Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Hered ; 112(5): 458-468, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34132805

RESUMEN

In North American gray wolves, black coat color is dominantly inherited via a 3 base pair coding deletion in the canine beta defensin 3 (CBD103) gene. This 3 base pair deletion, called the KB allele, was introduced through hybridization with dogs and subsequently underwent a selective sweep that increased its frequency in wild wolves. Despite apparent positive selection, KBB wolves have lower fitness than wolves with the KyB genotype, even though the 2 genotypes show no observable differences in black coat color. Thus, the KB allele is thought to have pleiotropic effects on as-yet unknown phenotypes. Given the role of skin-expressed CBD103 in innate immunity, we hypothesized that the KB allele influences the keratinocyte gene expression response to TLR3 pathway stimulation and/or infection by canine distemper virus (CDV). To test this hypothesis, we developed a panel of primary epidermal keratinocyte cell cultures from 24 wild North American gray wolves of both Kyy and KyB genotypes. In addition, we generated an immortalized Kyy line and used CRISPR/Cas9 editing to produce a KyB line on the same genetic background. We assessed the transcriptome-wide responses of wolf keratinocytes to the TLR3 agonist polyinosinic:polycytidylic acid (polyI:C), and to live CDV. K locus genotype did not predict the transcriptional response to either challenge, suggesting that variation in the gene expression response does not explain pleiotropic effects of the KB allele on fitness. This study supports the feasibility of using cell culture methods to investigate the phenotypic effects of naturally occurring genetic variation in wild mammals.


Asunto(s)
Virus del Moquillo Canino , Lobos , Alelos , Animales , Virus del Moquillo Canino/genética , Perros , Expresión Génica , Receptor Toll-Like 3/genética , Lobos/genética
2.
PLoS One ; 8(10): e78979, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24205356

RESUMEN

Overexpression of the basement membrane protein Laminin γ2 (Lamγ2) is a feature of many epidermal and oral dysplasias and all invasive squamous cell carcinomas (SCCs). This abnormality has potential value as an immunohistochemical biomarker of premalignancy but its mechanism has remained unknown. We recently reported that Lamγ2 overexpression in culture is the result of deregulated translation controls and depends on the MAPK-RSK signaling cascade. Here we identify eIF4B as the RSK downstream effector responsible for elevated Lamγ2 as well as MYC protein in neoplastic epithelial cells. Premalignant dysplastic keratinocytes, SCC cells, and keratinocytes expressing the E6 oncoprotein of human papillomavirus (HPV) type 16 displayed MAPK-RSK and mTOR-S6K1 activation and overexpressed Lamγ2 and MYC in culture. Immunohistochemical staining of oral dysplasias and SCCs for distinct, RSK- and S6K1-specific S6 phosphorylation events revealed that their respective upstream pathways become hyperactive at the same time during neoplastic progression. However, pharmacologic kinase inhibitor studies in culture revealed that Lamγ2 and MYC overexpression depends on MAPK-RSK activity, independent of PI3K-mTOR-S6K1. eIF4B knockdown reduced Lamγ2 and MYC protein expression, consistent with the known requirement for eIF4B to translate mRNAs with long, complex 5' untranslated regions (5'-UTRs). Accordingly, expression of a luciferase reporter construct preceded by the Lamγ2 5'-UTR proved to be RSK-dependent and mTOR-independent. These results demonstrate that RSK activation of eIF4B is causally linked to elevated Lamγ2 and MYC protein levels during neoplastic progression to invasive SCC. These findings have potential clinical significance for identifying premalignant lesions and for developing targeted drugs to treat SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Laminina/metabolismo , Sistema de Señalización de MAP Quinasas , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/fisiología , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Inmunohistoquímica , Queratinocitos/metabolismo , Laminina/genética , Invasividad Neoplásica/genética , Procesos Neoplásicos , ARN Mensajero/metabolismo
3.
Mod Pathol ; 26(11): 1498-507, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23765247

RESUMEN

As life expectancy lengthens, cases of non-viral-associated vulvar squamous cell carcinoma and its precursor lesion, so-called differentiated vulvar intraepithelial neoplasia (VIN), continue to increase in frequency. Differentiated VIN often is difficult to recognize and failure to detect it before invasion results in morbidity and mortality. Thus, identification of a reliable biomarker for this type of lesion would be of great clinical benefit. Our recent studies have identified activation (ser235/236 phosphorylation) of ribosomal protein S6 (p-S6) in basal epithelial cells as an event that precedes and accompanies laminin γ(2) overexpression in most preinvasive oral dysplasias. To test this as a potential biomarker of vulvar dysplasia, we immunostained seven differentiated VINs and nine papillomavirus-related 'classic' VINs, most of which were associated with carcinoma, for p-S6. All carcinomas, all differentiated VINs, and most classic VINs contained regions of p-S6 staining in the basal layer, whereas basal and parabasal cells of normal vulvar epithelium and hyperplastic and inflamed lesions lacking cellular atypia were p-S6 negative. Laminin γ(2) was expressed in a subset of VINs, always occurring within basal p-S6 positive regions, as we had found previously for oral dysplasias. Lichen sclerosus is considered a potential precursor of vulvar carcinoma. Two lichen sclerosus lesions of patients with a concurrent carcinoma and one of six lichen sclerosus lesions without atypia or known concurrent carcinoma were basal p-S6 positive. In summary, there is a distinct difference in p-S6 basal cell layer staining between benign and neoplastic vulvar squamous epithelium, with consistent staining of differentiated VIN and of some lichen sclerosus lesions. These results support further studies to assess the potential of p-S6 as a biomarker to identify vulvar lesions at risk of progressing to invasive cancer.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Carcinoma/química , Células Epiteliales/química , Inmunohistoquímica , Lesiones Precancerosas/química , Proteína S6 Ribosómica/análisis , Neoplasias de la Vulva/química , Anciano , Anciano de 80 o más Años , Carcinoma/patología , Carcinoma in Situ/patología , Progresión de la Enfermedad , Células Epiteliales/patología , Femenino , Humanos , Laminina/análisis , Persona de Mediana Edad , Invasividad Neoplásica , Fosforilación , Lesiones Precancerosas/patología , Valor Predictivo de las Pruebas , Factores de Riesgo , Liquen Escleroso Vulvar/metabolismo , Liquen Escleroso Vulvar/patología , Neoplasias de la Vulva/patología
4.
Am J Pathol ; 180(6): 2462-78, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22546478

RESUMEN

Lesions displaying a variety of dysplastic changes precede invasive oral and epidermal squamous cell carcinoma (SCC); however, there are no histopathological criteria for either confirming or staging premalignancy. SCCs and dysplasias frequently contain cells that abnormally express the γ2 subunit of laminin-332. We developed cell culture models to investigate γ2 dysregulation. Normal human keratinocytes displayed density-dependent repression of γ2, whereas premalignant keratinocytes and SCC cells overexpressed γ2 and secreted laminin assembly intermediates. Neoplastic cells had hyperactive EGFR/MAPK(ERK) signaling coordinate with overexpressed γ2, and EGFR and MEK inhibitors normalized γ2 expression. Keratinocytes engineered to express HPV16 E6 or activated mutant HRAS, cRAF1, or MEK1 lost density repression of γ2 and shared with neoplastic cells signaling abnormalities downstream of ERK, including increased phosphorylation of S6 and eIF4 translation factors. Notably, qPCR results revealed that γ2 overexpression was not accompanied by increased γ2 mRNA levels, consistent with ERK-dependent, eIF4B-mediated translation initiation of the stem-looped, 5'-untranslated region of γ2 mRNA in neoplastic cells. Inhibitors of MEK, but not of TORC1/2, blocked S6 and eIF4B phosphorylation and γ2 overexpression. Immunostaining of oral dysplasias identified γ2 overexpression occurring within fields of basal cells that had elevated p-S6 levels. These results reveal a causal relationship between ERK-dependent translation factor activation and laminin γ2 dysregulation and identify new markers of preinvasive neoplastic change during progression to SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Laminina/biosíntesis , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Neoplasias de la Boca/metabolismo , Lesiones Precancerosas/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Queratinocitos/metabolismo , Laminina/genética , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Modificación Traduccional de las Proteínas/fisiología , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Quinasas raf/fisiología , Proteínas ras/fisiología
5.
In Vitro Cell Dev Biol Anim ; 46(10): 841-55, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21042878

RESUMEN

Keratinocytes migrating from a wound edge or initiating malignant invasion greatly increase their expression of the basement membrane protein Laminin-322 (Lam332). In culture, keratinocytes initiate sustained directional hypermotility when plated onto an incompletely processed form of Lam332 (Lam332') or when treated with transforming growth factor beta (TGF-ß), an inducer of Lam332 expression. The development and tissue architecture of stratified squamous and prostate epithelia are very different, yet the basal cells of both express p63, α6ß4 integrin, and Lam332. Keratinocytes and prostate epithelial cells grow well in nutritionally optimized culture media with pituitary extract and certain mitogens. We report that prostate epithelial cells display hypermotility responses indistinguishable from those of keratinocytes. Several culture medium variables attenuated TGF-ß-induced hypermotility, including Ca(++), serum, and some pituitary extract preparations, without impairing growth, TGF-ß growth inhibition, or hypermotility on Lam322'. Distinct from its role as a mitogen, EGF proved to be a required cofactor for TGF-ß-induced hypermotility and could not be replaced by HGF or KGF. Prostate epithelial cells have a short replicative lifespan, restricted both by p16(INK4A) and telomere-related mechanisms. We immortalized the normal prostate epithelial cell line HPrE-1 by transduction to express bmi1 and TERT. Prostate epithelial cells lose expression of p63, ß4 integrin, and Lam332 when they transform to invasive carcinoma. In contrast, HPrE-1/bmi1/TERT cells retained expression of these proteins and normal TGF-ß signaling and hypermotility for >100 doublings. Thus, keratinocytes and prostate epithelial cells possess common hypermotility and senescence mechanisms and immortalized prostate cell lines can be engineered using defined methods to yield cells retaining normal properties.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Movimiento Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/fisiología , Queratinocitos/fisiología , Próstata/citología , Factor de Crecimiento Transformador beta/metabolismo , Western Blotting , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/metabolismo , Humanos , Inmunohistoquímica , Queratinocitos/metabolismo , Masculino , Retroviridae , Transducción Genética , Kalinina
6.
Clin Cancer Res ; 16(10): 2729-39, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20460482

RESUMEN

PURPOSE: The target antigens of graft-versus-leukemia that are tumor associated are incompletely characterized. EXPERIMENTAL DESIGN: We examined responses developing against CML66, an immunogenic antigen preferentially expressed in myeloid progenitor cells identified from a patient with chronic myelogenous leukemia who attained long-lived remission following CD4+ donor lymphocyte infusion (DLI). RESULTS: From this patient, CML66-reactive CD8+ T-cell clones were detected against an endogenously presented HLA-B*4403-restricted epitope (HDVDALLW). Neither CML66-specific antibody nor T-cell responses were detectable in peripheral blood before DLI. However, by 1 month after DLI, CD8+ T cells were present in peripheral blood and at 10-fold higher frequency in marrow. Subsequently, plasma antibody to CML66 developed in association with disease remission. Donor-derived CML66-reactive T cells were detected at low levels in vivo in marrow before DLI by ELISpot and by a nested PCR-based assay to detect clonotypic T-cell receptor sequences but not in blood of the patient pre-DLI nor of the graft donor. CONCLUSIONS: CD4+ DLI results in rapid expansion of preexisting marrow-resident leukemia-specific donor CD8+ T cells, followed by a cascade of antigen-specific immune responses detectable in blood. Our single-antigen analysis thus shows that durable posttransplant tumor immunity is directed in part against nonpolymorphic overexpressed leukemia antigens that elicit coordinated cellular and humoral immunity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/trasplante , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Transfusión de Linfocitos , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígenos HLA/inmunología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Activación de Linfocitos/inmunología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/terapia , Reacción en Cadena de la Polimerasa , Linfocitos T/inmunología
7.
Nature ; 460(7259): 1145-8, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19668190

RESUMEN

The overexpression of defined transcription factors in somatic cells results in their reprogramming into induced pluripotent stem (iPS) cells. The extremely low efficiency and slow kinetics of in vitro reprogramming suggest that further rare events are required to generate iPS cells. The nature and identity of these events, however, remain elusive. We noticed that the reprogramming potential of primary murine fibroblasts into iPS cells decreases after serial passaging and the concomitant onset of senescence. Consistent with the notion that loss of replicative potential provides a barrier for reprogramming, here we show that cells with low endogenous p19(Arf) (encoded by the Ink4a/Arf locus, also known as Cdkn2a locus) protein levels and immortal fibroblasts deficient in components of the Arf-Trp53 pathway yield iPS cell colonies with up to threefold faster kinetics and at a significantly higher efficiency than wild-type cells, endowing almost every somatic cell with the potential to form iPS cells. Notably, the acute genetic ablation of Trp53 (also known as p53) in cellular subpopulations that normally fail to reprogram rescues their ability to produce iPS cells. Our results show that the acquisition of immortality is a crucial and rate-limiting step towards the establishment of a pluripotent state in somatic cells and underscore the similarities between induced pluripotency and tumorigenesis.


Asunto(s)
Reprogramación Celular/fisiología , Senescencia Celular/fisiología , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular , División Celular , Línea Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Humanos , Queratinocitos , Cinética , Ratones , Ratones SCID , Células Madre Pluripotentes/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
8.
Stem Cells ; 27(6): 1388-99, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19489101

RESUMEN

Human embryonic stem (hES) cells can generate cells expressing p63, K14, and involucrin, which have been proposed to be keratinocytes. Although these hES-derived, keratinocyte-like (hESderK) cells form epithelioid colonies when cultured in a fibroblast feeder system optimal for normal tissue-derived keratinocytes, they have a very short replicative lifespan unless engineered to express HPV16 E6E7. We report here that hESderK cells undergo senescence associated with p16(INK4A) expression, unrelated to telomere status. Transduction to express bmi1, a repressor of the p16(INK4A)/p14(ARF) locus, conferred upon hESderK cells and keratinocytes a substantially extended lifespan. When exposed to transforming growth factor beta or to an incompletely processed form of Laminin-332, three lifespan-extended or immortalized hESderK lines that we studied became directionally hypermotile, a wound healing and invasion response previously characterized in keratinocytes. In organotypic culture, hESderK cells stratified and expressed involucrin and K10, as do epidermal keratinocytes in vivo. However, their growth requirements were less stringent than keratinocytes. We then extended the comparison to endoderm-derived, p63(+)/K14(+) urothelial and tracheobronchial epithelial cells. Primary and immortalized lines of these cell types had growth requirements and hypermotility responses similar to keratinocytes and bmi1 expression facilitated their immortalization by engineering to express the catalytic subunit of telomerase (TERT). In organotypic culture, they stratified and exhibited squamous metaplasia, expressing involucrin and K10. Thus, hESderK cells proved to be distinct from all three normal p63(+) cell types tested. These results indicate that hESderK cells cannot be identified conclusively as keratinocytes or even as ectodermal cells, but may represent an incomplete form of, or deviation from, normal p63(+) lineage development.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Senescencia Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Western Blotting , Línea Celular , Linaje de la Célula , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Queratinocitos/citología , Queratinocitos/metabolismo
9.
Am J Pathol ; 168(6): 1821-37, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16723698

RESUMEN

Keratinocytes become migratory to heal wounds, during early neoplastic invasion, and when undergoing telomere-unrelated senescence in culture. All three settings are associated with expression of the cell cycle inhibitor p16INK4A (p16) and of the basement membrane protein laminin 5 (LN5). We have investigated cause-and-effect relationships among laminin 5, p16, hypermotility, and growth arrest. Plating primary human keratinocytes on the gamma2 precursor form of laminin 5 (LN5') immediately induced directional hypermotility at approximately 125 microm/hour, followed by p16 expression and growth arrest. Cells deficient in p16 and either p14ARF or p53 became hypermotile in response to LN5' but did not arrest growth. Plating on LN5' triggered smad nuclear translocation, and all LN5' effects were blocked by a transforming growth factor (TGF) beta receptor I (TGFbetaRI) kinase inhibitor. In contrast, plating cells on collagen I triggered a TGFbetaRI kinase-independent hypermotility unaccompanied by smad translocation or growth arrest. Plating on control surfaces with TGFbeta induced hypermotility after a 1-day lag time and growth arrest by a p16-independent mechanism. Keratinocytes serially cultured with TGFbetaRI kinase inhibitor exhibited an extended lifespan, and immortalization was facilitated following transduction to express the catalytic subunit of telomerase (TERT). These results reveal fundamental features of a keratinocyte hyper-motility/growth-arrest response that is activated in wound healing, tumor suppression, and during serial culture.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Técnicas de Cultivo de Célula/métodos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Queratinocitos/metabolismo , Cicatrización de Heridas , Movimiento Celular , Proliferación Celular , Senescencia Celular , Femenino , Humanos , Masculino , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Kalinina
10.
Proc Natl Acad Sci U S A ; 103(6): 1792-7, 2006 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-16446420

RESUMEN

Cells of the human embryonic stem (hES) cell line H9, when cultured in the form of embryoid bodies, give rise to cells with markers of the keratinocyte of stratified squamous epithelia. Keratinocytes also form in nodules produced in scid mice by injected H9 cells; the hES-derived keratinocytes could be recovered in culture, where their colonies underwent a peculiar form of fragmentation. Whether formed from embryoid bodies or in nodules, hES-derived keratinocytes differed from postnatal keratinocytes in their much lower proliferative potential in culture; isolated single keratinocytes could not be expanded into mass cultures. Although their growth was not improved by transduction with the hTERT gene, these keratinocytes were immortalized by transduction with the E6E7 genes of HPV16. Clonally derived lines isolated from E6E7-transduced keratinocytes continued to express markers of the keratinocyte lineage, but the frequency with which they terminally differentiated was reduced compared with keratinocytes cultured from postnatal human epidermis. If other hES-derived somatic cell types also prove to be restricted in growth potential, not identical to the corresponding postnatal cell types, and to require immortalization for clonal isolation and expansion, these properties will have to be considered in planning their therapeutic use.


Asunto(s)
Queratinocitos/citología , Células Madre/citología , Diferenciación Celular , Línea Celular , Proliferación Celular , Separación Celular , Epitelio , Humanos , Queratinocitos/metabolismo , Fosforilación , Proteína de Retinoblastoma/metabolismo , Células Madre/metabolismo
11.
J Investig Dermatol Symp Proc ; 10(2): 72-85, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16358814

RESUMEN

The replicative lifespan of human keratinocytes in culture is restricted by a telomere-unrelated induction of p16INK4A (p16) and p14ARF. We have found that, in vivo, p16 is expressed by epidermal and oral keratinocytes at the migrating fronts of healing wounds and at the stromal interface of severely dysplastic and early invasive lesions and that such cells also invariably display increased expression of Laminin 5 (Lam5). In culture, p16 and Lam5 are coexpressed in keratinocytes at senescence, at the edges of wounds made in confluent cultures, and when cells are plated on dishes coated with the gamma2 precursor form of Lam5 (Lam5gamma2pre). Lam5/p16 coexpression in all three in vitro settings is associated with directional hypermotility and growth arrest. Hypermotility and growth arrest are uncoupled in p16- and p14ARF/p53-deficient keratinocytes and squamous cell carcinoma (SCC) cells; such cells become hypermotile is response to Lam5gamma2pre but do not growth arrest. Thus, the Lam5/p16 response is activated in normal wound healing, causing growth arrest of migratory keratinocytes that lead wound reepithelialization. This response also becomes activated at a critical stage of neoplastic progression, acting as a tumor suppressor mechanism. Rare premalignant cells that lose p16 remain motile and proliferative, thereby resulting in invasive growth as SCC.


Asunto(s)
Carcinoma de Células Escamosas/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Queratinocitos/química , Laminina/análisis , Neoplasias Glandulares y Epiteliales/patología , Cicatrización de Heridas , Línea Celular , Movimiento Celular , Proliferación Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Progresión de la Enfermedad , Humanos , Queratinocitos/fisiología , Laminina/fisiología , Factor de Crecimiento Transformador beta/farmacología , Proteína p53 Supresora de Tumor/fisiología
12.
Am J Pathol ; 166(6): 1827-40, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15920167

RESUMEN

Malignant pleural mesothelioma (MPM) is a highly lethal, poorly understood neoplasm that is typically associated with asbestos exposure. We performed transcriptional profiling using high-density oligonucleotide microarrays containing approximately 22,000 genes to elucidate potential molecular and pathobiological pathways in MPM using discarded human MPM tumor specimens (n = 40), normal lung specimens (n = 4), normal pleura specimens (n = 5), and MPM and SV40-immortalized mesothelial cell lines (n = 5). In global expression analysis using unsupervised clustering techniques, we found two potential subclasses of mesothelioma that correlated loosely with tumor histology. We also identified sets of genes with expression levels that distinguish between multiple tumor subclasses, normal and tumor tissues, and tumors with different morphologies. Microarray gene expression data were confirmed using quantitative reverse transcriptase-polymerase chain reaction and protein analysis for three novel candidate oncogenes (NME2, CRI1, and PDGFC) and one candidate tumor suppressor (GSN). Finally, we used bioinformatics tools (ie, software) to create and explore complex physiological pathways. Combined, all of these data may advance our understanding of mesothelioma tumorigenesis, pathobiology, or both.


Asunto(s)
Perfilación de la Expresión Génica , Genes Supresores de Tumor , Mesotelioma/genética , Oncogenes , Neoplasias Pleurales/genética , Western Blotting , Línea Celular Tumoral , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
13.
J Biol Chem ; 279(24): 25838-48, 2004 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-15070899

RESUMEN

Variola, the causative agent of smallpox, is a highly infectious double-stranded DNA virus of the orthopox genus that replicates within the cytoplasm of infected cells. For unknown reasons prominent skin manifestations, including "pox," mark the course of this systemic human disease. Here we characterized smallpox growth factor (SPGF), a protein containing an epidermal growth factor (EGF)-like domain that is conserved among orthopox viral genomes, and investigated its possible mechanistic link. We show that after recombinant expression, refolding, and purification, the EGF domain of SPGF binds exclusively to the broadly expressed cellular receptor, erb-B1 (EGF receptor), with subnanomolar affinity, stimulating the growth of primary human keratinocytes and fibroblasts. High affinity monoclonal antibodies specific for SPGF reveal in vivo immunoprotection in a murine vaccinia pneumonia model by a mechanism distinct from viral neutralization. These findings suggest that blockade of pathogenic factor actions, in general, may be advantageous to the infected host.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Sustancias de Crecimiento/fisiología , Virus de la Viruela/química , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Especificidad de Anticuerpos , Células Cultivadas , Secuencia Conservada , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Sustancias de Crecimiento/análisis , Sustancias de Crecimiento/química , Humanos , Péptidos y Proteínas de Señalización Intercelular , Interferón gamma/biosíntesis , Datos de Secuencia Molecular , Péptidos/metabolismo , Proteínas Virales/análisis , Proteínas Virales/química
15.
Int J Cancer ; 107(3): 407-15, 2003 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-14506741

RESUMEN

Infection of cervical keratinocytes by high-risk HPV is involved in the etiology of cervical carcinoma. Since viral products are immunogenic, development of cancer may require suppression of immune responses directed against infected epithelial cells. Many markers of host immune effector responses decrease as cervical intraepithelial neoplasia progresses. Among these is epithelial cell expression of the chemokine MCP-1, though the mechanism for its suppression is unclear. Here, we show that the E6 and E7 viral oncogenes from high-risk HPV, individually and together, suppress MCP-1 expression in primary epithelial cells derived from the female genital tract. This is not a consequence of global suppression of chemokine expression since other chemokines, including IP-10, IL-8 and RANTES, were less affected. Furthermore, 4 of 6 HPV-positive cervical carcinoma cell lines did not express MCP-1. Our data indicate that suppression of MCP-1 expression is part of the program of high-risk HPV E6/E7-induced transformation of primary epithelial cells. These observations are consistent with a model in which MCP-1 expression by infected keratinocytes, which would stimulate an immune attack on HPV-transformed cells, is suppressed for invasive cervical cancer to appear.


Asunto(s)
Cuello del Útero/inmunología , Quimiocina CCL2/antagonistas & inhibidores , Queratinocitos/inmunología , Proteínas Oncogénicas Virales/fisiología , Proteínas Represoras , Neoplasias del Cuello Uterino/inmunología , Células Cultivadas , Cuello del Útero/virología , Quimiocina CCL2/análisis , Quimiocinas/análisis , Células Epiteliales/inmunología , Femenino , Humanos , Queratinocitos/virología , Papillomaviridae/patogenicidad , Proteínas E7 de Papillomavirus , Factores de Tiempo , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/virología
16.
Am J Pathol ; 163(2): 477-91, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12875969

RESUMEN

The high frequency of mutation, deletion, and promoter silencing of the gene encoding p16(INK4A) (p16) in premalignant dysplasias and squamous cell carcinomas (SCC) of epidermis and oral epithelium classifies p16 as a tumor suppressor. However, the point during neoplastic progression at which this protein is expressed and presumably impedes formation of an SCC is unknown. Induction of p16 has been found to be responsible for the senescence arrest of normal human keratinocytes in culture, suggesting the possibility that excessive or spatially abnormal cell growth in vivo triggers p16 expression. We examined 73 skin and oral mucosal biopsy specimens immunohistochemically to test this hypothesis. p16 was not detectable in benign hyperplastic lesions, but instead was expressed heterogeneously in some dysplastic and carcinoma in situ lesions and consistently at areas of microinvasion and at superficial margins of advanced SCCs. p16-positive cells in these regions coexpressed the gamma2 chain of laminin 5, identified previously as a marker of invasion in some carcinomas. Normal keratinocytes undergoing senescence arrest in culture proved to coordinately express p16 and gamma2 and this was frequently associated with increased directional motility. Keratinocytes at the edges of wounds made in confluent early passage cultures also coexpressed p16 and gamma2, accompanying migration to fill the wound. These results have identified the point during neoplastic progression in stratified squamous epithelial at which the tumor suppressor p16 is expressed and suggest that normal epithelia may use the same mechanism to generate non-dividing, motile cells for wound repair.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Queratinocitos/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Movimiento Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Células Epidérmicas , Epidermis/metabolismo , Epidermis/patología , Humanos , Inmunohistoquímica , Queratinocitos/citología , Ratones , Mucosa Bucal/citología , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Invasividad Neoplásica , Subunidades de Proteína/metabolismo , Neoplasias Cutáneas/patología , Cicatrización de Heridas , Kalinina
17.
Mol Cell Biol ; 22(14): 5157-72, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12077343

RESUMEN

With increasing frequency during serial passage in culture, primary human keratinocytes express p16(INK4A) (p16) and undergo senescence arrest. Keratinocytes engineered to express hTERT maintain long telomeres but typically are not immortalized unless, by mutation or other heritable event, they avoid or greatly reduce p16 expression. We have confirmed that keratinocytes undergo p16-related senescence during growth in culture, whether in the fibroblast feeder cell system or in the specialized K-sfm medium formulation, and that this mechanism can act as a barrier to immortalization following hTERT expression. We have characterized the p16-related arrest mechanism more precisely by interfering specifically with several regulators of cell cycle control. Epidermal, oral mucosal, corneal limbal, and conjunctival keratinocytes were transduced to express a p16-insensitive mutant cdk4 (cdk4(R24C)), to abolish p16 control, and/or a dominant negative mutant p53 (p53DD), to abolish p53 function. Expression of either cdk4(R24C) or p53DD alone had little effect on life span, but expression of both permitted cells to divide 25 to 43 population doublings (PD) beyond their normal limit. Keratinocytes from a p16(+/-) individual transduced to express p53DD alone displayed a 31-PD life span extension associated with selective growth of variants that had lost the wild-type p16 allele. Cells in which both p53 and p16 were nonfunctional divided rapidly during their extended life span but experienced telomere erosion and ultimately ceased growth with very short telomeres. Expression of hTERT in these cells immortalized them. Keratinocytes engineered to express cdk4(R24C) and hTERT but not p53DD did not exhibit an extended life span. Rare immortal variants exhibiting p53 pathway defects arose from them, however, indicating that the p53-dependent component of keratinocyte senescence is telomere independent. Mutational loss of p16 and p53 has been found to be a frequent early event in the development of squamous cell carcinoma. Our results suggest that such mutations endow keratinocytes with extended replicative potential which may serve to increase the probability of neoplastic progression.


Asunto(s)
Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor/metabolismo , División Celular , Células Cultivadas , Medios de Cultivo , Quinasa 4 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Unión al ADN , Genes p53 , Humanos , Mutación , Telomerasa/genética , Telomerasa/metabolismo , Telómero/metabolismo , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...