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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Transplant ; 29(12): 1105-11, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26358816

RESUMEN

BACKGROUND: Patients with recurrent hepatitis C (HCV) infection post-liver transplant can be difficult to treat safely and effectively. A prior (COSMOS) study in patients with non-transplant HCV, using sofosbuvir plus simeprevir, had high efficacy and tolerability in treating patients with HCV genotype 1, even prior non-responders to interferon therapy and those with cirrhosis. Our aim was to evaluate the efficacy of sofosbuvir and simeprevir in patients with genotype 1 HCV post-liver transplant. METHODS: In this prospective, observational study, patients received sofosbuvir 400 mg plus simeprevir 150 mg daily for 12 wk without ribavirin. The primary end point was a sustained virologic response 12 wk after the end of therapy. RESULTS: Forty-two patients completed the treatment. Twenty-six percent started the treatment ≤ 6 months post-liver transplant. Nineteen percent of the included patients had cirrhosis, 14% with decompensation. At week 4 on the treatment, 21% of patients had detectable virus but at the end of the treatment, 100% were undetectable. Twelve weeks after the end of the treatment, 95% of the patients had undetectable hepatitis C. The regimen was generally well tolerated. CONCLUSION: The oral regimen of sofosbuvir plus simeprevir without ribavirin is efficacious and well tolerated in the treatment of patients with genotype 1 hepatitis C post-liver transplant.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/genética , Hepatitis C/prevención & control , Trasplante de Hígado/efectos adversos , Simeprevir/uso terapéutico , Sofosbuvir/uso terapéutico , Aloinjertos , ADN Viral/genética , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Genotipo , Hepacivirus/patogenicidad , Hepatitis C/virología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Recurrencia
2.
Ann Surg Oncol ; 19(9): 2853-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22434246

RESUMEN

BACKGROUND: Colorectal cancer (CRC) oncogenesis was considered to be determined by interactions between genetic and environmental factors. Specific interacting factors that influence CRC morbidity have yet to be fully investigated. METHODS: A multi-institutional collaborative study with 1511 CRC patients and 2098 control subjects was used to compare the odds ratios for the occurrence of polymorphisms at 11 known single nucleotide polymorphisms (SNPs). TaqMan PCR and questionnaires were used to evaluate the effects of environmental exposures. RESULTS: Variants of rs6983267 on 8q24 were the most significant markers of risk for CRC (odds ratio 1.16, 95% confidence interval 1.06-1.27, P = 0.0015). Non-insulin-dependent diabetes mellitus (DM), a higher body mass index at age 20, and meat consumption were environmental risk factors, whereas a tuna-rich diet and vitamin intake were protective factors. The cohort of rs6983267 SNP major (T) allele at 8q24 and DM had a 1.66-fold higher risk ratio than the cohort of major allele patients without DM. CONCLUSIONS: We confirmed that interactions between the genetic background and environmental factors are associated with increased risk for CRC. There is a robust risk of the minor G allele at the 8q24 rs6983267 SNP; however, a major T allele SNP could more clearly reveal a correlation with CRC specifically when DM is present.


Asunto(s)
Índice de Masa Corporal , Cromosomas Humanos Par 8 , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/genética , Complicaciones de la Diabetes/epidemiología , Factores de Edad , Alelos , Animales , Estudios de Casos y Controles , Neoplasias Colorrectales/patología , Intervalos de Confianza , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Modelos Logísticos , Masculino , Carne/efectos adversos , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Encuestas y Cuestionarios , Atún , Vitaminas
3.
J Gastroenterol Hepatol ; 23 Suppl 2: S205-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19120899

RESUMEN

BACKGROUND: The fragile histidine triad (FHIT) functions as a tumor suppressor, and giving adenoviral-FHIT (Ad-FHIT) is thus expected to be clinically beneficial. Much attention has recently been focused on which genes are commonly regulated by Ad-FHIT, and which genes are dominant in Ad-FHIT-induced apoptotic cells. METHODS: Ad-FHIT apoptosis-induced cells (H1299 and TE4) and non-apoptosis-induced cells (TE2) were used in the current experiments. The total RNA extracted from Ad-FHIT or control was labeled with Cy3-dCTP or Cy5-dCTP and hybridized with 19,192 genes on a chip. A microarray analysis for each gene was carried out with high reproducibility provided by seven independent experiments and duplicated oligos on a chip. RESULTS: We listed the upregulated genes based on the TE4:TE2 expression ratio, such as c-Src, Jak-1, and sialyltransferase, which are expected to be target pathways as well as the downregulated genes, including CASP8 and CASP10, after Ad-FHIT treatment in esophageal cancer. CONCLUSIONS: The current microarray analysis indicated that the apoptosis of esophageal cancer observed after giving Ad-FHIT was possibly induced by activation of the c-Src gene and inactivation of the CASP8 gene.


Asunto(s)
Ácido Anhídrido Hidrolasas/biosíntesis , Adenoviridae/genética , Apoptosis/genética , Neoplasias Esofágicas/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Proteínas de Neoplasias/biosíntesis , Ácido Anhídrido Hidrolasas/genética , Caspasa 8/genética , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Perfilación de la Expresión Génica/métodos , Silenciador del Gen , Genes src , Humanos , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados , Activación Transcripcional , Transducción Genética
4.
Cancer Res ; 66(5): 2683-90, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510588

RESUMEN

The FHIT gene is known to be susceptible to environmental carcinogens. Formation of prostaglandin E(2) (PGE(2)) is catalyzed by cyclooxygenase-2 (COX-2) and may influence malignant phenotype in colorectal cancer. We explored whether FHIT might play a role in progression of colorectal cancer through inflammation-associated PGE(2) activity. Immunohistochemical study of COX-2 and FHIT expression was done in 92 colorectal cancer tumors. We also used a FHIT-expressing cancer cell line (H460) induced by ponasterone A and two FHIT small interfering RNA-treated colorectal cancer cell lines (CCK81 and DLD1). After PGE(2) stimulation, we compared synthesis of PGE(2) (ELISA assay) and cell proliferation [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay]. Immunohistochemistry showed a significant association between COX-2 and FHIT expression in colorectal cancers (P < 0.01). In a subset of 41 COX-2-expressing tumors, 12 FHIT(-) tumors showed deeper cancer invasion than 29 FHIT(+) tumors (P < 0.01). Experimental study, however, showed there was no direct interaction between FHIT and COX-2. Considered with results from another experiment with epidermal growth factor receptor (EGFR), we hypothesize that FHIT and COX-2 might be regulated by a common molecule, such as EGFR. Additionally, there was an inverse and direct correlation between PGE(2) synthesis and FHIT in vitro, suggesting that FHIT's postulated antiaggressive effect on tumor goes through PGE(2) but not COX-2. Loss of FHIT expression in colorectal cancer suggests higher malignant potential. We conclude that FHIT suppressed cancer cell proliferation in this malignancy by directly inhibiting synthesis of PGE(2) but not affecting that of COX-2.


Asunto(s)
Ácido Anhídrido Hidrolasas/biosíntesis , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Dinoprostona/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Ácido Anhídrido Hidrolasas/genética , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Ciclooxigenasa 2/biosíntesis , Dinoprostona/biosíntesis , Dinoprostona/farmacología , Progresión de la Enfermedad , Receptores ErbB/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Transfección , Regulación hacia Arriba
5.
World J Gastroenterol ; 13(3): 383-90, 2007 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-17230606

RESUMEN

AIM: To study a more accurate quantification of hepatic fibrosis which would provide clinically useful information for monitoring the progression of chronic liver disease. METHODS: Using a cDNA microarray containing over 22000 clones, we analyzed the gene-expression profiles of non-cancerous liver in 74 patients who underwent hepatic resection. We calculated the ratio of azan-stained: total area, and determined the morphologic fibrosis index (MFI), as a mean of 9 section-images. We used the MFI as a reference standard to evaluate our method for assessing liver fibrosis. RESULTS: We identified 39 genes that collectively showed a good correlation (r > 0.50) between gene-expression and the severity of liver fibrosis. Many of the identified genes were involved in immune responses and cell signaling. To quantify the extent of liver fibrosis, we developed a new genetic fibrosis index (GFI) based on gene-expression profiling of 4 clones using a linear support vector regression analysis. This technique, based on a supervised learning analysis, correctly quantified the various degrees of fibrosis in both 74 training samples (r = 0.76, 2.2% vs 2.8%, P < 0.0001) and 12 independent additional test samples (r = 0.75, 9.8% vs 8.6%, P < 0.005). It was far better in assessing liver fibrosis than blood markers such as prothrombin time (r = -0.53), type IV collagen 7s (r = 0.48), hyaluronic acid (r = 0.41), and aspartate aminotransferase to platelets ratio index (APRI) (r = 0.38). CONCLUSION: Our cDNA microarray-based strategy may help clinicians to precisely and objectively monitor the severity of liver fibrosis.


Asunto(s)
Hepatopatías/genética , Hepatopatías/patología , Biomarcadores , Progresión de la Enfermedad , Fibrosis/genética , Fibrosis/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Hígado/patología , Pruebas de Función Hepática , Índice de Severidad de la Enfermedad
6.
Clin Cancer Res ; 11(19 Pt 1): 6800-6, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16203767

RESUMEN

PURPOSE: Human kallikrein genes (KLK) have been reported to be involved in human malignancies and several KLKs are promising biomarkers of prostate, ovarian, testicular, and breast cancers. Herein, we investigated the clinicopathologic and biological significance of KLK6 gene expression in human gastric cancer. PATIENTS AND METHODS: Using real-time reverse transcription-PCR, we analyzed the KLK6 expression status with respect to various clinicopathologic variables in 66 patients with gastric cancer. In addition, we established a KLK6 stably suppressed gastric cancer cell line (MKN28) using small interfering RNA-mediated gene silencing, and investigated its effects on the cell proliferation rate, cell cycle, and invasiveness. RESULTS: The KLK6 gene expression in cancerous tissue (0.37 +/- 0.53) was significantly (P < 0.000001) higher than that in noncancerous tissue (0.026 +/- 0.060). Elevated KLK6 expression was significantly associated with lymphatic invasion (P = 0.03). Furthermore, patients with a high KLK6 expression had a significantly poorer survival rate than those with a low KLK6 expression (P = 0.03). Therefore, we showed that KLK6 gene silencing with KLK6 small interfering RNA effectively suppressed the cell proliferation rate (P = 0.002), cell population in the S phase (P < 0.01), and invasiveness (P < 0.01) in comparison to mock-transfected cells. CONCLUSIONS: The KLK6 gene is markedly overexpressed in gastric cancer tissue and its expression status may be a powerful prognostic indicator for patients with gastric cancer. Our findings also suggest that KLK6 may possibly be a novel target for gastric cancer therapy by gene-silencing procedures.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Calicreínas/biosíntesis , Neoplasias Gástricas/metabolismo , Biomarcadores de Tumor , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cartilla de ADN/química , Femenino , Silenciador del Gen , Humanos , Inmunohistoquímica , Metástasis Linfática , Masculino , Invasividad Neoplásica , Pronóstico , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/terapia , Factores de Tiempo , Transfección , Resultado del Tratamiento
7.
Clin Cancer Res ; 11(4): 1368-71, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15746034

RESUMEN

PURPOSE: Somatic mutations of the epidermal growth factor receptor (EGFR) gene may predict the sensitivity of non-small cell lung carcinoma to gefitinib. However, no mutations have been reported for colorectal carcinoma. We therefore analyzed EGFR mutations in colorectal adenocarcinomas by the combined use of laser microdissection and sequencing of genomic DNA. EXPERIMENTAL DESIGN: We examined 11 representative colorectal adenocarcinoma cell lines and 33 clinical samples of colorectal carcinoma. In the clinical cases, we carefully dissected only carcinoma cells from frozen sections by laser microdissection. After DNA extraction and PCR, we examined EGFR mutations by sequencing genomic DNA. RESULTS: None of 11 colorectal carcinoma cell lines exhibited somatic mutations, but 4 of 33 clinical tumors (12%) exhibited mutations in the EGFR kinase domain. This may be the first report of somatic mutations in colorectal adenocarcinoma. CONCLUSIONS: Our findings suggest that a distinct minority of colorectal adenocarcinomas exhibit somatic mutations of EGFR, and these tumors may be susceptible to gefitinib treatment.


Asunto(s)
Neoplasias Colorrectales/genética , Receptores ErbB/genética , Mutación , Secuencia de Bases , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Análisis Mutacional de ADN , ADN de Neoplasias/química , ADN de Neoplasias/genética , Exones/genética , Células HT29 , Humanos , Mutación Missense
8.
Clin Exp Metastasis ; 22(1): 59-67, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16132579

RESUMEN

Comparing differential gene expression profiles established by cDNA microarray between normal cells (N), primary carcinoma cells (T), and metastatic carcinoma cells (M) may determine those critical genes directly associated with progression and metastasis of breast cancer. Total RNA was extracted by laser microdissection (LMD) from 20 slices of T, N and M from 6 cases. After amplification by a T7-based system, differentially expressed genes between T, N and M were identified by cDNA microarray. In addition, to clarify the mechanism for altered gene expression, we determined the methylation status by sequencing after bisulfite treatment for intriguing genes. As a result, the expression of motility related protein-1 (MRP-1/CD9), peripheral myelin protein-22 (PMP-22), and caspase 3 (CASP-3) were down-regulated in M compared to T. We focused especially on MRP-1 and found that the expression status of MRP-1 was significantly inversely associated with stage of disease in 56 cases of breast cancer (P<0.05), and the relapse free survival in 5 years was significantly higher in MRP-1 positive cases than those negative cases (P<0.05). Conversely, overexpression, by 11-fold, of signal transduction and translation factors were observed in T compared to N. The cancer specific methylation was observed only in CASP-3 in a case. In conclusion, the establishment of the present assay allows us to detect genes directly associated with each cell population within tumor tissue and gives us clues to identify metastasis-related genes comprehensively in clinical breast cancer cases.


Asunto(s)
Antígenos CD/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma/genética , Carcinoma/patología , Glicoproteínas de Membrana/genética , Caspasa 3 , Caspasas/genética , Metilación de ADN , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Microdisección , Proteínas de la Mielina/genética , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Tetraspanina 29 , Activación Transcripcional
9.
Oncol Rep ; 14(3): 707-12, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16077979

RESUMEN

Polymorphic catechol-O-methyltransferase (COMT) catalyzes the O-methylation of catechol estrogens. It has been reported that COMT polymorphism is a representative genetic trait related to the susceptibility of an individual to breast cancer. However, there is no consensus concerning the association between breast cancer in Japanese patients and COMT polymorphism. To analyze the polymorphism distribution in Japanese patients with breast cancer, a molecular genotyping method using a polymerase chain reaction-based restriction fragment length polymorphism (PCR-RFLP) was used. Based on an analysis of 201 Japanese patients with breast cancer and 352 healthy control subjects, a significant difference was observed in either the distribution of genotypes (p=0.03) or allele frequencies between the two groups (p=0.01). The relative risk of breast cancer for genotypes (COMT(Met/Met) and COMT(Val/Met)) including the variant allele (COMT(Met)) was 1.47 compared to the wild allele (COMT(Val)) and homozygote (COMT(Val/Val)). Furthermore, the distribution of genotypes in post-menopausal patients with breast cancer showed a significant difference with that of healthy subjects of the same menopausal status (p=0.01). No significant difference was found between the distribution of genotypes and clinicopathological features of the cancer. These results suggest that COMT polymorphism may thus be implicated as a genetic trait affecting the susceptibility of an individual to breast cancer in a Japanese population and be an important genetic risk factor in the development of breast cancer in post-menopausal women.


Asunto(s)
Neoplasias de la Mama/genética , Catecol O-Metiltransferasa/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo Genético , Adulto , Anciano , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , ADN de Neoplasias/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Japón , Persona de Mediana Edad , Oportunidad Relativa , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Posmenopausia/genética , Premenopausia/genética
10.
Clin Cancer Res ; 10(24): 8243-9, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15623600

RESUMEN

PURPOSE: Matrix metalloproteinase-7 (MMP-7) plays an important role in carcinoma invasion and metastasis of cancer. Recent studies focus on diverse roles of MMP-7, other than as a protease, during cancer progression. MMP-7 activates the epidermal growth factor (EGF) receptor by releasing an EGF ligand, tumor growth factor (TGF)-alpha. EXPERIMENTAL DESIGN: We examined expression of MMP-7 and EGF receptor in an immunohistochemical study of 40 colorectal cancer (CRC) cases. To determine the relationship between the EGF receptor and MMP-7, with a potential curative application, we compared the antitumor activity of the EGF receptor tyrosine kinase inhibitor (gefitinib) between MMP-7 transfectant, KYSE150 and HT29, and control cells. RESULTS: We found a statistically significant correlation (P = 0.04) between MMP-7 and activated (phosphorylated) EGF receptor expression, both being positive in six (15%) cases. Gefitinib reduced the cell number ratio more for MMP-7 transfectant than mock cells, and the proportion of apoptotic cells was 1.5 times higher in MMP-7 transfectant than mock cells by annexin/propidium iodide staining. This was mediated by activation of a TGF-beta signal as confirmed by the abundant expression of TGF-beta protein, the cytoplasmic to nuclear translocation of Smad4 protein by the administration of gefitinib, and the quantitative assay of the plasminogen activator inhibitor-1 promoter/luciferase construction. CONCLUSIONS: We propose that there are some cancers with up-regulated MMP-7 expression that leads to the activation of apoptotic activity of TGF-beta, which is susceptible to treatment with EGF receptor tyrosine kinase inhibitor.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Receptores ErbB/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Núcleo Celular/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/secundario , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Femenino , Gefitinib , Humanos , Técnicas para Inmunoenzimas , Masculino , Metaloproteinasa 7 de la Matriz/genética , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Transporte de Proteínas , Quinazolinas/farmacología , Proteína Smad4 , Transactivadores/metabolismo , Transfección , Células Tumorales Cultivadas
11.
Cancer Res ; 73(7): 2059-69, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23378342

RESUMEN

Circulating tumor cells (CTC) in blood have attracted attention both as potential seeds for metastasis and as biomarkers. However, most CTC detection systems might miss epithelial-mesenchymal transition (EMT)-induced metastatic cells because detection is based on epithelial markers. First, to discover novel markers capable of detecting CTCs in which EMT has not been repressed, microarray analysis of 132 colorectal cancers (CRC) from Japanese patients was conducted, and 2,969 genes were detected that were overexpressed relative to normal colon mucosa. From the detected genes, we selected those that were overexpressed CRC with distant metastasis. Then, we analyzed the CRC metastasis-specific genes (n = 22) to determine whether they were expressed in normal circulation. As a result, PLS3 was discovered as a CTC marker that was expressed in metastatic CRC cells but not in normal circulation. Using fluorescent immunocytochemistry, we validated that PLS3 was expressed in EMT-induced CTC in peripheral blood from patients with CRC with distant metastasis. PLS3-expressing cells were detected in the peripheral blood of approximately one-third of an independent set of 711 Japanese patients with CRC. Multivariate analysis showed that PLS3-positive CTC was independently associated with prognosis in the training set (n = 381) and the validation set [n = 330; HR = 2.17; 95% confidence interval (CI) = 1.38-3.40 and HR = 3.92; 95% CI = 2.27-6.85]. The association between PLS3-positive CTC and prognosis was particularly strong in patients with Dukes B (HR = 4.07; 95% CI = 1.50-11.57) and Dukes C (HR = 2.57; 95% CI = 1.42-4.63). PLS3 is a novel marker for metastatic CRC cells, and it possesses significant prognostic value.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , Neoplasias Hepáticas/secundario , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Recurrencia Local de Neoplasia/patología , Células Neoplásicas Circulantes/patología , Anciano , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Femenino , Humanos , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Metástasis Linfática , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Microfilamentos/genética , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/mortalidad , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Clin Invest ; 120(9): 3326-39, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20697159

RESUMEN

Cancer stem cells (CSCs) are generally dormant or slowly cycling tumor cells that have the ability to reconstitute tumors. They are thought to be involved in tumor resistance to chemo/radiation therapy and tumor relapse and progression. However, neither their existence nor their identity within many cancers has been well defined. Here, we have demonstrated that CD13 is a marker for semiquiescent CSCs in human liver cancer cell lines and clinical samples and that targeting these cells might provide a way to treat this disease. CD13+ cells predominated in the G0 phase of the cell cycle and typically formed cellular clusters in cancer foci. Following treatment, these cells survived and were enriched along the fibrous capsule where liver cancers usually relapse. Mechanistically, CD13 reduced ROS-induced DNA damage after genotoxic chemo/radiation stress and protected cells from apoptosis. In mouse xenograft models, combination of a CD13 inhibitor and the genotoxic chemotherapeutic fluorouracil (5-FU) drastically reduced tumor volume compared with either agent alone. 5-FU inhibited CD90+ proliferating CSCs, some of which produce CD13+ semiquiescent CSCs, while CD13 inhibition suppressed the self-renewing and tumor-initiating ability of dormant CSCs. Therefore, combining a CD13 inhibitor with a ROS-inducing chemo/radiation therapy may improve the treatment of liver cancer.


Asunto(s)
Biomarcadores de Tumor/análisis , Antígenos CD13/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Antígenos CD13/antagonistas & inhibidores , Ciclo Celular/efectos de los fármacos , Línea Celular , Fluorouracilo/administración & dosificación , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Ratones , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
13.
J Surg Oncol ; 93(1): 44-6, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16353180

RESUMEN

BACKGROUND AND OBJECTIVES: Recent studies have disclosed the presence of somatic mutations in the epidermal growth factor receptor (EGFR) gene in non-small cell lung cancers (NSCLC), and susceptibility to the EGFR tyrosine kinase inhibitor (gefitinib) was determined by the presence of mutations in the kinase domain of this gene. We thus predicted a clinical benefit of gefitinib against the 12% of primary colorectal cancers exhibiting a mutation reflective of this potential distinctive susceptibility. PATIENTS AND METHODS: The mutation status of the kinase domain in EGFR in different primary cancers has important clinical consequences, because the presence of a mutation is recognized as a reliable indicator for the effectiveness of gefitinib administration. In the current study, we investigated the presence of somatic mutations in exons 18-21 coding the ATP-binding domain in five gastric cancer cell lines and 39 primary gastric cancers and their corresponding normal tissues. RESULTS AND CONCLUSIONS: The kinase domain of EGFR is highly conserved in whole gastric cancer cell lines and cases, therefore treatment with gefitinib is unfortunately not recommended for such malignancy.


Asunto(s)
Genes erbB-1/genética , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Neoplasias Gástricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Femenino , Gefitinib , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/patología
14.
Stem Cells ; 24(3): 506-13, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16239320

RESUMEN

A subset of stem cells, termed "side population" (SP) cells, has been identified and characterized in several mammalian tissues and cell lines. However, SP cells have never been identified or isolated from gastrointestinal cancers. We used flow cytometry and the DNA-binding dye Hoechst 33342 to isolate SP cells from various human gastrointestinal system cancer cell lines. Fifteen of sixteen cancer cell lines from the gastrointestinal system contained 0.3%-2.2% SP cells. Next, we used an oligonucleotide microarray to analyze differentially expressed genes between SP and non-SP cells of hepatoma HuH7. The expression of GATA6, which is associated with embryonic development and hepatocytic differentiation, was significantly upregulated in HuH7 SP cells. The expression of ABCG2, ABCB1, and CEACAM6, which are associated with chemoresistance, was also significantly increased in SP cells. In addition, some epithelial markers and mesenchymal markers were overexpressed in SP cells. Reverse transcription-polymerase chain reaction and immunocytochemical staining validated these results and suggested a multilineage potential for HuH7 SP cells. In hepatoma HuH7 and colorectal SW480 cell lines, SP cells showed evidence for self-renewal, generating both SP and non-SP cells. Finally, chemoresistance to anticancer agents, including doxorubicin, 5-fluorouracil, and gemcitabine, were compared between HuH7 SP and non-SP cells using an ATP bioluminescence assay. The HuH7 SP cells expressed a higher resistance to doxorubicin, 5-fluorouracil, and gemcitabine compared with non-SP cells. These findings demonstrate that cancers of the gastrointestinal system do contain SP cells that show some characteristics of so-called stem cells.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Biomarcadores de Tumor/biosíntesis , Neoplasias Gastrointestinales/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/metabolismo , Antígenos de Diferenciación/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Desarrollo Embrionario/genética , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células Madre Neoplásicas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
15.
Dig Dis Sci ; 50(7): 1304-11, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16047477

RESUMEN

We investigated whether celecoxib augments the protective effect of polyethylene glycol (PEG) on colonic aberrant crypt foci (ACF) and tumor formation in F344 rats treated with azoxymethane (AOM). Three groups of rats received AOM: I (AOM alone), II (PEG), and III (PEG/celecoxib). PEG reduced the mean number of total ACF per colon from 190 to 141 (P < 0.05; 26% reduction) and > or = 4-crypt ACF from 95 to 58 (P < 0.01; 39%). Group III rats had a greater proportion of their ACF distally; whereas transverse colon ACF were reduced approximately 50%, distal ACF were reduced by only approximately 8% (P < 0.05). Of 13 large bowel tumors, 8 were in Group I, 4 in Group II, and 1 in Group III rats (P = 0.02). Thus in AOM-treated rats celecoxib appeared to enhance the PEG-induced reduction in colonic tumor formation, and in transverse but not distal or whole-colon ACF.


Asunto(s)
Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/prevención & control , Inhibidores de la Ciclooxigenasa/farmacología , Polietilenglicoles/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Azoximetano , Bromodesoxiuridina , Celecoxib , Neoplasias del Colon/inducido químicamente , Citoprotección , Sinergismo Farmacológico , Femenino , Inmunohistoquímica/métodos , Masculino , Ratas , Ratas Endogámicas F344 , Coloración y Etiquetado
16.
Jpn J Cancer Res ; 93(7): 789-97, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12149145

RESUMEN

The CXCL12 / CXCR4 system may be important in carcinoma. Expression of the alpha-chemokine SDF-1alpha (stromal cell derived factor-1alpha) / CXCL12 mRNA is reduced in many carcinomas, yet its tissue protein expression may guide metastasis. Here we first compare the mRNA and protein expression of CXCL12 and its receptor CXCR4 in human liver, hepatocellular carcinoma, and malignant cell lines, and then assess cell cycle variation in CXCR4 expression. CXCR4 mRNA was present in most normal human tissues and malignant cell lines; it was only marginally reduced in hepatomas, while CXCL12 was markedly reduced, P < 0.0001. Immuno-histochemical staining of adjacent non-malignant liver showed regional CXCR4 cytoplasmic and cell-surface staining, limited to those hepatocytes around the central vein, a distribution resembling that of CXCL12. CXCL12 protein was not present in hepatocellular carcinoma cells in vivo, nor was cytoplasmic CXCR4 staining; nuclear CXCR4 protein expression in some malignant hepatocytes and CXCR4 staining of capillary endothelial cells around tumor cells were noted. In some malignant cell lines that had no CXCL12 on northern blots CXCL12 was weakly detectable by RT-PCR or protein staining in the cytoplasm of a few cells. With a view to future manipulation of CXCL12 / CXCR4 expression and growth we noted that in HT-29 cells CXCR4 protein expression was less on confluent than on non-confluent cells and varied during the cell cycle. Higher expression was associated most closely with the percentage of cells in the S-phase and inversely with the percentage of cells in the G1-phase. Treatment of HT-29 cells with butyrate reduced CXCR4 cell surface expression and reduced the percentage of cells in S-phase. In summary, CXCL12 protein expression parallels its mRNA, being markedly reduced in malignant cell lines and hepatomas; in liver, the regional distributions of CXCL12 and cytoplasmic CXCR4 are similar; finally, in HT-29, CXCR4 expression correlates with the S-phase of the cell cycle and is reduced during butyrate-induced differentiation.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Quimiocinas CXC/biosíntesis , Neoplasias Hepáticas/metabolismo , Receptores CXCR4/biosíntesis , Northern Blotting , Butiratos/farmacología , Ciclo Celular , Diferenciación Celular , Quimiocina CXCL12 , Endotelio Vascular/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica , Hígado/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fase S , Factores de Tiempo , Células Tumorales Cultivadas
17.
J Hepatol ; 41(3): 399-406, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15336442

RESUMEN

BACKGROUNDS/AIMS: A more accurate and objective quantification of hepatic fibrosis would provide clinically useful information for the monitoring of chronic liver disease progression and therapy recommendation. METHODS: Using a cDNA microarray of 14,814 clones, we analyzed the gene-expression profiles of fibrotic livers in a rat model. RESULTS: We identified 750 up- and 345 down-regulated genes by combining a signal-to-noise score and a random permutation test (P<0.01). The functions of these genes provided insight into the underlying molecular mechanisms of both structural remodeling and functional deficits in cirrhosis. To quantify the extent of liver fibrosis, we have generated for the first time a 'genetic fibrosis index' based on gene-expression profiling of 95 genes by combining a Pearson correlation coefficient and a 'leave-one-out' cross-validation procedure. This technique based on a supervised learning analysis correctly quantified the various degrees of fibrosis in both 20 training samples (R(2)=0.829, P<0.001) and 6 test samples (R(2)=0.822, P<0.05). CONCLUSIONS: Our method will assist researchers in identifying rational targets for intervention and might help clinicians to objectively monitor the severity of liver fibrosis.


Asunto(s)
Perfilación de la Expresión Génica , Cirrosis Hepática Experimental/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , Regulación hacia Abajo , Cirrosis Hepática Experimental/complicaciones , Neoplasias Hepáticas Experimentales/etiología , Neoplasias Hepáticas Experimentales/genética , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA