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

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Pathol ; 185(3): 704-16, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25579842

RESUMEN

The two major melanoma histologic subtypes, superficial spreading and nodular melanomas, differ in their speed of dermal invasion but converge biologically once they invade and metastasize. Herein, we tested the hypothesis that distinct molecular alterations arising in primary melanoma cells might persist as these tumors progress to invasion and metastasis. Ribosomal protein S6 kinase, 90 kDa, polypeptide 1 (RSK1; official name RPS6KA1) was significantly hyperactivated in human melanoma lines and metastatic tissues derived from nodular compared with superficial spreading melanoma. RSK1 was constitutively phosphorylated at Ser-380 in nodular but not superficial spreading melanoma and did not directly correlate with BRAF or MEK activation. Nodular melanoma cells were more sensitive to RSK1 inhibition using siRNA and the pharmacological inhibitor BI-D1870 compared with superficial spreading cells. Gene expression microarray analyses revealed that RSK1 orchestrated a program of gene expression that promoted cell motility and invasion. Differential overexpression of the prometastatic matrix metalloproteinase 8 and tissue inhibitor of metalloproteinases 1 in metastatic nodular compared with metastatic superficial spreading melanoma was observed. Finally, using an in vivo zebrafish model, constitutive RSK1 activation increased melanoma invasion. Together, these data reveal a novel role for activated RSK1 in the progression of nodular melanoma and suggest that melanoma originating from different histologic subtypes may be biologically distinct and that these differences are maintained as the tumors invade and metastasize.


Asunto(s)
Movimiento Celular , Melanoma/metabolismo , Invasividad Neoplásica/patología , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Melanoma/patología , Fosforilación , Neoplasias Cutáneas/patología , Pez Cebra
2.
Proc Natl Acad Sci U S A ; 110(17): 6973-8, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23576742

RESUMEN

T cells expressing antigen-specific T-cell receptors (TCRs) can mediate effective tumor regression, but they often also are accompanied by autoimmune responses. To determine the TCR affinity threshold defining the optimal balance between effective antitumor activity and autoimmunity in vivo, we used a unique self-antigen system comprising seven human melanoma gp100(209-217)-specific TCRs spanning physiological affinities (1-100 µM). We found that in vitro and in vivo T-cell responses are determined by TCR affinity, except in one case that was compensated by substantial CD8 involvement. Strikingly, we found that T-cell antitumor activity and autoimmunity are closely coupled but plateau at a defined TCR affinity of 10 µM, likely due to diminished contribution of TCR affinity to avidity above the threshold. Together, these results suggest that a relatively low-affinity threshold is necessary for the immune system to avoid self-damage, given the close relationship between antitumor activity and autoimmunity. The low threshold, in turn, indicates that adoptive T-cell therapy treatment strategies using in vitro-generated high-affinity TCRs do not necessarily improve efficacy.


Asunto(s)
Autoinmunidad/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de Varianza , Pruebas Inmunológicas de Citotoxicidad , Ensayo de Inmunoadsorción Enzimática , Fluorescencia , Humanos , Inmunohistoquímica , Transducción Genética , Antígeno gp100 del Melanoma/inmunología
3.
Cancer ; 121(1): 51-9, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25155861

RESUMEN

BACKGROUND: Identification of primary melanoma patients at the highest risk of recurrence remains a critical challenge, and monitoring for recurrent disease is limited to costly imaging studies. We recently reported our array-based discovery of prognostic serum miRNAs in melanoma. In the current study, we examined the clinical utility of these serum-based miRNAs for prognosis as well as detection of melanoma recurrence. METHODS: Serum levels of 12 miRNAs were tested using qRT-PCR at diagnosis in 283 melanoma patients (training cohort, n = 201; independent validation, n = 82; median follow-up, 68.8 months). A refined miRNA signature was chosen and evaluated. We also tested the potential clinical utility of the miRNAs in early detection and monitoring of recurrence using multiple longitudinal samples (pre- and postrecurrence) in a subset of 82 patients (n = 225). In addition, we integrated our miRNA signature with publicly available Cancer Genome Atlas data to examine the relevance of these miRNAs to melanoma biology. RESULTS: Four miRNAs (miR-150, miR-30d, miR-15b, and miR-425) in combination with stage separated patients by recurrence-free survival (RFS) and overall survival (OS) and improved prediction of recurrence over stage alone in both the training and validation cohorts (training RFS and OS, P < .001; validation RFS, P < .001; OS, P = .005). Serum miR-15b levels significantly increased over time in recurrent patients (P < .001), adjusting for endogenous controls as well as age, sex, and initial stage. In nonrecurrent patients, miR-15b levels were not significantly changed with time (P =.17). CONCLUSIONS: Data demonstrate that serum miRNAs can improve melanoma patient stratification over stage and support further testing of miR-15b to guide patient surveillance.


Asunto(s)
Melanoma/diagnóstico , Melanoma/genética , MicroARNs/sangre , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/genética , Adulto , Anciano , Anciano de 80 o más Años , Detección Precoz del Cáncer , Femenino , Regulación Neoplásica de la Expresión Génica , Historia Antigua , Humanos , Masculino , Melanoma/sangre , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Adulto Joven
4.
Oncology ; 85(3): 173-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24008821

RESUMEN

OBJECTIVES: Age is an understudied factor when considering treatment options for melanoma. Here, we examine the impact of age on primary melanoma treatment in a prospective cohort of patients. METHODS: We used logistic regression models to examine the associations between age and initial treatment, using recurrence and melanoma-specific survival as endpoints. RESULTS: 444 primary melanoma patients were categorized into three groups by age at diagnosis: 19-45 years (24.3%), 46-70 (50.2%), and 71-95 (25.5%). In multivariate models, older patients experienced a higher risk of recurrence (hazard ratio 3.34, 95% confidence interval, CI, 1.53-7.25; p < 0.01). No significant differences were observed in positive biopsy margin rates or extent of surgical margins across age groups. Patients in the middle age group were more likely to receive adjuvant therapy than those in the older group (odds ratio 2.78, 95% CI 1.19-6.45; p = 0.02) and showed a trend to longer disease-free survival when receiving adjuvant therapy (p = 0.09). CONCLUSION: Our data support age as an independent negative prognostic factor in melanoma. Our data suggest that age does not affect primary surgical treatment but may affect decisions of whether or not patients receive postoperative treatment(s). Further work is needed to better understand the biological variables affecting treatment decisions and efficacy in older patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma/mortalidad , Recurrencia Local de Neoplasia/mortalidad , Neoplasias Cutáneas/mortalidad , Adulto , Distribución por Edad , Factores de Edad , Anciano , Anciano de 80 o más Años , Quimioterapia Adyuvante , Supervivencia sin Enfermedad , Femenino , Disparidades en Atención de Salud , Humanos , Modelos Logísticos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Oportunidad Relativa , Selección de Paciente , Pronóstico , Estudios Prospectivos , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Resultado del Tratamiento , Estados Unidos/epidemiología
5.
Cancers (Basel) ; 16(1)2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38201446

RESUMEN

BACKGROUND: We previously reported a higher incidence of a pathogenic germline variant in the kinase insert domain receptor (KDR) in melanoma patients compared to the general population. Here, we dissect the impact of this genotype on melanoma tumor growth kinetics, tumor phenotype, and response to treatment with immune checkpoint inhibitors (ICIs) or targeted therapy. METHODS: The KDR genotype was determined and the associations between the KDR Q472H variant (KDR-Var), angiogenesis, tumor immunophenotype, and response to MAPK inhibition or ICI treatment were examined. Melanoma B16 cell lines were transfected with KDR-Var or KDR wild type (KDR-WT), and the differences in tumor kinetics were evaluated. We also examined the impact of KDR-Var on the response of melanoma cells to a combination of VEGFR inhibition with MAPKi. RESULTS: We identified the KDR-Var genotype in 81/489 (37%) patients, and it was associated with a more angiogenic (p = 0.003) and immune-suppressive tumor phenotype. KDR-Var was also associated with decreased PFS to MAPKi (p = 0.022) and a trend with worse PFS to anti-PD1 therapy (p = 0.06). KDR-Var B16 murine models had increased average tumor volume (p = 0.0027) and decreased CD45 tumor-infiltrating lymphocytes (p = 0.0282). The anti-VEGFR treatment Lenvatinib reduced the tumor size of KDR-Var murine tumors (p = 0.0159), and KDR-Var cells showed synergistic cytotoxicity to the combination of dabrafenib and lenvatinib. CONCLUSIONS: Our data demonstrate a role of germline KDR-Var in modulating melanoma behavior, including response to treatment. Our data also suggest that anti-angiogenic therapy might be beneficial in patients harboring this genotype, which needs to be tested in clinical trials.

6.
Nat Commun ; 14(1): 1867, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-37015919

RESUMEN

Metastatic melanoma develops once transformed melanocytic cells begin to de-differentiate into migratory and invasive melanoma cells with neural crest cell (NCC)-like and epithelial-to-mesenchymal transition (EMT)-like features. However, it is still unclear how transformed melanocytes assume a metastatic melanoma cell state. Here, we define DNA methylation changes that accompany metastatic progression in melanoma patients and discover Nuclear Receptor Subfamily 2 Group F, Member 2 - isoform 2 (NR2F2-Iso2) as an epigenetically regulated metastasis driver. NR2F2-Iso2 is transcribed from an alternative transcriptional start site (TSS) and it is truncated at the N-terminal end which encodes the NR2F2 DNA-binding domain. We find that NR2F2-Iso2 expression is turned off by DNA methylation when NCCs differentiate into melanocytes. Conversely, this process is reversed during metastatic melanoma progression, when NR2F2-Iso2 becomes increasingly hypomethylated and re-expressed. Our functional and molecular studies suggest that NR2F2-Iso2 drives metastatic melanoma progression by modulating the activity of full-length NR2F2 (Isoform 1) over EMT- and NCC-associated target genes. Our findings indicate that DNA methylation changes play a crucial role during metastatic melanoma progression, and their control of NR2F2 activity allows transformed melanocytes to acquire NCC-like and EMT-like features. This epigenetically regulated transcriptional plasticity facilitates cell state transitions and metastatic spread.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Línea Celular Tumoral , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Transición Epitelial-Mesenquimal/genética , Epigénesis Genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción COUP II/metabolismo
7.
Mod Pathol ; 25(7): 1000-10, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22425909

RESUMEN

The sentinel lymph node is the initial site of metastasis. Downregulation of antitumor immunity has a role in nodal progression. Our objective was to investigate the relationship between immune modulation and sentinel lymph node positivity, correlating it with outcome in melanoma patients. Lymph node/primary tissues from melanoma patients prospectively accrued and followed at New York University Medical Center were evaluated for the presence of regulatory T cells (Foxp3(+)) and dendritic cells (conventional: CD11c(+), mature: CD86(+)) using immunohistochemistry. Primary melanoma immune cell profiles from sentinel lymph node-positive/-negative patients were compared. Logistic regression models inclusive of standard-of-care/immunological primary tumor characteristics were constructed to predict the risk of sentinel lymph node positivity. Immunological responses in the positive sentinel lymph node were also compared with those in the negative non-sentinel node from the same nodal basin and matched negative sentinel lymph node. Decreased immune response was defined as increased regulatory T cells or decreased dendritic cells. Associations between the expression of these immune modulators, clinicopathological variables, and clinical outcome were evaluated using univariate/multivariate analyses. Primary tumor conventional dendritic cells and regression were protective against sentinel lymph node metastasis (odds ratio=0.714, 0.067; P=0.0099, 0.0816, respectively). Antitumor immunity was downregulated in the positive sentinel lymph node with an increase in regulatory T cells compared with the negative non-sentinel node from the same nodal basin (P=0.0005) and matched negative sentinel lymph node (P=0.0002). The positive sentinel lymph node also had decreased numbers of conventional dendritic cells compared with the negative sentinel lymph node (P<0.0001). Adding sentinel lymph node regulatory T cell expression improved the discriminative power of a recurrence risk assessment model using clinical stage. Primary tumor regression was associated with prolonged disease-free (P=0.025) and melanoma-specific (P=0.014) survival. Our results support an assessment of local immune profiles in both the primary tumor and sentinel lymph node to help guide therapeutic decisions.


Asunto(s)
Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Melanoma/inmunología , Melanoma/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Adulto , Anciano , Área Bajo la Curva , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática/inmunología , Masculino , Melanoma/mortalidad , Persona de Mediana Edad , Curva ROC , Biopsia del Ganglio Linfático Centinela , Neoplasias Cutáneas/mortalidad
8.
Sci Adv ; 8(7): eabi7127, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35179962

RESUMEN

The contribution of epigenetic dysregulation to metastasis remains understudied. Through a meta-analysis of gene expression datasets followed by a mini-screen, we identified Plant Homeodomain Finger protein 8 (PHF8), a histone demethylase of the Jumonji C protein family, as a previously unidentified prometastatic gene in melanoma. Loss- and gain-of-function approaches demonstrate that PHF8 promotes cell invasion without affecting proliferation in vitro and increases dissemination but not subcutaneous tumor growth in vivo, thus supporting its specific contribution to the acquisition of metastatic potential. PHF8 requires its histone demethylase activity to enhance melanoma cell invasion. Transcriptomic and epigenomic analyses revealed that PHF8 orchestrates a molecular program that directly controls the TGFß signaling pathway and, as a consequence, melanoma invasion and metastasis. Our findings bring a mechanistic understanding of epigenetic regulation of metastatic fitness in cancer, which may pave the way for improved therapeutic interventions.


Asunto(s)
Histona Demetilasas , Melanoma , Proliferación Celular , Epigénesis Genética , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Melanoma/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
9.
Pigment Cell Melanoma Res ; 33(3): 466-479, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31663663

RESUMEN

Next-generation sequencing has enabled genetic and genomic characterization of melanoma to an unprecedent depth. However, the high mutational background plus the limited depth of coverage of whole-genome sequencing performed on cutaneous melanoma samples make the identification of novel driver mutations difficult. We sought to explore the somatic mutation portfolio in exonic and gene regulatory regions in human melanoma samples, for which we performed targeted sequencing of tumors and matched germline DNA samples from 89 melanoma patients, identifying known and novel recurrent mutations. Two recurrent mutations found in the RPS27 promoter associated with decreased RPS27 mRNA levels in vitro. Data mining and IHC analyses revealed a bimodal pattern of RPS27 expression in melanoma, with RPS27-low patients displaying worse prognosis. In vitro characterization of RPS27-high and RPS27-low melanoma cell lines, as well as loss-of-function experiments, demonstrated that high RPS27 status provides increased proliferative and invasive capacities, while low RPS27 confers survival advantage in low attachment and resistance to therapy. Additionally, we demonstrate that 10 other cancer types harbor bimodal RPS27 expression, and in those, similarly to melanoma, RPS27-low expression associates with worse clinical outcomes. RPS27 promoter mutation could thus represent a mechanism of gene expression modulation in melanoma patients, which may have prognostic and predictive implications.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Metaloproteínas/genética , Mutación/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Proteínas Ribosómicas/genética , Animales , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sitios Genéticos , Genómica , Humanos , Metaloproteínas/metabolismo , Ratones , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Ribosómicas/metabolismo
10.
J Invest Dermatol ; 139(2): 430-438, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30148988

RESUMEN

The extent of PTEN loss that confers clinical and biological impact in melanoma is unclear. We evaluated the clinical and biologic relevance of PTEN dosage in melanoma and tested the postulate that partial PTEN loss is due to epigenetic mechanisms. PTEN expression was assessed by immunohistochemistry in a stage III melanoma cohort (n = 190) with prospective follow up. Overall, 21 of 190 (11%) tumors had strong PTEN expression, 51 of 190 (27%) had intermediate PTEN, 44 of 190 (23%) had weak PTEN, and 74 of 190 (39%) had absent PTEN. Both weak and absent PTEN expression predicted shorter survival in multivariate analyses (hazard ratio = 2.13, P < 0.01). We show a continuous negative correlation between PTEN and activated Akt in melanoma cells with titrated PTEN expression and in two additional independent tumor datasets. PTEN genomic alterations (deletion, mutation), promoter methylation, and protein destabilization did not fully explain PTEN loss in melanoma, whereas PTEN levels increased with treatment of melanoma cells with the histone deacetylase inhibitor LBH589. Our data indicate that partial PTEN loss is due to modifiable epigenetic mechanisms and drives Akt activation and worse prognosis, suggesting a potential approach to improve the clinical outcome for a subset of patients with advanced melanoma.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Fosfohidrolasa PTEN/genética , Neoplasias Cutáneas/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Femenino , Estudios de Seguimiento , Dosificación de Gen , Humanos , Masculino , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , Estudios Prospectivos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piel/patología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Análisis de Supervivencia , Adulto Joven
11.
Hum Pathol ; 85: 65-71, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30428388

RESUMEN

Microglandular adenosis (MGA) is a rare breast lesion reported to be associated with invasive carcinoma in up to 20% to 30% of cases and has been proposed as a nonobligate precursor to basal-like breast cancers. We identified a case of matrix-producing metaplastic carcinoma with morphologic and immunohistochemical evidence of progression from MGA to atypical MGA, carcinoma in situ, and invasive carcinoma. We performed whole-exome sequencing of each component (MGA, atypical MGA, carcinoma in situ, and cancer) to characterize the mutational landscape of these foci. There was a significant copy number overlap between all foci, including a segmental amplification of the CCND1 locus (partial chromosome 11 trisomy) and MYC (8q24.12-13). Using a bioinformatics approach, we were able to identify 3 putative mutational clusters and recurrent, stop-gain nonsynonymous mutations in both ZNF862 and TP53 that were shared across all foci. Finally, we identified a novel deleterious splice-acceptor site mutation of chr5:5186164 G>T (chromosome 5p15) encoding the gene, ADAMTS16, in the invasive component.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Enfermedad Fibroquística de la Mama/patología , Lesiones Precancerosas/patología , Proteínas ADAMTS/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Progresión de la Enfermedad , Femenino , Enfermedad Fibroquística de la Mama/metabolismo , Humanos , Inmunohistoquímica , Lesiones Precancerosas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
12.
Neuron ; 37(3): 449-61, 2003 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-12575952

RESUMEN

Subthreshold-activating somatodendritic A-type potassium channels have fundamental roles in neuronal signaling and plasticity which depend on their unique cellular localization, voltage dependence, and kinetic properties. Some of the components of A-type K(+) channels have been identified; however, these do not reproduce the properties of the native channels, indicating that key molecular factors have yet to be unveiled. We purified A-type K(+) channel complexes from rat brain membranes and found that DPPX, a protein of unknown function that is structurally related to the dipeptidyl aminopeptidase and cell adhesion protein CD26, is a novel component of A-type K(+) channels. DPPX associates with the channels' pore-forming subunits, facilitates their trafficking and membrane targeting, reconstitutes the properties of the native channels in heterologous expression systems, and is coexpressed with the pore-forming subunits in the somatodendritic compartment of CNS neurons.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/enzimología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/fisiología , Cerebelo/citología , Dendritas/enzimología , Dipeptidil Peptidasa 4/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Matriz Extracelular/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Neuronas/ultraestructura , Oocitos/fisiología , Canales de Potasio/química , Pruebas de Precipitina , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Canales de Potasio Shal , Xenopus
13.
Brain Res ; 1094(1): 1-12, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16764835

RESUMEN

Transient subthreshold-activating somato-dendritic A-type K(+) currents (I(SA)s) have fundamental roles in neuronal function. They cause delayed excitation, influence spike repolarization, modulate the frequency of repetitive firing, and have important roles in signal processing in dendrites. We previously reported that DPPX proteins are key components of the channels mediating these currents (Kv4 channels) (Nadal, M.S., Ozaita, A., Amarillo, Y., Vega-Saenz, E., Ma, Y., Mo, W., Goldberg, E.M., Misumi, Y., Ikehara, Y., Neubert, T.A., Rudy, B., 2003. The CD26-related dipeptidyl aminopeptidase-like protein DPPX is a critical component of neuronal A-type K+ channels. Neuron 37, 449-461). The DPPX gene encodes alternatively spliced transcripts that generate single-spanning transmembrane proteins with a short, divergent intracellular domain and a large extracellular domain. We characterized the modulatory effects on Kv4.2-mediated currents and the rat brain distribution of three splice variants of the DPPX subfamily of proteins. These three splice isoforms--DPPX-S, DPPX-L, and DPPX-K--are expressed in adult rat brain and modify the voltage dependence and kinetic properties of Kv4.2 channels expressed in Xenopus oocytes. Analysis of a deletion mutant that lacks the variable N-terminus showed that the N-terminus is not necessary for the modulation of Kv4 channels. Using in situ hybridization analysis, we found that the three splice variants are prominently expressed in brain regions where Kv4 subunits are also expressed. DPPX-K and DPPX-S mRNAs have a widespread distribution, whereas DPPX-L transcripts are concentrated in few specific areas of the rat brain. The emerging diversity of DPPX splice variants, differing only in the N-terminus of the protein, opens up intriguing possibilities for the modulation of Kv4 channels.


Asunto(s)
Empalme Alternativo/genética , Encéfalo/metabolismo , Membrana Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Canales de Potasio/metabolismo , Canales de Potasio Shal/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/anatomía & histología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Femenino , Masculino , Potenciales de la Membrana/genética , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/aislamiento & purificación , Oocitos , Canales de Potasio/genética , Canales de Potasio/aislamiento & purificación , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Canales de Potasio Shal/química , Canales de Potasio Shal/genética , Xenopus
14.
Brain Res Mol Brain Res ; 123(1-2): 91-103, 2004 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-15046870

RESUMEN

Human and rat Kv10.1a and b cDNAs encode silent K+ channel pore-forming subunits that modify the electrophysiological properties of Kv2.1. These alternatively spliced variants arise by the usage of an alternative site of splicing in exon 1 producing an 11-amino acid insertion in the linker between the first and second transmembrane domains in Kv10.1b. In human, the Kv10s mRNA were detected by Northern blot in brain kidney lung and pancreas. In brain, they were expressed in cortex, hippocampus, caudate, putamen, amygdala and weakly in substantia nigra. In rat, Kv10.1 products were detected in brain and weakly in testes. In situ hybridization in rat brain shows that Kv10.1 mRNAs are expressed in cortex, olfactory cortical structures, basal ganglia/striatal structures, hippocampus and in many nuclei of the amygdala complex. The CA3 and dentate gyrus of the hippocampus present a gradient that show a progression from high level of expression in the caudo-ventro-medial area to a weak level in the dorso-rostral area. The CA1 and CA2 areas had low levels throughout the hippocampus. Several small nuclei were also labeled in the thalamus, hypothalamus, pons, midbrain, and medulla oblongata. Co-injection of Kv2.1 and Kv10.1a or b mRNAs in Xenopus oocytes produced smaller currents that in the Kv2.1 injected oocytes and a moderate reduction of the inactivation rate without any appreciable change in recovery from inactivation or voltage dependence of activation or inactivation. At higher concentration, Kv10.1a also reduces the activation rate and a more important reduction in the inactivation rate. The gene that encodes for Kv10.1 mRNAs maps to chromosome 2p22.1 in human, 6q12 in rat and 17E4 in mouse, locations consistent with the known systeny for human, rat and mouse chromosomes.


Asunto(s)
Química Encefálica/genética , Encéfalo/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Empalme Alternativo/genética , Secuencia de Aminoácidos/genética , Animales , Secuencia de Bases/genética , Cromosomas Humanos Par 2/genética , ADN Complementario/análisis , ADN Complementario/genética , Canales de Potasio de Tipo Rectificador Tardío , Humanos , Potenciales de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Oocitos/metabolismo , Filogenia , Canales de Potasio/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/metabolismo , Ratas , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Canales de Potasio Shab , Vísceras/metabolismo , Xenopus
15.
Brain Res Mol Brain Res ; 102(1-2): 18-27, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12191490

RESUMEN

The human KCNK4 gene encodes several transcripts that generate two-pore K+ channel subunits. We describe the identification and cloning of two transcripts of this gene: human KT4.1a (HKT4.1a) and HKT4.1b. They encode proteins of 393 and 419 amino acids, respectively. HKT4.1a and mouse TRAAK (mTRAAK) are 83% identical, polymerase chain reaction (PCR) experiments performed with rat and human samples as well as the comparison of the HKT4.1 and mTRAAK UTRs strongly suggest that both the human and mouse cDNAs are products of ortholog genes. In contrast to the reported exclusive expression mTRAAK in the nervous system, human and rat KCNK4 gene products are expressed widely in several tissues. Northern blot analysis revealed the presence of three bands of 1.9, 3.0, and 4.8 kb in human, while in rat four bands of 1.8, 3.6, 5.2 and 8.6 kb were observed. Human KCNK4 transcripts were expressed mainly in the heart and brain but also in the liver, skeletal muscle, kidney and pancreas. In rat, the transcripts were strongly expressed in the brain but were also detected in the lung, kidney, liver, spleen, skeletal muscle, testes and at lower levels in the heart. Expression of HKT4.1b in Xenopus oocytes drives the resting potential close to the potassium equilibrium voltage. The expressed channels are not gated by voltage and are permanently open. The channels are not blocked by the classical K+ channel blockers TEA, 4-AP, Cs+, Ba++, quinine or quinidine. Analysis of genomic sequences reveals that seven exons participate to produce HKT4.1a and 11 exons to produce HKT4.1b cDNAs. The KCNK4 gene maps to chromosome 11q13.


Asunto(s)
Empalme Alternativo/genética , Membrana Celular/metabolismo , Células Eucariotas/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/aislamiento & purificación , Regiones no Traducidas 5'/genética , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , Clonación Molecular , Femenino , Regulación de la Expresión Génica/genética , Humanos , Potenciales de la Membrana/genética , Datos de Secuencia Molecular , Oocitos , Canales de Potasio/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Xenopus
16.
Cancer Cell ; 20(1): 104-18, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21741600

RESUMEN

To metastasize, a tumor cell must acquire abilities such as the capacity to colonize new tissue and evade immune surveillance. Recent evidence suggests that microRNAs can promote the evolution of malignant behaviors by regulating multiple targets. We performed a microRNA analysis of human melanoma, a highly invasive cancer, and found that miR-30b/30d upregulation correlates with stage, metastatic potential, shorter time to recurrence, and reduced overall survival. Ectopic expression of miR-30b/30d promoted the metastatic behavior of melanoma cells by directly targeting the GalNAc transferase GALNT7, resulted in increased synthesis of the immunosuppressive cytokine IL-10, and reduced immune cell activation and recruitment. These data support a key role of miR-30b/30d and GalNAc transferases in metastasis, by simultaneously promoting cellular invasion and immunosuppression.


Asunto(s)
Tolerancia Inmunológica/genética , MicroARNs/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Animales , Secuencia de Bases , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genoma Humano/genética , Humanos , Interleucina-10/metabolismo , Melanoma/enzimología , Melanoma/genética , Melanoma/patología , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , N-Acetilgalactosaminiltransferasas/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polisacáridos/metabolismo , Regulación hacia Arriba , Polipéptido N-Acetilgalactosaminiltransferasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA