Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Exp Dermatol ; 25(1): 38-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26308156

RESUMEN

Keloids are progressively expanding scars, mostly prevalent in individuals of African descent. Previous data identified increased mast cell number and activation state in keloids suggesting a role in disease progression. The major eicosanoid secreted by mast cells is prostaglandin D2 (PGD2), a relatively unstable pro-inflammatory mediator which can be spontaneously converted to 15-deoxy-(Delta12,14)-prostaglandin J2(15d-PGJ2) or enzymatically metabolized to 9α,11ß-PGF2 by aldo-keto reductase 1C3 (AKR1C3). In this work, we investigated the possible role of PGD2 and its metabolites in keloids using CRL1762 keloid fibroblasts (KF) and immunohistochemical staining. Our data suggested approximately 3-fold increase of tryptase-positive mast cell count in keloids compared with normal skin. Furthermore, AKR1C3 was overexpressed in the fibrotic area of keloids while relatively weak staining detected in normal skin. Metabolism of PGD2 to 9α,11ß-PGF2 by both, KF and normal fibroblasts, was dependent on AKR1C3 as this reaction was attenuated in the presence of the AKR1C3 inhibitor, 2'-hydroxyflavanone, or in cells with decreased AKR1C3 expression. 15d-PGJ2, but not the other tested PGs, inhibited KF proliferation, attenuated KF-mediated collagen gel contraction and increased caspase-3 activation. In addition, treatment with 15d-PGJ2 activated P38-MAPK, induced reactive oxygen species and upregulated superoxide dismutase-1 (SOD-1). Finally, inhibition of P38-MAPK further augmented 15d-PGJ2-induced caspase-3 cleavage and attenuated its effect on SOD-1 transcription. This work suggests that localized dual inhibition of AKR1C3 and P38-MAPK may inhibit keloid progression. Inhibiting AKR1C3 activity may generate oxidative environment due to redirection of PGD2 metabolism towards 15d-PGJ2 while inhibition of P38-MAPK will sensitize keloid cells to ROS-induced apoptosis.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Queloide/metabolismo , Prostaglandina D2/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Apoptosis , Caspasa 3/metabolismo , Proliferación Celular , Colágeno/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Mastocitos/metabolismo , Estrés Oxidativo , Reacción en Cadena de la Polimerasa , Prostaglandina D2/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo , Piel/embriología , Superóxido Dismutasa-1/metabolismo
2.
Nutr Cancer ; 62(1): 66-73, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20043261

RESUMEN

Selenium (Se) supplementation is reported to decrease the incidence and total mortality of cancer. Whereas in vitro and in vivo studies have shown a decrease in prostate, lung, and liver cancers, this has not been shown in thyroid cancer. ARO (anaplastic), NPA (BRAF positive papillary), WRO (BRAF negative papillary), and FRO (follicular) cells treated with 150 microM seleno-l-methionine (SM) were assessed for viability at 24, 48, and 72 h. Treated FRO cells were examined for cell cycle using flow cytometry, for apoptosis using terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay, and for gene expression using microarray. Genes identified as upregulated were confirmed by real-time PCR (RT-PCR) and proteins by Western blot analysis. SM treatment significantly decreased the proliferation of all cell lines. TUNEL assay showed no evidence of apoptosis, and flow cytometry showed a significant cell-cycle arrest in S (271% increase, P = 0.006) and G2/M (61% increase, P = 0.002) compared to control. Microarray revealed 21 differentially expressed genes with greater than twofold change. A relative overexpression of growth arrest and DNA damage inducible (GADD)34 and GADD153 in treated cells was confirmed with RT-PCR and Western blot. SM inhibits thyroid cancer cell proliferation through a time dependent upregulation of the GADD family of genes and arrest in S and G2/M phases of the cell cycle. This is the first report of selenium induced inhibition of thyroid cancer cell growth.


Asunto(s)
Antígenos de Diferenciación/genética , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Selenio/farmacología , Neoplasias de la Tiroides/patología , Factor de Transcripción CHOP/genética , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/genética , Receptores ErbB/análisis , Receptores ErbB/genética , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Análisis por Micromatrices , Proteína Fosfatasa 1 , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenometionina/farmacología , Neoplasias de la Tiroides/química , Factor de Transcripción CHOP/análisis
3.
Oncol Rep ; 21(4): 1017-21, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19288003

RESUMEN

Clinical familial adenomatous polyposis (FAP) syndrome represents a high risk pre-invasive precursor for colon cancer, and is characterized by germ line mutation in the adenomatous polyposis coli (APC) tumor suppressor gene. Cellular models with relevant genetic and biological characteristics should provide important mechanistic leads for predisposition and preventive intervention. Cloned colon epithelial cell line from the Apc850 Min/+ mouse represented a model for FAP. Cell cycle progression, cellular apoptosis and anchorage-independent growth represented the biomarkers for carcinogenic risk. The Apc mutant 850Min COL-Cl1 cells exhibited decreased G0/G1:S+G2/M ratio, increased S+G2/M:subG0 ratio, and increased anchorage-independent colony formation, indicating loss of homeostatic growth control and gain of anchorage-independent growth. Growth of these cells in serum-depleted medium was promoted by mitogenic insulin and epidermal growth factor, and inhibited by anti-mitogenic transforming growth factor-beta1 and dexamethasone. Treatment with low dose combinations of synthetic enzyme inhibitor difluoro methylornithine (DFMO), synthetic non-steroidal anti-inflammatory drug sulindac (SUL), and naturally occurring epigallocatechin gallate (EGCG), and eicosapen-taenoic acid (EPA) produced cytostatic growth arrest and inhibited anchorage-independent colony formation. These data identify a novel cell culture model and validate a mechanism-based approach to prioritize combinations of effective chemopreventive compounds for prevention/therapy of colon cancer.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Neoplasias del Colon/prevención & control , Poliposis Adenomatosa del Colon/patología , Animales , Anticarcinógenos/uso terapéutico , Catequina/análogos & derivados , Catequina/uso terapéutico , Línea Celular , Eflornitina/uso terapéutico , Ácido Eicosapentaenoico/uso terapéutico , Genes APC , Ratones , Ratones Endogámicos C57BL , Mutación
4.
J Cell Biochem ; 104(5): 1781-92, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18348204

RESUMEN

Curcumin (diferuloymethane), a natural compound used in traditional medicine, exerts an antiproliferative effect on various tumor cell lines by an incompletely understood mechanism. It has been shown that low doses of curcumin downregulate DNA topoisomerase II alpha (TOP2A) which is upregulated in many malignances. The activity of TOP2A is required for RNA polymerase II transcription on chromatin templates. Recently, it has been reported that CTCF, a multifunctional transcription factor, recruits the largest subunit of RNA polymerase II (LS Pol II) to its target sites genome-wide. This recruitment of LS Pol II is more pronounced in proliferating cells than in fully differentiated cells. As expression of imprinted genes is often altered in tumors, we investigated the potential effect of curcumin treatment on transcription of the imprinted H19 gene, located distally from the CTCF binding site, in human tumor cell lines HCT 116, SW 620, HeLa, Cal 27, Hep-2 and Detroit 562. Transcription of TOP2A and concomitantly H19 was supressed in all tumor cell lines tested. Monoallelic IGF2 expression was maintained in curcumin-treated cancer cells, indicating the involvement of mechanism/s other than disturbance of CTCF insulator function at the IGF2/H19 locus. Curcumin did not alter H19 gene transcription in primary cell cultures derived from normal human tissues.


Asunto(s)
Curcumina/farmacología , Regulación hacia Abajo/efectos de los fármacos , ARN no Traducido/genética , Transcripción Genética/efectos de los fármacos , Alelos , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Factor II del Crecimiento Similar a la Insulina/genética , NAD/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , ARN Largo no Codificante
5.
Oncol Rep ; 17(4): 909-14, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17342335

RESUMEN

Germ line mutations in the tumor suppressor adenomatous polyposis coli (APC) gene, predispose for the clinical familial adenomatous polyposis (FAP) syndrome, a high risk precursor for early onset colon cancer. Similar mutations in the murine homolog of the APC gene, however, produce adenomas predominantly in the small intestine, rather than in the colon. The objectives of the present study were: i) to develop a preclinical cell culture model for human FAP syndrome and ii) to validate this model as a rapid mechanism-based approach for evaluation of the preventive efficacy of combinations of synthetic pharmacological agents or naturally-occurring phytochemicals, for the risk of colon carcinogenesis. The clonally selected 850Min COL-Cl1 cell line derived from histologically normal colon of ApcMin/+ mouse exhibited aberrant proliferation (64.7% decrease in population doubling time, 820% increase in saturation density, and 81.4% decrease in spontaneous apoptosis), relative to that observed in the colon epithelial cell line C57 COL established from Apc [+/+] C57BL/6J mouse. In addition, unlike the Apc [+/+] C57 COL cells, the Apc mutant cells exhibited enhanced risk for spontaneous carcinogenic transformation as evidenced by 100% increase in anchorage-independent colony formation (C57 COL: 0/12; 850Min COL-Cl1: 12/12, mean colony number 23.6+/-2.7). Treatment of Apc mutant cells with low dose combination of select mechanistically distinct synthetic chemopreventive agents such as celecoxib (CLX) + difluoro methylornithine (DFMO), or naturally-occurring epigallocatechin gallate (EGCG) + curcumin (CUR) produced 160-400% and 220-430% decrease in the viable cell number respectively, relative to these agents used independently. Furthermore, relative to independent agents, CLX+DFMO and EGCG+CUR combinations produced 31.5-82.1% and 45.9-105.4% greater reduction in the number of anchorage-independent colonies. Thus, aberrant proliferation and increased risk for carcinogenesis in the Apc mutant cells, and their susceptibility to low dose combinations of mechanistically distinct chemopreventive agents validate a rapid approach to prioritize efficacious combinations for long-term animal studies and future clinical trials on prevention of colon cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica , Neoplasias del Colon/prevención & control , Modelos Biológicos , Lesiones Precancerosas/prevención & control , Proliferación Celular , Quimioprevención , Evaluación Preclínica de Medicamentos , Humanos , Riesgo
6.
Oncol Lett ; 13(4): 2477-2482, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28454423

RESUMEN

The Luminal A subtype of breast cancer expresses the estrogen receptor (ER)-α and progesterone receptor (PR), but not the human epidermal growth factor receptor (HER)-2 oncogene. This subtype of breast cancer responds to endocrine therapy involving the use of selective estrogen receptor modulators and/or inhibitors of estrogen biosynthesis. However, these therapeutic agents are frequently associated with long-term systemic toxicity and acquired tumor resistance, emphasizing the need to identify non-toxic alternative treatments for chemo-endocrine therapy responsive breast cancer. The present study utilized the human mammary carcinoma-derived, ER+/PR+/HER-2- MCF-7 cell line as a model of the Luminal A subtype of breast cancer to examine the growth inhibitory effect of the Chinese nutritional herb Epimedium grandiflorum (EG) and determine the mechanisms underlying this effect. MCF-7 cells maintained in a serum-depleted culture medium retained their ability to grow in response to 17ß-estradiol (E2). Treatment of the MCF-7 cells with EG resulted in dose-dependent inhibition of E2-promoted growth. Mechanistically, EG inhibited E2-promoted cell cycle progression through G1 stage arrest and modulated the cellular metabolism of E2, increasing the formation of the anti-proliferative metabolites 2-hydroxyestrone and estriol. Long-term treatment of MCF-7 cells with EG inhibited E2-promoted anchorage independent growth, a surrogate in vitro biomarker of tumorigenesis. In conclusion, the results of the present study demonstrate the growth inhibitory effects of EG on MCF-7 cells and identified clinically relevant mechanistic leads for its anti-tumorigenic efficacy.

7.
Int J Oncol ; 28(6): 1523-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16685453

RESUMEN

Human colon cancer is a multi-factorial, multi-step disease wherein genetic and dietary factors represent important regulators of initiation, promotion and progression. While the etiology of sporadic colon cancer remains largely unidentified, familial adenomatous polyposis (FAP) and hereditary non-polyposis colon cancer (HNPCC) represent predisposing genetic syndromes for early-onset familial/hereditary colon cancer. These syndromes are characterized by germ-line mutations in the adenomatous polyposis coli (APC) and/or DNA mismatch repair genes, respectively. Currently available preclinical animal models for human FAP and HNPCC syndromes, expressing clinically relevant germ-line mutations, exhibit adenomas in the small intestine rather than in the colorectum. These models are, therefore, subject to extrapolation for direct clinical translatability of the data for colon carcinogenesis and chemoprevention. Experimental models expressing clinically relevant genetic defects (APC and/or DNA mismatch repair gene mutations) in an appropriate target site (colon) may represent novel approaches that reduce extrapolation of the data for their clinical relevance. This report provides an overview on carcinogenesis and chemoprevention in preclinical models of FAP and HNPCC syndromes, and summarizes recent data on i) development of new cell culture models for FAP and HNPCC syndromes; and ii) validation of developed models for rapid, mechanism-based screening of new pharmacological or naturally occurring chemopreventive agents.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/prevención & control , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/prevención & control , Disparidad de Par Base , Biomarcadores de Tumor/análisis , Mutación de Línea Germinal , Humanos
8.
Oncol Lett ; 12(5): 3949-3957, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27895755

RESUMEN

In estrogen receptor (ER)+ MCF-7 cells, ER represents a ligand-activated transcription factor, and 17ß-estradiol (E2) represents its physiological ligand. Maintenance of the human breast carcinoma-derived MCF-7 cells with 0.7% serum selected a proliferative sub-population of E2-responsive cells with transiently non-functional ER due to limited availability of E2. Culture of MCF-7 cells in the presence of either 0.7% serum, <1 nM E2 or 0.7% serum + 20 nM E2 selected isogenic cells with either non-functional ER (ER-NF) or functional ER (ER-F) phenotype. The two phenotypes responded to the growth-promoting effects of E2 and to the growth-inhibitory effects of the selective ER modulator tamoxifen, indicating retention of E2 responsiveness. Comparative dose-response experiments with Chinese nutritional herbs on ER-NF and ER-F cells identified the inhibitory concentration (IC)50 values for these herbs, while the IC50 ratios for the ER-NF:ER-F phenotypes facilitated their rank ordering in terms of efficacy. Out of the 11 efficacious herbs tested, five herbs exhibited ER-F > ER-NF inhibitory activity, four exhibited ER-F = ER-NF inhibitory activity and two exhibited ER-NF > ER-F inhibitory activity. Extracts from representative herbs, Lycium barbarum bark, Epimedium grandiflorum and Cornus officinalis, from each of the three groups inhibited anchorage-independent growth, induced G1 or G2/M arrest and/or apoptosis, and generated anti-proliferative E2 metabolites. The differential growth inhibition in ER-NF and ER-F phenotypes, together with the mechanistic efficacy of representative herbs, identified potential leads for their efficacy on ER+ and/or ER- breast cancer.

9.
Cancers (Basel) ; 7(3): 1586-604, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26287245

RESUMEN

Cutaneous Melanoma (CM) is a leading cause of cancer deaths, with reports indicating a rising trend in the incidence rate of melanoma among Hispanics in certain U.S. states. The level of melanin pigmentation in the skin is suggested to render photoprotection from the DNA-damaging effects of Ultraviolet Radiation (UVR). UVR-induced DNA damage leads to cytogenetic defects visualized as the formation of micronuclei, multinuclei and polymorphic nuclei in cells, and a hallmark of cancer risk. The causative relationship between Sun exposure and CM is controversial, especially in Hispanics and needs further evaluation. This study was initiated with melanocytes from White, Hispanic and Black neonatal foreskins which were exposed to UVR to assess their susceptibility to UVR-induced modulation of cellular growth, cytogenetic damage, intracellular and released melanin. Our results show that White and Hispanic skin melanocytes with similar levels of constitutive melanin are susceptible to UVR-induced cytogenetic damage, whereas Black skin melanocytes are not. Our data suggest that the risk of developing UVR-induced CM in a skin type is correlated with the level of cutaneous pigmentation and its ethnic background. This study provides a benchmark for further investigation on the damaging effects of UVR as risk for CM in Hispanics.

10.
Int J Oncol ; 22(3): 509-15, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12579302

RESUMEN

Human breast cancer is a multifactorial, multistep disease wherein genetic, endocrine and dietary factors represent crucial regulators of initiation, promotion and progression. Preclinical investigations utilizing human breast carcinoma derived cell lines either in culture, or upon xenotransplantation, have provided valuable leads for molecular pathogenesis of cancer progression and also for novel therapeutic modalities. The mechanistic significance of genetic factors on early events of initiation/promotion, however, is dependent on extrapolation, and is therefore, equivocal. Human tissue derived explant culture/cell culture models utilizing non-involved target tissue at risk for carcinogenic transformation provide a novel approach that minimizes extrapolation for clinical relevance and thereby maximizes the translational impact. This report provides an overview of laboratory investigations focused on: i) development of the model, ii) optimization of mechanistic biomarker assays for carcinogenic transformation, and iii) validation of the model as a high throughput mechanistic screen for preclinical efficacy of natural phytochemicals.


Asunto(s)
Anticarcinógenos/farmacología , Neoplasias de la Mama/prevención & control , Carcinoma Ductal/prevención & control , Ensayos de Selección de Medicamentos Antitumorales , Anticarcinógenos/aislamiento & purificación , Anticarcinógenos/uso terapéutico , Biomarcadores , Mama/citología , Mama/efectos de los fármacos , Neoplasias de la Mama/patología , Carcinoma Ductal/patología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/fisiología , Línea Celular Tumoral/efectos de los fármacos , Transformación Celular Neoplásica , Células Epiteliales/efectos de los fármacos , Femenino , Humanos , Plantas Comestibles/química
11.
Int J Oncol ; 21(4): 809-15, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12239620

RESUMEN

In the multistep progressive pathogenesis of human breast cancer, comedo ductal carcinoma in situ (DCIS) represents a preinvasive precursor lesion for therapy resistant invasive cancer. Human tissue derived cell culture models exhibiting molecular similarities to clinical DCIS facilitate an important preclinical mechanistic approach for evaluation of preventive efficacy of natural and synthetic chemopreventive compounds. Natural phytochemicals present in fresh fruits, vegetables and grain products are likely to offer protection against cancer. The clinical efficacy of these natural phytochemicals, however, depends on extrapolation, and is therefore equivocal. The present study determined whether the natural soy isoflavone genistein (GEN) inhibited aberrant proliferation in 184-B5/HER cells (a model for human comedo DCIS) and identified possible mechanisms responsible for its efficacy. Human reduction mammoplasty derived HER-2/neu oncogene expressing preneoplastic 184-B5/HER cells represented the experimental system. Flow cytometry and cellular epifluorescence based assays were utilized to quantitate the alterations in cell cycle progression, cellular apoptosis, and in the status of cell cycle regulatory and apoptosis-associated gene product expression. The 184-B5/HER cells exhibited specific immunofluorescence to p185HER, p53, EGFR, but not to ERalpha, thus resembling comedo DCIS. Treatment of 184-B5/HER cells with GEN resulted in a dose-dependent decrease in the viable cell population, increase in the G0/G1:S + G2/M ratio and enhancement of sub G0/G1 (apoptotic population). Exposure to the maximum cytostatic 10 microM dose of GEN down-regulated HER-2/neu mediated signal transduction as evidenced by a 73.9% decrease (p=0.001) in p185HER specific, and a 89.8% decrease (p=0.001) in phosphotyrosine specific immunofluorescence. The increase in G0/G1:S + G2/M ratio in response to the treatment with 10 microM GEN was associated with a 85.5% decrease (p=0.001) in immunoreactivity to PCNA and a 128.6% increase (p=0.004) in immunoreactivity to the cyclin dependent kinase inhibitor p16INK4. The induction of apoptosis by GEN was associated with a 52.8% decrease (p=0.001) in the immunoreactivity to antiapoptotic Bcl-2 and with a 195.9% (p=0.001) increase in the immunoreactivity to proapoptotic Bax. Thus, preventive efficacy of GEN in HER-2/neu+/ER- 184-B5/HER cells may be due to its ability to down-regulate HER-2/neu mediated signal transduction, increase the expression of the cyclin dependent kinase inhibitor p16INK4, and induce Bcl-2 dependent apoptosis. These data provide evidence that GEN may be a potential chemopreventive lead compound for human comedo DCIS. The 184-B5/HER cells, may therefore, provide a high throughput mechanistic bioassay to identify new chemopreventive agents for human breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Genisteína/farmacología , Isoflavonas/farmacología , Receptor ErbB-2/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , División Celular , ADN/metabolismo , Femenino , Humanos , Factores de Tiempo , Células Tumorales Cultivadas
12.
Int J Mol Med ; 10(4): 427-32, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12239589

RESUMEN

Germline mutations of the Apc tumor suppressor gene result in increased risk for gastrointestinal carcinogenesis. The Apc1638N [+/-] mouse exhibits accelerated gastrointestinal carcinogenesis that is modifiable by select pharmacological and dietary agents. Experiments in the present study were conducted on a subculturable epithelial 1638NCOL cell line established from histologically normal colon of Apc1638N [+/-] mouse to examine the effects of selected chemopreventive agents that differ in their mechanism of action. Extent of growth arrest, number of cell population doublings, cell cycle progression and aneuploid G0/G1: S + G2/M ratio represented the quantitative endpoints for the susceptibility and efficacy of chemopreventive agents. Treatment of exponentially growing 1638NCOL cells with maximum cytostatic dose of 9cisRA, DFMO or SUL (100 microM) produced a 60-70% growth arrest, that with TAM and AMF (10 microM) produced a 20-40% growth arrest, while that with OLT (100 microM) produced a 25% growth arrest. This response was associated with corresponding decrease in the number of cell population doubling. 9cisRA, SUL or AMF increased the aneuploid G0/G1: S + G2/M ratio by inducing G1 checkpoint arrest, while DFMO, TAM and OLT decreased the ratio by inducing G2 checkpoint arrest. Thus, cell cycle phase-dependent susceptibility of the Apc [+/-] 1638NCOL cell line to mechanistically distinct chemopreventive agents validates a novel colon epithelial cell culture model for mechanistic, preventive or therapeutic studies on Apc regulated colon carcinogenesis.


Asunto(s)
Anticarcinógenos/farmacología , Neoplasias del Colon/prevención & control , Epitelio/efectos de los fármacos , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , División Celular/efectos de los fármacos , Línea Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados
13.
Int J Oncol ; 42(2): 583-96, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23229765

RESUMEN

Gene expression profiling can be useful for phenotypic classification, investigation of functional pathways, and to facilitate the search for disease risk genes through the integration of transcriptional data with available genomic information. To enhance our understanding of the genetic and molecular basis of basal cell carcinoma (BCC) we performed global gene expression analysis to generate a disease-associated transcriptional profile. A gene signature composed of 331 differentially expressed genes (DEGs) was generated from comparing 4 lesional and 4 site-matched control samples using Affymetrix Human Genome U95A microarrays. Hierarchical clustering based on the obtained gene signature separated the samples into their corresponding phenotype. Pathway analysis identified several significantly overrepresented pathways including PPAR-γ signaling, TGF-ß signaling and lipid metabolism, as well as confirmed the importance of SHH and p53 in the pathogenesis of BCC. Comparison of our microarray data with previous microarray studies revealed 13 DEGs overlapping in 3 studies. Several of these overlapping genes function in lipid metabolism or are components of the extracellular matrix, suggesting the importance of these and related pathways in BCC pathogenesis. BCC-associated DEGs were mapped to previously reported BCC susceptibility loci including 1p36, 1q42, 5p13.3, 5p15 and 12q11-13. Our analysis also revealed transcriptional 'hot spots' on chromosome 5 which help to confirm (5p13 and 5p15) and suggest novel (5q11.2-14.3, 5q22.1-23.3 and 5q31-35.3) disease susceptibility loci/regions. Integrating microarray analyses with reported genetic information helps to confirm and suggest novel disease susceptibility loci/regions. Identification of these specific genomic and/or transcriptional targets may lead to novel diagnostic and therapeutic modalities.


Asunto(s)
Carcinogénesis/genética , Carcinoma Basocelular/genética , Regulación Neoplásica de la Expresión Génica , Metabolismo de los Lípidos/genética , Anciano , Anciano de 80 o más Años , Carcinoma Basocelular/patología , Perfilación de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Masculino , PPAR gamma/genética , PPAR gamma/metabolismo , Factor de Crecimiento Transformador beta/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Oncol Lett ; 3(4): 744-750, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22740986

RESUMEN

Clinical breast cancer progresses via a multi-step carcinogenic process wherein genetic, molecular, endocrine and dietary factors play significant roles in the pathogenesis, prevention and therapy of the disease. Preclinical cell culture models, expressing clinically relevant genetic and endocrine defects and exhibiting quantifiable cancer risk, may provide facile, clinically translatable approaches to identify molecular targets and susceptible mechanistic pathways for the efficacy of novel interventional approaches. This review summarizes laboratory investigations focused on i) developing murine and human mammary tissue-derived cell culture models; ii) optimizing mechanism-based quantitative endpoint biomarker assays specific for carcinogenic risk and preventive/therapeutic efficacy; and iii) providing quantifiable proof-of-principle evidence for validation of the present cell culture approaches, capable of prioritizing efficacious lead compounds for subsequent in vivo animal studies and clinical trials for the prevention/therapy of breast cancer. Epithelial cell culture models are developed and characterized where the carcinogenic process is initiated by the targeted expression of clinically relevant oncogenes. The cell culture systems from mouse mammary tissue are in vitro approaches that complement the Ras and Myc transgenic mouse models. The human mammary tissue-derived systems are in vitro models for chemoendocrine, therapy-resistant, clinical, pre-invasive ER(-)/PR(-)/HER-2(+) comedo ductal carcinoma in situ, ER(+)/PR(+) chemoendocrine therapy-responsive breast cancer and ER(-)/PR(-)/HER-2(-) triple-negative chemoendocrine, therapy-resistant breast cancer. The oncogene-initiated phenotypes exhibit loss of homeostatic growth control, downregulation of cell apoptosis and gain of carcinogenic risk in vitro, as well as transplantable tumor development in vivo. Numerous mechanistically distinct, synthetic pharmacological agents, as well as naturally occurring dietary compounds, re-establish homeostatic growth control via cell cycle arrest and/or induction of cell apoptosis, downregulate oncogene-mediated cell signaling pathways, modulate the expression of numerous cell cycle regulatory and apoptosis-specific proteins and reduce carcinogenic risk in pre-neoplastic and carcinoma-derived cell culture models. These data validate the present cell culture approaches as novel, mechanism-based screens to evaluate and prioritize promising lead compounds for the prevention/therapy of clinical breast cancer.

15.
Oncol Rep ; 25(5): 1195-201, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21399881

RESUMEN

Genetically engineered mice with targeted alterations in clinically relevant oncogenes, tumor suppressor genes or DNA mismatch repair genes provide unique predictive animal models for human carcinogenesis, and cancer prevention/therapy. However, some of the genetically engineered mouse models lack target organ specificity for colon carcinogenesis. We have established, characterized and validated stable epithelial cell lines from 'normal' and 'genetically' predisposed target organs that offer innovative and mechanistic approaches, complementing in vivo studies on existing animal models for clinical breast and colon cancer. Epithelial cell lines with up- regulated Ras or myc oncogene, mutated Apc tumor suppressor gene and Mlh1 DNA mismatch repair gene provide facile experimental systems for organ site carcinogenesis and cancer prevention. Altered expression of cancer specific biomarkers and their modulation by several synthetic pharmacological agents such as retinoids, selective estrogen receptor modulators, non-steroidal anti-inflammatory drugs and specific enzyme inhibitors have been reported from our laboratory. Oncogene expressing MMEC-Ras and MMEC-myc mammary epithelial cells, Apc mutant 850Min COL and 1638N COL, and DNA mismatch repair/Apc mutant Mlh1/1638N COL colon epithelial cells exhibit aberrant cell cycle progression, down-regulated apoptosis and enhanced carcinogenic risk in vitro and tumor formation in vivo. We have reported that relative to the parental 'normal' non-neoplastic cells, genetically 'altered' pre-neoplastic cells exhibit enhanced sensitivity for growth arrest by multiple mechanistically distinct pharmacological agents. Comparative experiments on isogenic 'normal' and genetically 'altered' target cell lines facilitate cancer selective efficacy and identification of susceptible mechanistic pathways. Treatment of these genetically 'altered' pre-neoplastic cells with low dose combination of mechanistically distinct pharmacological agents as well as naturally occurring phytochemicals induce cytostatic growth arrest, alter cell cycle progression and reduce carcinogenic risk. The availability of validated technology for model development, and for mechanism based biomarker assays now establishes a novel platform to rapidly test carcinogenicity and preventive/therapeutic efficacy of novel pharmacological agents as well as naturally occurring phytochemicals. Thus, these data permit rational prioritization of efficacious lead compounds for preclinical testing and future clinical trials for prevention/therapy of breast and colon cancer.


Asunto(s)
Neoplasias de la Mama , Línea Celular Tumoral , Neoplasias del Colon , Animales , Anticarcinógenos/farmacología , Biomarcadores de Tumor , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones Transgénicos
17.
ALTEX ; 24(1): 16-21, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17361317

RESUMEN

Mouse models for colon cancer that harbor a germ line mutation in the tumor suppressor gene Adenomatous polyposis coli (Apc) exhibit a primary genetic defect that predisposes to a high incidence of adenomatous polyps in the small intestine rather than in the colon. Colon cell culture models expressing quantifiable markers for carcinogenic risk may represent an alternative approach to reduce, refine or replace long-term animal experimentation. The newly developed colon epithelial cell lines 1638N COL-Cl(1) (clonal derivative of the parental Apc mutant cell line 1638N COL) and 1638N COL-Pr(1) (tumor derivative of the clone), established from an Apc1638N [+/-] mutant mouse, exhibit aberrant cell cycle progression, downregulated apoptosis, enhanced carcinogenic risk and tumor formation, indicating that aberrantly proliferative preneoplastic1638N COL-Cl(1) cells exhibit a quantifiable risk for carcinogenesis. Treatment of these preneoplastic Apc mutant cells with a combination of celecoxib and 5-fluorouracil at clinically achievable low concentrations produced a 2.1 fold to 5.5 fold higher efficacy for cytostatic growth arrest and a 40.2% to 52.4% higher efficacy for inhibition of carcinogenic risk, relative to that obtained by these agents used individually. Thus, a low dose combination of mechanistically distinct agents resulted in enhanced efficacy. These data validate a novel cell culture model and a rapid mechanism-based approach to prioritize efficacious drug combinations for animal studies and clinical trials on cancer prevention and, thereby, support the 3R concept by refining and/or reducing the use of animals in biomedical research relevant to prevention/therapy of colon cancer.


Asunto(s)
Alternativas a las Pruebas en Animales , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/prevención & control , Genes APC , Lesiones Precancerosas/prevención & control , Animales , Anticarcinógenos/farmacología , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
18.
Bioorg Med Chem ; 14(18): 6349-58, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16777417

RESUMEN

Betulonic acid, derived from betulinol, a pentacyclic styrene, has shown a highly specific anti-prostate cancer activity in in vitro cell cultures. However, due to the lack of solubility of betulonic acid in aqueous medium, its potent anti-cancer activity in vivo has not been determined to the fullest extent. The present study describes the chemical synthesis of hydrophilic Boc-lysinated-betulonic acid, which has improved its solubility in an aqueous biocompatible solvent. Evaluation in cytotoxicity assays, Boc-lysinated-betulonic acid dissolved in phosphate-buffered saline (PBS) containing 22% ethanol and 4% human serum albumin, has shown 95.7% inhibition of LNCaP prostate cancer cells in culture after 72 h incubation at a concentration of 100 microM, but with little effect on normally proliferating fibroblast cells. In the in vivo assay, male athymic mice transplanted with human prostate LNCaP xenografts were injected with Boc-lysinated-betulonic acid intraperitoneally at a dose of 30 mg/kg daily for 17 days. The treated mice exhibited 92% inhibition of tumor growth as compared to controls. Histological sections of the tumors showed that Boc-lysinated-betulonic acid arrested mitosis and induced apoptosis, which was confirmed by TUNEL assay, Yo-Pro-1 staining, and the release of cleaved caspase-3 from the ex vivo in tumor culture. These studies, for the first time, demonstrate that a non-toxic hydrophilic lysinated derivative of betulonic acid and its solubility in a biocompatible aqueous medium has enhanced the bioavailability of the drug and has thus unleashed its full anti-prostate cancer activity.


Asunto(s)
Antineoplásicos/farmacología , Lisina/química , Ácido Oleanólico/análogos & derivados , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inmunohistoquímica , Técnicas In Vitro , Lisina/análogos & derivados , Masculino , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Conformación Molecular , Ácido Oleanólico/síntesis química , Ácido Oleanólico/química , Ácido Oleanólico/farmacología , Neoplasias de la Próstata/patología , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA