Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Ann Oncol ; 34(3): 275-288, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36526124

RESUMEN

BACKGROUND: KEAP1 mutations have been associated with reduced survival in lung adenocarcinoma (LUAD) patients treated with immune checkpoint inhibitors (ICIs), particularly in the presence of STK11/KRAS alterations. We hypothesized that, beyond co-occurring genomic events, clonality prediction may help identify deleterious KEAP1 mutations and their counterparts with retained sensitivity to ICIs. PATIENTS AND METHODS: Beta-binomial modelling of sequencing read counts was used to infer KEAP1 clonal inactivation by combined somatic mutation and loss of heterozygosity (KEAP1 C-LOH) versus partial inactivation [KEAP1 clonal diploid-subclonal (KEAP1 CD-SC)] in the Memorial Sloan Kettering Cancer Center (MSK) MetTropism cohort (N = 2550). Clonality/LOH prediction was compared to a streamlined clinical classifier that relies on variant allele frequencies (VAFs) and tumor purity (TP) (VAF/TP ratio). The impact of this classification on survival outcomes was tested in two independent cohorts of LUAD patients treated with immunotherapy (MSK/Rome N = 237; DFCI N = 461). Immune-related features were studied by exploiting RNA-sequencing data (TCGA) and multiplexed immunofluorescence (DFCI mIF cohort). RESULTS: Clonality/LOH inference in the MSK MetTropism cohort overlapped with a clinical classification model defined by the VAF/TP ratio. In the ICI-treated MSK/Rome discovery cohort, predicted KEAP1 C-LOH mutations were associated with shorter progression-free survival (PFS) and overall survival (OS) compared to KEAP1 wild-type cases (PFS log-rank P = 0.001; OS log-rank P < 0.001). Similar results were obtained in the DFCI validation cohort (PFS log-rank P = 0.006; OS log-rank P = 0.014). In both cohorts, we did not observe any significant difference in survival outcomes when comparing KEAP1 CD-SC and wild-type tumors. Immune deconvolution and multiplexed immunofluorescence revealed that KEAP1 C-LOH and KEAP1 CD-SC differed for immune-related features. CONCLUSIONS: KEAP1 C-LOH mutations are associated with an immune-excluded phenotype and worse clinical outcomes among advanced LUAD patients treated with ICIs. By contrast, survival outcomes of patients whose tumors harbored KEAP1 CD-SC mutations were similar to those with KEAP1 wild-type LUADs.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/patología , Proteína 1 Asociada A ECH Tipo Kelch/genética , Factor 2 Relacionado con NF-E2/genética , Mutación , Pérdida de Heterocigocidad , Inmunoterapia
2.
Ann Oncol ; 31(12): 1746-1754, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32866624

RESUMEN

BACKGROUND: Immune checkpoint inhibitors (ICIs) have demonstrated significant overall survival (OS) benefit in lung adenocarcinoma (LUAD). Nevertheless, a remarkable interpatient heterogeneity characterizes immunotherapy efficacy, regardless of programmed death-ligand 1 (PD-L1) expression and tumor mutational burden (TMB). KEAP1 mutations are associated with shorter survival in LUAD patients receiving chemotherapy. We hypothesized that the pattern of KEAP1 co-mutations and mutual exclusivity may identify LUAD patients unresponsive to immunotherapy. PATIENTS AND METHODS: KEAP1 mutational co-occurrences and somatic interactions were studied in the whole MSKCC LUAD dataset. The impact of coexisting alterations on survival outcomes in ICI-treated LUAD patients was verified in the randomized phase II/III POPLAR/OAK trials (blood-based sequencing, bNGS cohort, N = 253). Three tissue-based sequencing studies (Rome, MSKCC and DFCI) were used for independent validation (tNGS cohort, N = 289). Immunogenomic features were analyzed using The Cancer Genome Atlas (TCGA) LUAD study. RESULTS: On the basis of KEAP1 mutational co-occurrences, we identified four genes potentially associated with reduced efficacy of immunotherapy (KEAP1, PBRM1, SMARCA4 and STK11). Independent of the nature of co-occurring alterations, tumors with coexisting mutations (CoMut) had inferior survival as compared with single-mutant (SM) and wild-type (WT) tumors (bNGS cohort: CoMut versus SM log-rank P = 0.048, CoMut versus WT log-rank P < 0.001; tNGS cohort: CoMut versus SM log-rank P = 0.037, CoMut versus WT log-rank P = 0.006). The CoMut subset harbored higher TMB than the WT disease and the adverse significance of coexisting alterations was maintained in LUAD with high TMB. Significant immunogenomic differences were observed between the CoMut and WT groups in terms of core immune signatures, T-cell receptor repertoire, T helper cell signatures and immunomodulatory genes. CONCLUSIONS: This study indicates that coexisting alterations in a limited set of genes characterize a subset of LUAD unresponsive to immunotherapy and with high TMB. An immune-cold microenvironment may account for the clinical course of the disease.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/terapia , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Humanos , Inmunoterapia , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Mutación , Factor 2 Relacionado con NF-E2 , Ensayos Clínicos Controlados Aleatorios como Asunto , Microambiente Tumoral
3.
Vet Pathol ; 54(3): 405-412, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28438108

RESUMEN

Canine osteosarcoma is highly resistant to current chemotherapy; thus, clarifying the mechanisms of tumor cell resistance to treatments is an urgent need. We tested the geldanamycin derivative 17-AAG (17-allylamino-17-demethoxygeldanamycin) prototype of Hsp90 (heat shock protein 90) inhibitors in 2 canine osteosarcoma cell lines, D22 and D17, derived from primary and metastatic tumors, respectively. With the aim to understand the interplay between cell death, autophagy, and mitophagy, in light of the dual effect of autophagy in regulating cancer cell viability and death, D22 and D17 cells were treated with different concentrations of 17-AAG (0.5 µM, 1 µM) for 24 and 48 hours. 17-AAG-induced apoptosis, necrosis, autophagy, and mitophagy were assessed by transmission electron microscopy, flow cytometry, and immunofluorescence. A simultaneous increase in apoptosis, autophagy, and mitophagy was observed only in the D22 cell line, while D17 cells showed low levels of apoptotic cell death. These results reveal differential cell response to drug-induced stress depending on tumor cell type. Therefore, pharmacological treatments based on proapoptotic chemotherapy in association with autophagy regulators would benefit from a predictive in vitro screening of the target cell type.


Asunto(s)
Antineoplásicos/farmacología , Benzoquinonas/farmacología , Neoplasias Óseas/veterinaria , Enfermedades de los Perros/patología , Lactamas Macrocíclicas/farmacología , Osteosarcoma/veterinaria , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Benzoquinonas/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Línea Celular Tumoral , Enfermedades de los Perros/tratamiento farmacológico , Perros , Relación Dosis-Respuesta a Droga , Lactamas Macrocíclicas/uso terapéutico , Microscopía Electrónica de Transmisión/veterinaria , Mitofagia/efectos de los fármacos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología
4.
G Chir ; 35(11-12): 257-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25644725

RESUMEN

The Cancer Stem Cells (CSC) hypothesis is based on three fundamental ideas: 1) the similarities in the mechanisms that regulate self-renewal of normal stem cells and cancer cells; 2) the possibility that tumour cells might arise from normal stem cells; 3) the notion that tumours might contain 'cancer stem cells' - rare cells with indefinite proliferative potential that drive the formation and growth of tumours. The roles for cancer stem cells have been demonstrated for some cancers, such as cancers of the hematopoietic system, breast, brain, prostate, pancreas and liver. The attractive idea about cancer stem cell hypothesis is that it could partially explain the concept of minimal residual disease. After surgical macroscopically zero residual (R0) resections, even the persistence of one single cell nestling in one of the so called "CSCs niches" could give rise to distant relapse. Furthermore the metastatic cells can remain in a "dormant status" and give rise to disease after long period of apparent disease free. These cells in many cases have acquired resistance traits to chemo and radiotherapy making adjuvant treatment vain. Clarifying the role of the cancer stem cells and their implications in the oncogenesis will play an important role in the management of cancer patient by identifying new prospective for drugs and specific markers to prevent and monitoring relapse and metastasis. The identification of the niche where the CSCs reside in a dormant status might represent a valid instrument to follow-up patients also after having obtained a R0 surgical resection. What we believe is that if new diagnostic instruments were developed specifically to identify the localization and status of activity of the CSCs during tumor dormancy, this would lead to impressive improvement in the early detection and management of relapse and metastasis.


Asunto(s)
Neoplasias/patología , Neoplasias/cirugía , Células Madre Neoplásicas , Humanos
5.
Mol Cell Biochem ; 374(1-2): 233-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23225229

RESUMEN

Increased oxidative stress is known to play a role in the pathogenesis of atherosclerosis, and polymorphisms in genes encoding for enzymes involved in modulation of oxidant stress, such as paraoxonases (PONs), provide a potentially powerful approach to study the risk of disease susceptibility. Aim of our study is to investigate the possible association among PONs polymorphisms, clinical and metabolic factors, and atherothrombotic events in an Italian population. We evaluated in 105 subjects, with or without atherosclerotic risk factors, the presence of PON1 L55M, PON1 Q192R, and PON2 S311C genetic variants, as well as lipid profile, the concentration of aminothiols (blood reduced glutathione, plasma total glutathione, homocysteine, cysteine, cysteinyl glycine), and malondialdehyde as markers of lipid peroxidation. Clinical, biochemical, and genetic variables were correlated with a history of atherothrombosis. Previous atherothrombotic events were found in 42 patients (40 %): myocardial infarction in 24, stroke or transient ischemic attack in 18. By multiple logistic regression analysis, hypertension (OR = 5.538; 95 % CI 2.202-13.902, P < 0.001), HDL-cholesterol concentration (OR = 0.947; 95 % CI 0.910-0.985, P = 0.007), and the presence of C allele in PON2 gene (OR = 3.595; 95 % CI 1.247-10.361, P = 0.018) were independently associated with atherothrombotic events. Our study sheds light on the role of PON2 as a possible cofactor in determining the risk of events together with the well-known risk markers HDL-cholesterol and hypertension.


Asunto(s)
Arildialquilfosfatasa/genética , Trombosis/genética , Alelos , Cisteína/sangre , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Glutatión/sangre , Homocisteína/sangre , Humanos , Hipertensión/genética , Ataque Isquémico Transitorio/genética , Peroxidación de Lípido , Lípidos/sangre , Masculino , Malondialdehído/sangre , Persona de Mediana Edad , Infarto del Miocardio/genética , Estrés Oxidativo , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Accidente Cerebrovascular/genética
6.
Vet Sci ; 9(11)2022 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-36356060

RESUMEN

Human epidermal growth factor receptor 2 (HER2) is a tyrosine kinase receptor that promotes tumor cell growth and is implicated in the pathogenesis of human breast cancer. The role of HER2 in canine mammary carcinomas (CMCs) is not clear. Therefore, this study aimed to examine the protein expression and cytogenetic changes of HER2 and their correlation with other clinical-pathological parameters in CMC. We retrospectively selected 112 CMCs. HER2, ER, and Ki67 were assessed by immunohistochemistry. HER2 antibody validation was investigated by immunoblot on mammary tumor cell lines. Fluorescence in situ hybridization (FISH) was performed with probes for HER2 and CRYBA1 (control gene present on CFA9). HER2 protein overexpression was detected in 15 carcinomas (13.5%). A total of 90 carcinomas were considered technically adequate by FISH, and 8 out of 90 CMC (10%) were HER2 amplified, 3 of which showed a cluster-type pattern. HER2 overexpression was correlated with an increased number of HER2 gene copies (p = 0.01; R = 0.24) and overall survival (p = 0.03), but no correlation with ER, Ki67, grade, metastases, and tumor-specific survival was found. Surprisingly, co-amplification or polysomy was identified in three tumors, characterized by an increased copy number of both HER2 and CRYBA1. A morphological translocation-fusion pattern was recognized in 20 carcinomas (22%), with a co-localized signal of HER2 and CRYBA1. HER2 is not associated with clinical-pathological parameters of increased malignancy in canine mammary tumors, but it is suitable for studying different amplification patterns.

7.
J Exp Med ; 184(6): 2411-6, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8976196

RESUMEN

Ceramides are intramembrane diffusible mediators involved in transducing signals originated from a variety of cell surface receptors. Different adaptive and differentiative cellular responses, including apoptotic cell death, use ceramide-mediated pathways as an essential part of the program. Here, we show that human dendritic cells respond to CD40 ligand, as well as to tumor necrosis factor-alpha and IL-1 beta, with intracellular ceramide accumulation, as they are induced to differentiate. Dendritic cells down-modulate their capacity to take up soluble antigens in response to exogenously added or endogenously produced ceramides. This is followed by an impairment in presenting soluble antigens to specific T cell clones, while cell viability and the capacity to stimulate allogeneic responses or to present immunogenic peptides is fully preserved. Thus, ceramide-mediated pathways initiated by different cytokines can actively modulate professional antigen-presenting cell function and antigen-specific immune responses.


Asunto(s)
Antígenos/metabolismo , Ceramidas/biosíntesis , Células Dendríticas/fisiología , Interleucina-1/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Células Presentadoras de Antígenos/inmunología , Transporte Biológico , Ligando de CD40 , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Endocitosis/efectos de los fármacos , Humanos , Cinética , Glicoproteínas de Membrana/farmacología , Linfocitos T/inmunología
8.
J Exp Med ; 187(6): 897-902, 1998 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-9500792

RESUMEN

Ceramides deriving from sphingomyelin hydrolysis are important mediators of apoptotic signals originating from Fas (APO-1/CD95). However, definitive evidence for the role played by individual sphingomyelinases is still lacking. We have analyzed lymphoblastoid cell lines derived from patients affected by Niemann Pick disease (NPD), an autosomal recessive disorder caused by loss-of-function mutations within the acidic sphingomyelinase (ASM) gene. NPD lymphoblasts, which display normal neutral sphingomyelinase activity, fail to activate ASM in response to Fas cross-linking, unlike normal lymphoblasts. NPD lymphoblasts also fail to accumulate GD3 ganglioside, a downstream mediator of ceramide-induced cell death (De Maria, R., L. Lenti, F. Malisan, F. D'Agostino, B. Tomassini, A. Zeuner, M.R. Rippo, R. Testi. 1997. Science. 277:1652-1655), and display a substantially inefficient apoptosis after Fas cross-linking. Inefficient apoptosis is due to lack of ASM activity, because proximal signaling from Fas in NPD lymphoblasts is not impaired and apoptosis can be efficiently triggered by passing the ASM defect with exogenous ceramides. Moreover, mannose receptor-mediated transfer of ASM into NPD lymphoblasts rescues their ability to transiently activate ASM, accumulate GD3, and rapidly undergo apoptosis after Fas cross-linking. These results provide definitive genetic evidence for the role of ASM in the progression of apoptotic signals originating from Fas.


Asunto(s)
Apoptosis , Gangliósidos/metabolismo , Linfocitos/fisiología , Esfingomielina Fosfodiesterasa/fisiología , Receptor fas/fisiología , Animales , Células CHO , Cricetinae , Humanos , Ratones
9.
J Exp Med ; 186(8): 1193-200, 1997 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-9334358

RESUMEN

Fas is an apoptosis-inducing surface receptor involved in controlling tissue homeostasis and function at multiple sites. Here we show that beta cells from the pancreata of newly diagnosed insulin-dependent diabetes mellitus (IDDM) patients express Fas and show extensive apoptosis among those cells located in proximity to Fas ligand-expressing T lymphocytes infiltrating the IDDM islets. Normal human pancreatic beta cells that do not constitutively express Fas, become strongly Fas positive after interleuken (IL)-1beta exposure, and are then susceptible to Fas-mediated apoptosis. NG-monomethyl-L-arginine, an inhibitor of nitric oxide (NO) synthase, prevents IL-1beta-induced Fas expression, whereas the NO donors sodium nitroprusside and nitric oxide releasing compound (NOC)-18, induce functional Fas expression in normal pancreatic beta cells. These findings suggest that NO-mediated upregulation of Fas contributes to pancreatic beta cell damage in IDDM.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Óxido Nítrico/fisiología , Receptor fas/fisiología , Adulto , Apoptosis/inmunología , Movimiento Celular/inmunología , Niño , Diabetes Mellitus Tipo 1/etiología , Proteína Ligando Fas , Femenino , Humanos , Interleucina-1/farmacología , Islotes Pancreáticos/metabolismo , Ligandos , Masculino , Glicoproteínas de Membrana/inmunología , Subgrupos de Linfocitos T/patología , Receptor fas/biosíntesis , Receptor fas/metabolismo
10.
J Exp Med ; 180(4): 1547-52, 1994 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-7523573

RESUMEN

Intracellular pathways leading from membrane receptor engagement to apoptotic cell death are still poorly characterized. We investigated the intracellular signaling generated after cross-linking of CD95 (Fas/Apo-1 antigen), a broadly expressed cell surface receptor whose engagement results in triggering of cellular apoptotic programs. DX2, a new functional anti-CD95 monoclonal antibody was produced by immunizing mice with human CD95-transfected L cells. Crosslinking of CD95 with DX2 resulted in the activation of a sphingomyelinase (SMase) in promyelocytic U937 cells, as well as in other human tumor cell lines and in CD95-transfected murine cells, as demonstrated by induction of in vivo sphingomyelin (SM) hydrolysis and generation of ceramide. Direct in vitro measurement of enzymatic activity within CD95-stimulated U937 cell extracts, using labeled SM vesicles as substrates, showed strong SMase activity, which required pH 5.0 for optimal substrate hydrolysis. Finally, all CD95-sensitive cell lines tested could be induced to undergo apoptosis after exposure to cell-permeant C2-ceramide. These data indicate that CD95 cross-linking induces SM breakdown and ceramide production through an acidic SMase, thus providing the first information regarding early signal generation from CD95, and may be relevant in defining the biochemical nature of intracellular messengers leading to apoptotic cell death.


Asunto(s)
Antígenos de Superficie/fisiología , Apoptosis , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Secuencia de Bases , Línea Celular , Ceramidas/fisiología , Activación Enzimática , Humanos , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Receptor fas
11.
J Exp Med ; 180(5): 1999-2004, 1994 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7964477

RESUMEN

The expression and function of CD69, a member of the natural killer cell gene complex family of signal transducing receptors, was investigated on human monocytes. CD69 was found expressed on all peripheral blood monocytes, as a 28- and 32-kD disulfide-linked dimer. Molecular cross-linking of CD69 receptors induced extracellular Ca2+ influx, as revealed by flow cytometry. CD69 cross-linking resulted also in phospholipase A2 activation, as detected by in vivo arachidonic acid release measurement from intact cells and by direct in vitro measurement of enzymatic activity using radiolabeled phosphatidylcholine vesicles. Prostaglandin E 2 alpha, 6-keto-prostaglandin F 1 alpha, and leukotriene B4 were detected by radioimmunoassay in supernatants from CD69-stimulated monocytes, suggesting the activation of both cyclooxygenase and lipoxygenase pathways after CD69 stimulation. CD69 cross-linking, moreover, was able to induce strong nitric oxide (NO) production from monocytes, as detected by accumulation of NO oxydixed derivatives, and cyclic GMP. It is important to note that NO generation was responsible for CD69-mediated increase in spontaneous cytotoxicity against L929 murine transformed fibroblast cell line and induction of redirected cytotoxicity towards P815 FcRII+ murine mastocytoma cell line. These data indicate that CD69 can act as a potent stimulatory molecule on the surface of human peripheral blood monocytes.


Asunto(s)
Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Células Asesinas Naturales/fisiología , Lectinas Tipo C , Monocitos/fisiología , Receptores Inmunológicos , Transducción de Señal , Antígenos de Superficie/fisiología , Ácido Araquidónico/metabolismo , Calcio/metabolismo , Citotoxicidad Inmunológica , Humanos , Glicoproteínas de Membrana/fisiología , Subfamilia B de Receptores Similares a Lectina de Células NK , Óxido Nítrico/biosíntesis , Fosfolipasas A/metabolismo , Fosfolipasas A2 , Receptores de Células Asesinas Naturales
12.
Vet J ; 264: 105538, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33012439

RESUMEN

Cellular adaptation to a hypoxic microenvironment is essential for tumour progression and is largely mediated by HIF-1α and hypoxia-regulated factors, including CXCR4, VEGF-A and GLUT-1. In human osteosarcoma, hypoxia is associated with resistance to chemotherapy as well as with metastasis and poor survival, whereas little is known about its role in canine osteosarcoma (cOSA). This study aimed primarily to evaluate the prognostic value of several known hypoxic markers in cOSA. Immunohistochemical analysis for HIF-1α, CXCR4, VEGF-A and GLUT-1 was performed on 56 appendicular OSA samples; correlations with clinicopathological features and outcome was investigated. The second aim was to investigate the in vitro regulation of markers under chemically induced hypoxia (CoCl2). Two primary canine osteosarcoma cell lines were selected, and Western blotting, immunofluorescence and qRT-PCR were used to study protein and gene expression. Dogs with high-grade OSA (35.7%) were more susceptible to the development of metastases (P = 0.047) and showed high HIF-1α protein expression (P = 0.007). Moreover, HIF-1α overexpression (56%) was correlated with a shorter disease-free interval (DFI; P = 0.01), indicating that it is a reliable negative prognostic marker. The in vitro experiments identified an accumulation of HIF-1α in cOSA cells after chemically induced hypoxia, leading to a significant increase in GLUT-1 transcript (P = 0.02). HIF-1α might be a promising prognostic marker, highlighting opportunities for the use of therapeutic strategies targeting the hypoxic microenvironment in cOSA. These results reinforce the role of the dog as a comparative animal model since similar hypoxic mechanisms are reported in human osteosarcoma.


Asunto(s)
Neoplasias Óseas/veterinaria , Hipoxia de la Célula/fisiología , Enfermedades de los Perros/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Osteosarcoma/veterinaria , Animales , Biomarcadores de Tumor/análisis , Neoplasias Óseas/química , Neoplasias Óseas/fisiopatología , Línea Celular Tumoral , Enfermedades de los Perros/patología , Perros , Femenino , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 1/análisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Inmunohistoquímica/veterinaria , Masculino , Metástasis de la Neoplasia/fisiopatología , Osteosarcoma/química , Osteosarcoma/fisiopatología , Pronóstico , Receptores CXCR4/análisis , Factor A de Crecimiento Endotelial Vascular/análisis
13.
Cell Death Differ ; 15(9): 1491-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18497759

RESUMEN

Glioblastoma multiforme is a severe form of cancer most likely arising from the transformation of stem or progenitor cells resident in the brain. Although the tumorigenic population in glioblastoma is defined as composed by cancer stem cells (CSCs), the cellular target of the transformation hit remains to be identified. Glioma stem cells (SCs) are thought to have a differentiation potential restricted to the neural lineage. However, using orthotopic versus heterotopic xenograft models and in vitro differentiation assays, we found that a subset of glioblastomas contained CSCs with both neural and mesenchymal potential. Subcutaneous injection of CSCs or single CSC clones from two of seven patients produced tumor xenografts containing osteo-chondrogenic areas in the context of glioblastoma-like tumor lesions. Moreover, CSC clones from four of seven cases generated both neural and chondrogenic cells in vitro. Interestingly, mesenchymal differentiation of the tumor xenografts was associated with reduction of both growth rate and mitotic index. These findings suggest that in a subclass of glioblastomas the tumorigenic hit occurs on a multipotent stem cell, which may reveal its plasticity under specific environmental stimuli. The discovery of such biological properties might provide considerable information to the development of new therapeutic strategies aimed at forcing glioblastoma stem cell differentiation.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Mesodermo/citología , Células Madre Neoplásicas/citología , Adulto , Anciano , Animales , Diferenciación Celular , Células Clonales , Femenino , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Células Madre Neoplásicas/química , Células Madre Neoplásicas/patología , Neuronas/citología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Death Differ ; 15(3): 504-14, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18049477

RESUMEN

Lung carcinoma is often incurable and remains the leading cancer killer in both men and women. Recent evidence indicates that tumors contain a small population of cancer stem cells that are responsible for tumor maintenance and spreading. The identification of the tumorigenic population that sustains lung cancer may contribute significantly to the development of effective therapies. Here, we found that the tumorigenic cells in small cell and non-small cell lung cancer are a rare population of undifferentiated cells expressing CD133, an antigen present in the cell membrane of normal and cancer-primitive cells of the hematopoietic, neural, endothelial and epithelial lineages. Lung cancer CD133(+) cells were able to grow indefinitely as tumor spheres in serum-free medium containing epidermal growth factor and basic fibroblast growth factor. The injection of 10(4) lung cancer CD133(+) cells in immunocompromised mice readily generated tumor xenografts phenotypically identical to the original tumor. Upon differentiation, lung cancer CD133(+) cells acquired the specific lineage markers, while loosing the tumorigenic potential together with CD133 expression. Thus, lung cancer contains a rare population of CD133(+) cancer stem-like cells able to self-renew and generates an unlimited progeny of non-tumorigenic cells. Molecular and functional characterization of such a tumorigenic population may provide valuable information to be exploited in the clinical setting.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Pequeñas/patología , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Células Pequeñas/metabolismo , Diferenciación Celular , Resistencia a Antineoplásicos , Femenino , Glicoproteínas/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones , Ratones SCID , Células Madre Neoplásicas/metabolismo , Péptidos/metabolismo , Fenotipo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Science ; 277(5332): 1652-5, 1997 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-9287216

RESUMEN

Gangliosides participate in development and tissue differentiation. Cross-linking of the apoptosis-inducing CD95 protein (also called Fas or APO-1) in lymphoid and myeloid tumor cells triggered GD3 ganglioside synthesis and transient accumulation. CD95-induced GD3 accumulation depended on integral receptor "death domains" and on activation of a family of cysteine proteases called caspases. Cell-permeating ceramides, which are potent inducers of apoptosis, also triggered GD3 synthesis. GD3 disrupted mitochondrial transmembrane potential (DeltaPsim), and induced apoptosis, in a caspase-independent fashion. Transient overexpression of the GD3 synthase gene directly triggered apoptosis. Pharmacological inhibition of GD3 synthesis and exposure to GD3 synthase antisense oligodeoxynucleotides prevented CD95-induced apoptosis. Thus, GD3 ganglioside mediates the propagation of CD95-generated apoptotic signals in hematopoietic cells.


Asunto(s)
Apoptosis , Ceramidas/fisiología , Gangliósidos/metabolismo , Receptor fas/fisiología , Ceramidas/farmacología , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores Enzimáticos/farmacología , Gangliósidos/biosíntesis , Gangliósidos/farmacología , Aparato de Golgi/metabolismo , Humanos , Potenciales de la Membrana , Mitocondrias/fisiología , Morfolinas/farmacología , Oligonucleótidos Antisentido/farmacología , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Receptor fas/metabolismo
16.
Science ; 275(5302): 960-3, 1997 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-9020075

RESUMEN

The mechanisms responsible for thyrocyte destruction in Hashimoto's thyroiditis (HT) are poorly understood. Thyrocytes from HT glands, but not from nonautoimmune thyroids, expressed Fas. Interleukin-1beta (IL-1beta), abundantly produced in HT glands, induced Fas expression in normal thyrocytes, and cross-linking of Fas resulted in massive thyrocyte apoptosis. The ligand for Fas (FasL) was shown to be constitutively expressed both in normal and HT thyrocytes and was able to kill Fas-sensitive targets. Exposure to IL-1beta induced thyrocyte apoptosis, which was prevented by antibodies that block Fas, suggesting that IL-1beta-induced Fas expression serves as a limiting factor for thyrocyte destruction. Thus, Fas-FasL interactions among HT thyrocytes may contribute to clinical hypothyroidism.


Asunto(s)
Apoptosis , Glicoproteínas de Membrana/metabolismo , Glándula Tiroides/metabolismo , Tiroiditis Autoinmune/etiología , Receptor fas/metabolismo , Anticuerpos Monoclonales/inmunología , Células Cultivadas , Citocinas/farmacología , Proteína Ligando Fas , Humanos , Técnicas para Inmunoenzimas , Interleucina-1/farmacología , Glicoproteínas de Membrana/biosíntesis , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Reacción en Cadena de la Polimerasa , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Glándula Tiroides/patología , Tiroiditis Autoinmune/metabolismo , Tiroiditis Autoinmune/patología , Células Tumorales Cultivadas , Receptor fas/biosíntesis , Receptor fas/inmunología
17.
Science ; 285(5433): 1553-8, 1999 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-10477517

RESUMEN

Studies on pluripotent hematopoietic stem cells (HSCs) have been hindered by lack of a positive marker, comparable to the CD34 marker of hematopoietic progenitor cells (HPCs). In human postnatal hematopoietic tissues, 0.1 to 0.5% of CD34(+) cells expressed vascular endothelial growth factor receptor 2 (VEGFR2, also known as KDR). Pluripotent HSCs were restricted to the CD34+KDR+ cell fraction. Conversely, lineage-committed HPCs were in the CD34+KDR- subset. On the basis of limiting dilution analysis, the HSC frequency in the CD34+KDR+ fraction was 20 percent in bone marrow (BM) by mouse xenograft assay and 25 to 42 percent in BM, peripheral blood, and cord blood by 12-week long-term culture (LTC) assay. The latter values rose to 53 to 63 percent in LTC supplemented with VEGF and to greater than 95 percent for the cell subfraction resistant to growth factor starvation. Thus, KDR is a positive functional marker defining stem cells and distinguishing them from progenitors.


Asunto(s)
Antígenos CD34/análisis , Hematopoyesis , Células Madre Hematopoyéticas/citología , Proteínas Tirosina Quinasas Receptoras/análisis , Receptores de Factores de Crecimiento/análisis , Animales , Células de la Médula Ósea/citología , Linaje de la Célula , Separación Celular , Células Cultivadas , Factores de Crecimiento Endotelial/farmacología , Femenino , Sangre Fetal/citología , Feto , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Humanos , Linfocinas/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Embarazo , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular , Ovinos , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
18.
Vet Rec ; 164(15): 459-64, 2009 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-19363227

RESUMEN

The effects of 17beta-oestradiol (E2) on gene expression in cultures of bovine primary prostate stromal cells (BPSCs) and prostate gland tissue were studied. In the first part of the study, BPSCs were grown in the presence of E2 from the first passage to the end of the experiment; a second group was treated in the same way but the treatment was suspended for 48 hours before the end of the experiment; a third group of BPSCs served as a control. In the second part of the study, five male veal calves, aged 130 days, were treated four times intramuscularly with 10 mg of E2 at intervals of two weeks and then euthanased two weeks after the last treatment. Quantitative PCR and immunohistochemistry were used to evaluate the expression of fibroblast growth factor (FGF) receptors (FGFRs), FGFs, progesterone receptor, androgen receptor and oestrogen receptor in BPSCs and prostate tissue. E2 induced a significant over-expression of progesterone receptor in both BPSCs and prostate tissue. There was also a marked up-regulation of FGFR types 1, 2 and 3 genes observed in the BPSCs.


Asunto(s)
Estradiol/farmacología , Factores de Crecimiento de Fibroblastos/biosíntesis , Próstata/citología , Próstata/metabolismo , Receptores de Estrógenos/metabolismo , Regulación hacia Arriba , Animales , Biomarcadores/metabolismo , Bovinos , Células Cultivadas , Factores de Crecimiento de Fibroblastos/genética , Masculino , Reacción en Cadena de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa/veterinaria , ARN/análisis , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
19.
J Hematol Oncol ; 12(1): 111, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665051

RESUMEN

Breast cancer (BC) is a complex disease with primary or acquired incurability characteristics in a significant part of patients. Immunotherapeutical agents represent an emerging option for breast cancer treatment, including the human epidermal growth factor 2 positive (HER2+) subtype. The immune system holds the ability to spontaneously implement a defensive response against HER2+ BC cells through complex mechanisms which can be exploited to modulate this response for obtaining a clinical benefit. Initial immune system modulating strategies consisted mostly in vaccine therapies, which are still being investigated and improved. However, the entrance of trastuzumab into the scenery of HER2+ BC treatment was the real game changing event, which embodied a dominant immune-mediated mechanism. More recently, the advent of the immune checkpoint inhibitors has caused a new paradigm shift for immuno-oncology, with promising initial results also for HER2+ BC. Breast cancer has been traditionally considered poorly immunogenic, being characterized by relatively low tumor mutation burden (TMB). Nevertheless, recent evidence has revealed high tumor infiltrating lymphocytes (TILs) and programmed cell death-ligand 1 (PD-L1) expression in a considerable proportion of HER2+ BC patients. This may translate into a higher potential to elicit anti-cancer response and, therefore, wider possibilities for the use and implementation of immunotherapy in this subset of BC patients. We are herein presenting and critically discussing the most representative evidence concerning immunotherapy in HER2+ BC cancer, both singularly and in combination with therapeutic agents acting throughout HER2-block, immune checkpoint inhibition and anti-cancer vaccines. The reader will be also provided with hints concerning potential future projection of the most promising immutherapeutic agents and approaches for the disease of interest.


Asunto(s)
Neoplasias de la Mama/terapia , Predisposición Genética a la Enfermedad , Inmunoterapia , Receptor ErbB-2/genética , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/genética , Femenino , Humanos
20.
Neuropharmacology ; 55(4): 568-76, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18621067

RESUMEN

Targeted-therapies enhancing differentiation of glioma-initiating cells (GICs) are potential innovative approaches to the treatment of malignant gliomas. These cells support tumour growth and recurrence and are resistant to radiotherapy and chemotherapy. We have found that GICs express mGlu3 metabotropic glutamate receptors. Activation of these receptors sustained the undifferentiated state of GICs in culture by negatively modulating the action of bone morphogenetic proteins, which physiologically signal through the phosphorylation of the transcription factors, Smads. The cross-talk between mGlu3 receptors and BMP receptors was mediated by the activation of the mitogen-activated protein kinase pathway. Remarkably, pharmacological blockade of mGlu3 receptors stimulated the differentiation of cultured GICs into astrocytes, an effect that appeared to be long lasting, independent of the growth conditions, and irreversible. In in vivo experiments, a 3-month treatment with the brain-permeant mGlu receptor antagonist, LY341495 limited the growth of infiltrating brain tumours originating from GICs implanted into the brain parenchyma of nude mice. While clusters of tumour cells were consistently found in the brain of control mice, they were virtually absent in a large proportion of mice treated with LY341495. These findings pave the way to a new non-cytotoxic treatment of malignant gliomas based on the use of mGlu3 receptor antagonists.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Receptores de Glutamato Metabotrópico/fisiología , Transducción de Señal/fisiología , Aminoácidos/farmacología , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Glioma/tratamiento farmacológico , Glioma/patología , Glioma/fisiopatología , Humanos , Imagen por Resonancia Magnética , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/efectos de los fármacos , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Xantenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA