonal T cell proliferation through an inducible nitric oxide synthase (iNOS)-dependent mechanism. Additionally, M regs eliminated cocultured T cells in an allospecific fashion. In a heterotopic heart transplant model, a single intravenous administration of 5 × 10(6) donor-strain M regs before transplantation significantly prolonged allograft survival in fully immunocompetent recipients using both the stringent C3H-to-BALB/c (32.6 ± 4.5 versus 8.7 ± 0.2 days) and B6-to-BALB/c (31.1 ± 12 versus 9.7 ± 0.4 days) strain combinations. Nos2-deficient M regs did not prolong allograft survival, proving that M reg function in vivo is iNOS-dependent and mediated by living cells. M regs were detectable for at least 2 weeks postinfusion in allogeneic recipients. In their origin, development, phenotypic relationship with other in vitro-derived macrophages and functions, there are solid grounds to assert a near-equivalence of mouse and human M regs. It is concluded that mouse M regs represent a novel, phenotypically distinct subset of suppressor macrophages. Clinical applications of M reg therapy as an adjunct immunosuppressive therapy are currently being investigated within The ONE Study.


Asunto(s)
Supervivencia de Injerto , Trasplante de Corazón , Terapia de Inmunosupresión/métodos , Interferón gamma/farmacología , Macrófagos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Animales , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Trasplante de Corazón/métodos , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Análisis por Micromatrices , Monocitos/citología , Monocitos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Trasplante Homólogo
6.
J Immunol ; 187(5): 2072-8, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21804023

RESUMEN

Regulatory macrophages (M regs) were administered to two living-donor renal transplant recipients. Both patients were minimized to low-dose tacrolimus monotherapy within 24 wk of transplantation and subsequently maintained excellent graft function. After central venous administration, most M regs remained viable and were seen to traffic from the pulmonary vasculature via the blood to liver, spleen, and bone marrow. By 1 y posttransplantation, both patients displayed patterns of peripheral blood gene expression converging upon the IOT-RISET signature. Furthermore, both patients maintained levels of peripheral blood FOXP3 and TOAG-1 mRNA expression within the range consistent with nonrejection. It is concluded that M regs warrant further study as a potential immune-conditioning therapy for use in solid-organ transplantation. The results of this work are being used to inform the design of The ONE Study, a multinational clinical trial of immunomodulatory cell therapy in renal transplantation.


Asunto(s)
Movimiento Celular , Quimiotaxis de Leucocito/inmunología , Rechazo de Injerto/prevención & control , Inmunoterapia/métodos , Trasplante de Riñón/inmunología , Macrófagos/citología , Separación Celular , Femenino , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Rechazo de Injerto/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/trasplante , Masculino , Persona de Mediana Edad , Linfocitos T/inmunología , Adulto Joven
7.
Stem Cells ; 29(5): 847-57, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21394831

RESUMEN

MicroRNAs (miRNAs) have been shown to play an important role in hematopoiesis. To elucidate the role of miRNAs in the early steps of hematopoiesis, we directly compared donor-matched CD133(+) cells with the more differentiated CD34(+) CD133(-) and CD34(-) CD133(-) cells from bone marrow on the miRNA and mRNA level. Using quantitative whole genome miRNA microarray and sequencing-based profiling, we found that between 109 (CD133(+) ) and 216 (CD34(-) CD133(-) ) miRNAs were expressed. Quantification revealed that the 25 highest expressed miRNAs accounted for 73% of the total miRNA pool. miR-142-3p was the highest expressed miRNA with up to 2,000 copies per cell in CD34(+) CD133(-) cells. Eighteen miRNAs were significantly differentially expressed between CD133(+) and CD34(+) CD133(-) cells. We analyzed their biological role by examining the coexpression of miRNAs and its bioinformatically predicted mRNA targets and luciferase-based reporter assays. We provide the first evidence for a direct regulation of CD133 by miR-142-3p as well as tropomyosin 1 and frizzled homolog 5 by miR-29a. Overexpression of miRNAs in CD133(+) cells demonstrated that miR-142-3p has a negative influence on the overall colony-forming ability. In conclusion, the miRNAs expressed differentially between the CD133(+) and CD34(+) CD133(-) cells are involved in inhibition of differentiation, prevention of apoptosis, and cytoskeletal remodeling. These results are highly relevant for stem cell-based therapies with CD133(+) cells and delineate for the first time how the stem cell character of CD133(+) cells is defined by the expression of specific miRNAs.


Asunto(s)
Antígenos CD34/metabolismo , Antígenos CD/metabolismo , Diferenciación Celular/fisiología , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , MicroARNs/genética , Péptidos/metabolismo , ARN Mensajero/genética , Antígeno AC133 , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Humanos , MicroARNs/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , ARN Mensajero/fisiología
8.
RNA ; 15(12): 2375-84, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19861428

RESUMEN

MicroRNAs (miRNAs) are a species of small RNAs approximately 21-23-nucleotides long that have been shown to play an important role in many different cellular, developmental, and physiological processes. Accordingly, numerous PCR-, sequencing-, or hybridization-based methods have been established to identify and quantify miRNAs. Their short length results in a high dynamic range of melting temperatures and therefore impedes a proper selection of detection probes or optimized PCR primers. While miRNA microarrays allow for massive parallel and accurate relative measurement of all known miRNAs, they have so far been less useful as an assay for absolute quantification. Here, we present a microarray-based approach for global and absolute quantification of miRNAs. The method relies on the parallel hybridization of the sample of interest labeled with Cy5 and a universal reference of 954 synthetic miRNAs in equimolar concentrations that are labeled with Cy3 on a microarray slide containing probes for all human, mouse, rat, and viral miRNAs (miRBase 12.0). Each single miRNA is quantified with respect to the universal reference canceling biases related to sequence, labeling, or hybridization. We demonstrate the accuracy of the method by various spike-in experiments. Furthermore, we quantified miRNA copy numbers in liver samples and CD34(+)/CD133(-) hematopoietic progenitor cells.


Asunto(s)
MicroARNs/análisis , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Viral/análisis , Animales , Secuencia de Bases , Dosificación de Gen , Células Madre Hematopoyéticas/química , Humanos , Hígado/química , Ratones , Ratas
9.
Nat Commun ; 12(1): 1453, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674603

RESUMEN

A major roadblock prohibiting effective cellular immunotherapy of pancreatic ductal adenocarcinoma (PDAC) is the lack of suitable tumor-specific antigens. To address this challenge, here we combine flow cytometry screenings, bioinformatic expression analyses and a cyclic immunofluorescence platform. We identify CLA, CD66c, CD318 and TSPAN8 as target candidates among 371 antigens and generate 32 CARs specific for these molecules. CAR T cell activity is evaluated in vitro based on target cell lysis, T cell activation and cytokine release. Promising constructs are evaluated in vivo. CAR T cells specific for CD66c, CD318 and TSPAN8 demonstrate efficacies ranging from stabilized disease to complete tumor eradication with CD318 followed by TSPAN8 being the most promising candidates for clinical translation based on functionality and predicted safety profiles. This study reveals potential target candidates for CAR T cell based immunotherapy of PDAC together with a functional set of CAR constructs specific for these molecules.


Asunto(s)
Adenocarcinoma/metabolismo , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Inmunoterapia/métodos , Neoplasias Pancreáticas/metabolismo , Tetraspaninas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/terapia , Animales , Antígenos de Neoplasias/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Citocinas/metabolismo , Proteínas Ligadas a GPI/metabolismo , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Factores Inmunológicos , Activación de Linfocitos , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Linfocitos T/inmunología , Tetraspaninas/genética , Neoplasias Pancreáticas
10.
Haematologica ; 95(12): 2134-43, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20713457

RESUMEN

BACKGROUND: CD4(+) regulatory T cells are a specialized subset of T cells that actively control immune responses. Several experimental protocols have been used to expand natural regulatory T cells and to generate adaptive type 1 regulatory T cells for regulatory T-cell-based therapies. DESIGN AND METHODS: The ability of exogenous recombinant human interleukin-10 to induce alloantigen-specific anergy in T cells was investigated and compared to that of interleukin-10 derived from tolerogenic dendritic cells, in mixed lymphocyte cultures. A detailed characterization of the effector functions of the resulting anergized T cells is reported. RESULTS: Interleukin-10, whether exogenous or derived from tolerogenic dendritic cells, induces a population of alloantigen-specific T cells (interleukin-10-anergized T cells) containing type 1 regulatory T cells, which are anergic and actively suppress alloantigen-specific effector T cells present within the mixed population. Interleukin-10-induced anergy is transforming growth factor-ß independent, and is associated with a decreased frequency of alloantigen-specific cytotoxic T lymphocyte precursors, but interleukin-10-anergized T cells are still responsive to third-party, bacterial, and viral antigens. Tolerogenic dendritic cells are more powerful than exogenous interleukin-10 in generating type 1 regulatory T-cell precursors, and are also effective in the context of HLA-matched donors. CONCLUSIONS: Based on these studies, we have developed an efficient and reproducible in vitro method to generate antigen-specific type 1 regulatory T-cell precursors starting from total peripheral blood cells with minimal cell manipulation and suitable for generating type 1 regulatory T cells for regulatory T-cell-based therapies.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Anergia Clonal/inmunología , Isoantígenos/inmunología , Linfocitos T/inmunología , Candida albicans/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Anergia Clonal/efectos de los fármacos , Citomegalovirus/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Interleucina-10/farmacología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Monocitos/inmunología , Monocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Toxoide Tetánico/inmunología , Factor de Crecimiento Transformador beta/farmacología
11.
Nat Commun ; 10(1): 2263, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118448

RESUMEN

All memory T cells mount an accelerated response on antigen reencounter, but significant functional heterogeneity is present within the respective memory T-cell subsets as defined by CCR7 and CD45RA expression, thereby warranting further stratification. Here we show that several surface markers, including KLRB1, KLRG1, GPR56, and KLRF1, help define low, high, or exhausted cytokine producers within human peripheral and intrahepatic CD4+ memory T-cell populations. Highest simultaneous production of TNF and IFN-γ is observed in KLRB1+KLRG1+GPR56+ CD4 T cells. By contrast, KLRF1 expression is associated with T-cell exhaustion and reduced TNF/IFN-γ production. Lastly, TCRß repertoire analysis and in vitro differentiation support a regulated, progressive expression for these markers during CD4+ memory T-cell differentiation. Our results thus help refine the classification of human memory T cells to provide insights on inflammatory disease progression and immunotherapy development.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Hepatopatías/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Similares a Lectina de Células NK/metabolismo , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/metabolismo , Citocinas/inmunología , Humanos , Memoria Inmunológica , Hígado/patología , Hepatopatías/sangre , Hepatopatías/patología , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/inmunología , Receptores Similares a Lectina de Células NK/inmunología
12.
Autophagy ; 15(11): 1899-1916, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30982460

RESUMEN

Infection and inflammation are able to induce diet-independent Na+-accumulation without commensurate water retention in afflicted tissues, which favors the pro-inflammatory activation of mouse macrophages and augments their antibacterial and antiparasitic activity. While Na+-boosted host defense against the protozoan parasite Leishmania major is mediated by increased expression of the leishmanicidal NOS2 (nitric oxide synthase 2, inducible), the molecular mechanisms underpinning this enhanced antibacterial defense of mouse macrophages with high Na+ (HS) exposure are unknown. Here, we provide evidence that HS-increased antibacterial activity against E. coli was neither dependent on NOS2 nor on the phagocyte oxidase. In contrast, HS-augmented antibacterial defense hinged on HIF1A (hypoxia inducible factor 1, alpha subunit)-dependent increased autophagy, and NFAT5 (nuclear factor of activated T cells 5)-dependent targeting of intracellular E. coli to acidic autolysosomal compartments. Overall, these findings suggest that the autolysosomal compartment is a novel target of Na+-modulated cell autonomous innate immunity. Abbreviations: ACT: actins; AKT: AKT serine/threonine kinase 1; ATG2A: autophagy related 2A; ATG4C: autophagy related 4C, cysteine peptidase; ATG7: autophagy related 7; ATG12: autophagy related 12; BECN1: beclin 1; BMDM: bone marrow-derived macrophages; BNIP3: BCL2/adenovirus E1B interacting protein 3; CFU: colony forming units; CM-H2DCFDA: 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate, acetyl ester; CTSB: cathepsin B; CYBB: cytochrome b-245 beta chain; DAPI: 4,6-diamidino-2-phenylindole; DMOG: dimethyloxallyl glycine; DPI: diphenyleneiodonium chloride; E. coli: Escherichia coli; FDR: false discovery rate; GFP: green fluorescent protein; GSEA: gene set enrichment analysis; GO: gene ontology; HIF1A: hypoxia inducible factor 1, alpha subunit; HUGO: human genome organization; HS: high salt (+ 40 mM of NaCl to standard cell culture conditions); HSP90: heat shock 90 kDa proteins; LDH: lactate dehydrogenase; LPS: lipopolysaccharide; Lyz2/LysM: lysozyme 2; NFAT5/TonEBP: nuclear factor of activated T cells 5; MΦ: macrophages; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MFI: mean fluorescence intensity; MIC: minimum inhibitory concentration; MOI: multiplicity of infection; MTOR: mechanistic target of rapamycin kinase; NaCl: sodium chloride; NES: normalized enrichment score; n.s.: not significant; NO: nitric oxide; NOS2/iNOS: nitric oxide synthase 2, inducible; NS: normal salt; PCR: polymerase chain reaction; PGK1: phosphoglycerate kinase 1; PHOX: phagocyte oxidase; RFP: red fluorescent protein; RNA: ribonucleic acid; ROS: reactive oxygen species; sCFP3A: super cyan fluorescent protein 3A; SBFI: sodium-binding benzofuran isophthalate; SLC2A1/GLUT1: solute carrier family 2 (facilitated glucose transporter), member 1; SQSTM1/p62: sequestosome 1; ULK1: unc-51 like kinase 1; v-ATPase: vacuolar-type H+-ATPase; WT: wild type.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/inmunología , Sodio/farmacología , Factores de Transcripción/metabolismo , Animales , Autofagosomas/microbiología , Autofagia/genética , Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Concentración de Iones de Hidrógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación/metabolismo , Lisosomas/genética , Lisosomas/inmunología , Lisosomas/metabolismo , Lisosomas/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Macrófagos/ultraestructura , Manitol/toxicidad , Ratones , Microscopía Electrónica de Transmisión , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Presión Osmótica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sodio/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética
13.
Int J Cancer ; 123(11): 2605-15, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18770863

RESUMEN

Squamous cell carcinoma (SCC) and basal cell carcinoma (BCC) represent the 2 most common types of nonmelanoma skin cancer. Both derive from keratinocytes but show a distinct biological behavior. Here we present transcriptional profiling data of a large cohort of tumor patients (SCC, n = 42; BCC, n = 114). Differentially expressed genes reflect known features of SCC and BCC including the typical cytokeratin pattern as well as upregulation of characteristic cell proliferation genes. Additionally, we found increased expression of interferon (IFN)-regulated genes (including IFI27, IFI30, Mx1, IRF1 and CXCL9) in SCC, and to a lower extent in BCC. The expression of IFN-regulated genes correlated with the extent of the lesional immune-cell infiltrate. Immunohistological examinations confirmed the expression of IFN-regulated genes in association with a CXCR3+ cytotoxic inflammatory infiltrate on the protein level. Of note, a small subset of SCC samples with low expression of IFN-regulated genes included most organ transplant recipients receiving immunosuppressive medication. Collectively, our findings support the concept that IFN-associated host responses play an important role in tumor immunosurveillance in the skin.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Regulación Neoplásica de la Expresión Génica/genética , Interferones/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Transcripción Genética/genética , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Proliferación Celular , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Queratinas/genética , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología , Neoplasias Basocelulares/genética , Neoplasias Basocelulares/inmunología , Neoplasias Basocelulares/patología , Neoplasias Cutáneas/patología
14.
Nat Commun ; 9(1): 2858, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-30030423

RESUMEN

Human regulatory macrophages (Mreg) have shown early clinical promise as a cell-based adjunct immunosuppressive therapy in solid organ transplantation. It is hypothesised that recipient CD4+ T cell responses are actively regulated through direct allorecognition of donor-derived Mregs. Here we show that human Mregs convert allogeneic CD4+ T cells to IL-10-producing, TIGIT+ FoxP3+-induced regulatory T cells that non-specifically suppress bystander T cells and inhibit dendritic cell maturation. Differentiation of Mreg-induced Tregs relies on multiple non-redundant mechanisms that are not exclusive to interaction of Mregs and T cells, including signals mediated by indoleamine 2,3-dioxygenase, TGF-ß, retinoic acid, Notch and progestagen-associated endometrial protein. Preoperative administration of donor-derived Mregs to living-donor kidney transplant recipients results in an acute increase in circulating TIGIT+ Tregs. These results suggest a feed-forward mechanism by which Mreg treatment promotes allograft acceptance through rapid induction of direct-pathway Tregs.


Asunto(s)
Macrófagos/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T Reguladores/inmunología , Aloinjertos , Animales , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/metabolismo , Rechazo de Injerto , Humanos , Interleucina-10/metabolismo , Trasplante de Riñón , Receptores de Lipopolisacáridos/metabolismo , Ratones , Fenotipo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Trasplante Homólogo
15.
PLoS One ; 12(1): e0169624, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28085915

RESUMEN

BACKGROUND: There is high medical need for safe long-term immunosuppression monotherapy in kidney transplantation. Selective targeting of post-transplant alloantigen-(re)activated effector-T cells by anti-TNF antibodies after global T cell depletion may allow safe drug minimization, however, it is unsolved what might be the best maintenance monotherapy. METHODS: In this open, prospective observational single-centre trial, 20 primary deceased donor kidney transplant recipients received 2x20 mg Alemtuzumab (d0/d1) followed by 5 mg/kg Infliximab (d2). For 14 days all patients received only tacrolimus, then they were allocated to either receive tacrolimus (TAC, n = 13) or sirolimus (SIR, n = 7) monotherapy, respectively. Protocol biopsies and extensive immune monitoring were performed and patients were followed-up for 60 months. RESULTS: TAC-monotherapy resulted in excellent graft survival (5yr 92%, 95%CI: 56.6-98.9) and function, normal histology, and no proteinuria. Immune monitoring revealed low intragraft inflammation (urinary IP-10) and hints for the development of operational tolerance signature in the TAC- but not SIR-group. Remarkably, the TAC-monotherapy was successful in all five presensitized (ELISPOT+) patients. However, recruitment into SIR-arm was stopped (after n = 7) because of high incidence of proteinuria and acute/chronic rejection in biopsies. No opportunistic infections occurred during follow-up. CONCLUSIONS: In conclusion, our novel fast-track TAC-monotherapy protocol is likely to be safe and preliminary results indicated an excellent 5-year outcome, however, a full-scale study will be needed to confirm our findings. TRIAL REGISTRATION: EudraCT Number: 2006-003110-18.


Asunto(s)
Glicoproteínas/antagonistas & inhibidores , Rechazo de Injerto/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Trasplante de Riñón/efectos adversos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto , Antígenos CD , Antígenos de Neoplasias , Biomarcadores/metabolismo , Antígeno CD52 , Femenino , Perfilación de la Expresión Génica , Rechazo de Injerto/etiología , Humanos , Masculino , Persona de Mediana Edad , Monitorización Inmunológica , Estudios Prospectivos , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico , Adulto Joven
16.
Int J Mol Med ; 18(3): 395-403, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16865223

RESUMEN

Renal cell carcinoma (RCC) is known to effectively prevent immune recognition. However, little is known about the mechanisms that underlie this phenomenon. Thus, the identification of immunogenic molecules associated with RCC and the elucidation of the corresponding signaling pathways are crucial to the development of effective treatments. We performed transcriptional and functional profiling with cDNA microarrays (1070 cDNA probes) on a total of 17 RCCs, 11 clear cell and 6 papillary, and on corresponding normal tissue. Samples were clustered based on their expression profiles. We found a total of 45 genes to be regulated equally by both tumor types compared to the normal tissue. A set of 13 differentially expressed genes was identified between the examined tumor subtypes. Functional analysis was performed for both gene sets and showed a significant enrichment of cell surface genes regulated in both tumor subtypes. Within these we found five surface marker genes to be upregulated (TNFRSF10B, CD70, TNFR1, PDGFRB, and BAFF) which are involved in immune responses via the regulation of lymphocytes and can also induce apoptosis. Their overexpression in both tumor subtypes suggests a possible involvement in the immune escape strategies of RCC. The combination of transcriptional and functional profiling revealed potential target molecules for novel therapy strategies that must be studied in more detail.


Asunto(s)
Carcinoma Papilar/genética , Carcinoma de Células Renales/genética , Perfilación de la Expresión Génica , Neoplasias Renales/genética , Análisis por Conglomerados , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
17.
J Vis Exp ; (113)2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27501218

RESUMEN

The use of in vitro cell line models for cancer research has been a useful tool. However, it has been shown that these models fail to reliably mimic patient tumors in different assays(1). Human tumor xenografts represent the gold standard with respect to tumor biology, drug discovery, and metastasis research (2-4). Tumor xenografts can be derived from different types of material like tumor cell lines, tumor tissue from primary patient tumors(4) or serially transplanted tumors. When propagated in vivo, xenografted tissue is infiltrated and vascularized by cells of mouse origin. Multiple factors such as the tumor entity, the origin of xenografted material, growth rate and region of transplantation influence the composition and the amount of mouse cells present in tumor xenografts. However, even when these factors are kept constant, the degree of mouse cell contamination is highly variable. Contaminating mouse cells significantly impair downstream analyses of human tumor xenografts. As mouse fibroblasts show high plating efficacies and proliferation rates, they tend to overgrow cultures of human tumor cells, especially slowly proliferating subpopulations. Mouse cell derived DNA, mRNA, and protein components can bias downstream gene expression analysis, next-generation sequencing, as well as proteome analysis (5). To overcome these limitations, we have developed a fast and easy method to isolate untouched human tumor cells from xenografted tumor tissue. This procedure is based on the comprehensive depletion of cells of mouse origin by combining automated tissue dissociation with the benchtop tissue dissociator and magnetic cell sorting. Here, we demonstrate that human target cells can be can be obtained with purities higher than 96% within less than 20 min independent of the tumor type.


Asunto(s)
Neoplasias , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Trasplante Heterólogo
18.
PLoS One ; 10(11): e0143538, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26618511

RESUMEN

RATIONALE: Central questions such as cardiomyocyte subtype emergence during cardiogenesis or the availability of cardiomyocyte subtypes for cell replacement therapy require selective identification and purification of atrial and ventricular cardiomyocytes. However, current methodologies do not allow for a transgene-free selective isolation of atrial or ventricular cardiomyocytes due to the lack of subtype specific cell surface markers. METHODS AND RESULTS: In order to develop cell surface marker-based isolation procedures for cardiomyocyte subtypes, we performed an antibody-based screening on embryonic mouse hearts. Our data indicate that atrial and ventricular cardiomyocytes are characterized by differential expression of integrin α6 (ITGA6) throughout development and in the adult heart. We discovered that the expression level of this surface marker correlates with the intracellular subtype-specific expression of MLC-2a and MLC-2v on the single cell level and thereby enables the discrimination of cardiomyocyte subtypes by flow cytometry. Based on the differential expression of ITGA6 in atria and ventricles during cardiogenesis, we developed purification protocols for atrial and ventricular cardiomyocytes from mouse hearts. Atrial and ventricular identities of sorted cells were confirmed by expression profiling and patch clamp analysis. CONCLUSION: Here, we introduce a non-genetic, antibody-based approach to specifically isolate highly pure and viable atrial and ventricular cardiomyocytes from mouse hearts of various developmental stages. This will facilitate in-depth characterization of the individual cellular subsets and support translational research applications.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Atrios Cardíacos/citología , Ventrículos Cardíacos/citología , Integrina alfa6/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Células Cultivadas , Atrios Cardíacos/embriología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Integrina alfa6/genética , Ratones , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo
19.
Front Immunol ; 5: 16, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24550909

RESUMEN

T-cell therapy after hematopoietic stem cell transplantation (HSCT) has been used alone or in combination with immunosuppression to cure hematologic malignancies and to prevent disease recurrence. Here, we describe the outcome of patients with high-risk/advanced stage hematologic malignancies, who received T-cell depleted (TCD) haploidentical-HSCT (haplo-HSCT) combined with donor T lymphocytes pretreated with IL-10 (ALT-TEN trial). IL-10-anergized donor T cells (IL-10-DLI) contained T regulatory type 1 (Tr1) cells specific for the host alloantigens, limiting donor-vs.-host-reactivity, and memory T cells able to respond to pathogens. IL-10-DLI were infused in 12 patients with the goal of improving immune reconstitution after haplo-HSCT without increasing the risk of graft-versus-host-disease (GvHD). IL-10-DLI led to fast immune reconstitution in five patients. In four out of the five patients, total T-cell counts, TCR-Vß repertoire and T-cell functions progressively normalized after IL-10-DLI. These four patients are alive, in complete disease remission and immunosuppression-free at 7.2 years (median follow-up) after haplo-HSCT. Transient GvHD was observed in the immune reconstituted (IR) patients, despite persistent host-specific hypo-responsiveness of donor T cells in vitro and enrichment of cells with Tr1-specific biomarkers in vivo. Gene-expression profiles of IR patients showed a common signature of tolerance. This study provides the first indication of the feasibility of Tr1 cell-based therapy and paves way for the use of these Tr1 cells as adjuvant treatment for malignancies and immune-mediated disorders.

20.
Stem Cell Res ; 10(2): 133-46, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23237958

RESUMEN

The cellular heterogeneity that is generated during the differentiation of pluripotent stem cells into specific neural subpopulations represents a major obstacle for experimental and clinical progress. To address this problem we developed an optimized strategy for magnetic isolation of PSA-NCAM positive neuronal precursors from embryonic stem cells (ESCs) derived neuronal cultures. PSA-NCAM enrichment at an early step of the in vitro differentiation process increased the number of ES cell derived neurons and reduced cellular diversity. Gene expression analysis revealed that mainly genes involved in neuronal activity were over-represented after purification. In vitro derived PSA-NCAM(+) enriched precursors were characterized in vivo through grafting into the forebrain of adult mice. While unsorted control cells 40 days post graft gave rise to a mixed population composed of immature precursors, early postmitotic neurons and glial cells, PSA-NCAM(+) enriched cells differentiated predominantly into NeuN positive cells. Furthermore, PSA-NCAM enriched population showed efficient migration towards the olfactory bulb after transplantation into the rostral migratory stream of the forebrain neurogenic system. Thus, enrichment of neuronal precursors based on PSA-NCAM expression represents a general and straightforward approach to narrow cellular heterogeneity during neuronal differentiation of pluripotent cells.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Separación Inmunomagnética , Células-Madre Neurales/citología , Neuronas/citología , Actinas/metabolismo , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Supervivencia Celular/genética , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Ácidos Siálicos/metabolismo , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA