Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Am J Pathol ; 189(5): 942-945, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31029232

RESUMEN

This Editorial introduces this month's special Breast Ductal Carcinoma in Situ Theme Issue, a series of reviews intended to highlight the relationship of ductal carcinoma in situ as a precursor to breast cancer and emphasize the need for appropriate treatment in hopes of minimizing the progression to invasive disease.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Progresión de la Enfermedad , Femenino , Humanos , Invasividad Neoplásica
3.
J Clin Pharmacol ; 57 Suppl 10: S60-S66, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28921651

RESUMEN

Somatic mutation analysis of human cancers has become the standard of practice. Whether screening for single gene variants or sequencing hundreds of cancer-related genes, this genomic information is the basis for precision medicine initiatives in oncology. Genomic profiling results in information that allows oncologists to make a more educated selection of appropriate therapeutic strategies that more often combine traditional cytotoxic chemotherapy and radiation with novel targeted therapies. Here we discuss the nuances of implementing somatic mutation testing in a clinical setting.


Asunto(s)
Genómica/métodos , Neoplasias/genética , Medicina de Precisión/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Terapia Molecular Dirigida , Mutación , Neoplasias/patología , Neoplasias/terapia
4.
Am J Pathol ; 187(10): 2199-2207, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28822803

RESUMEN

Breast cancer represents a heterogeneous collection of diseases with disparate clinical behaviors, responses to treatment, and patient outcomes, despite common histopathological features at diagnosis. Examination of molecular signatures of breast cancer (based on complex gene expression patterns) enabled identification of several intrinsic molecular subtypes: luminal A, luminal B, human epidermal growth factor receptor 2 enriched, and basal like. The intrinsic subtypes are associated with measures of clinical aggressiveness, but do not perfectly predict patient outcomes. Several molecular signatures have been developed for prediction and prognostication of breast cancer outcomes. This review describes the molecular classification of breast cancer and the use of predictive/prognostic molecular signatures for guiding treatment decisions in breast cancer patients.


Asunto(s)
Neoplasias de la Mama/genética , Perfilación de la Expresión Génica , Neoplasias de la Mama/clasificación , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico
5.
Am J Pathol ; 187(10): 2130-2132, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28822804

RESUMEN

This Editorial highlights the reviews in the Breast Cancer Theme Issue that features topics related to next-generation breast cancer omics.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Metabolómica/métodos , Proteómica/métodos , Transcriptoma/genética , Femenino , Humanos
6.
Curr Opin Pharmacol ; 31: 104-113, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27886642

RESUMEN

Breast cancer is associated with risk factors such as advancing age and obesity. However, the linkages between these risk factors for breast cancer development and initiation of the disease are not yet clear. Obesity may drive breast cancer development through increases in circulating estrogens in postmenopausal women. Mammary cell susceptibility to neoplastic transformation requires both genetic and epigenetic alterations, including changes in DNA methylation. Obesity is also subject to epigenetic regulation. In this review, the nature of epigenetic changes, specifically changes to the methylome, are discussed in the context of obesity and breast cancer, and a potential mechanism for the interaction of obesity and breast cancer is proposed. This proposed mechanism identifies opportunities for intervention (using drugs or biologic therapies) to prevent breast cancer development in the obese patient.


Asunto(s)
Neoplasias de la Mama/etiología , Metilación de ADN , Obesidad/complicaciones , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/prevención & control , Epigénesis Genética , Estrógenos/metabolismo , Femenino , Humanos , Posmenopausia , Factores de Riesgo
7.
Am J Pathol ; 185(7): 1846-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26093982
8.
Am J Pathol ; 185(2): 282-92, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25541331

RESUMEN

Triple-negative breast cancers (including basal-like and claudin-low molecular subtypes) represent 20% to 25% of all breast cancers, but disproportionately contribute to breast cancer-associated death. We have identified a novel fundamental biological property of triple-negative breast cancers: most triple-negative breast cancers express aberrant DNA hypermethylation due to overexpression of DNA methyltransferase 3b (and hyperactivity of the DNA methyltransferase enzymes). DNA methyltransferase 3b overexpression occurs secondary to loss of miRNA-mediated post-transcriptional regulation. The resulting hyperactivity of DNA methyltransferase 3b produces concurrent DNA methylation-dependent silencing of numerous critical gene targets (including tumor suppressors and pro-apoptotic genes) and resistance to cytotoxic chemotherapy. This observation presents new opportunities for development of innovative treatment strategies on the basis of the epigenome as a novel therapeutic target in triple-negative breast cancers. Epigenetic therapy represents a new principle in cancer treatment in which restoration of critical molecular pathways occurs secondary to reexpression of silenced genes that encode negative mediators of cancer cell growth.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Metilación de ADN , ADN de Neoplasias/metabolismo , Resistencia a Antineoplásicos , Epigénesis Genética , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN de Neoplasias/genética , Femenino , Humanos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , ADN Metiltransferasa 3B
9.
Int J Oncol ; 44(2): 563-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24297604

RESUMEN

Basal-like breast cancers frequently express aberrant DNA hypermethylation associated with concurrent silencing of specific genes secondary to DNMT3b overexpression and DNMT hyperactivity. DNMT3b is known to be post-transcriptionally regulated by microRNAs. The objective of the current study was to determine the role of microRNA dysregulation in the molecular mechanism governing DNMT3b overexpression in primary breast cancers that express aberrant DNA hypermethylation. The expression of microRNAs (miRs) that regulate (miR-29a, miR-29b, miR-29c, miR-148a and miR-148b) or are predicted to regulate DNMT3b (miR­26a, miR-26b, miR-203 and miR-222) were evaluated among 70 primary breast cancers (36 luminal A-like, 13 luminal B-like, 5 HER2­enriched, 16 basal-like) and 18 normal mammoplasty tissues. Significantly reduced expression of miR-29c distinguished basal-like breast cancers from other breast cancer molecular subtypes. The expression of aberrant DNA hypermethylation was determined in a subset of 33 breast cancers (6 luminal A-like, 6 luminal B-like, 5 HER2-enriched and 16 basal-like) through examination of methylation­sensitive biomarker gene expression (CEACAM6, CDH1, CST6, ESR1, GNA11, MUC1, MYB, TFF3 and SCNN1A), 11/33 (33%) cancers exhibited aberrant DNA hypermethylation including 9/16 (56%) basal-like cancers, but only 2/17 (12%) non-basal-like cancers (luminal A-like, n=1; HER2-enriched, n=1). Breast cancers with aberrant DNA hypermethylation express diminished levels of miR-29a, miR-29b, miR-26a, miR-26b, miR-148a and miR-148b compared to cancers lacking aberrant DNA hypermethylation. A total of 7/9 (78%) basal-like breast cancers with aberrant DNA hypermethylation exhibit diminished levels of ≥6 regulatory miRs. The results show that i) reduced expression of miR-29c is characteristic of basal-like breast cancers, ii) miR and methylation-sensitive gene expression patterns identify two subsets of basal-like breast cancers, and iii) the subset of basal-like breast cancers with reduced expression of multiple regulatory miRs express aberrant DNA hypermethylation. Together, these findings strongly suggest that the molecular mechanism governing the DNMT3b-mediated aberrant DNA hypermethylation in primary breast cancer involves the loss of post-transcriptional regulation of DNMT3b by regulatory miRs.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Carcinoma Basocelular/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Carcinoma Basocelular/patología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ADN Metiltransferasa 3B
10.
Am J Pathol ; 183(4): 1113-1124, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23993780

RESUMEN

Breast cancer is noted for disparate clinical behaviors and patient outcomes, despite common histopathological features at diagnosis. Molecular pathogenesis studies suggest that breast cancer is a collection of diseases with variable molecular underpinnings that modulate therapeutic responses, disease-free intervals, and long-term survival. Traditional therapeutic strategies for individual patients are guided by the expression status of the estrogen and progesterone receptors (ER and PR) and human epidermal growth factor receptor 2 (HER2). Although such methods for clinical classification have utility in selection of targeted therapies, short-term patient responses and long-term survival remain difficult to predict. Molecular signatures of breast cancer based on complex gene expression patterns have utility in prediction of long-term patient outcomes, but are not yet used for guiding therapy. Examination of the correspondence between these methods for breast cancer classification reveals a lack of agreement affecting a significant percentage of cases. To realize true personalized breast cancer therapy, a more complete analysis and evaluation of the molecular characteristics of the disease in the individual patient is required, together with an understanding of the contributions of specific genetic and epigenetic alterations (and their combinations) to management of the patient. Here, we discuss the molecular and cellular heterogeneity of breast cancer, the impact of this heterogeneity on practical breast cancer classification, and the challenges for personalized breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Heterogeneidad Genética , Medicina de Precisión , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Femenino , Perfilación de la Expresión Génica , Salud Global , Humanos , Resultado del Tratamiento
11.
Exp Mol Pathol ; 95(3): 276-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24045095

RESUMEN

A subset of human breast cancer cell lines exhibits aberrant DNA hypermethylation that is characterized by hyperactivity of the DNA methyltransferase enzymes, overexpression of DNMT3b, and concurrent methylation-dependent silencing of numerous epigenetic biomarker genes. The objective of this study was to determine if this aberrant DNA hypermethylation (i) is found in primary breast cancers, (ii) is associated with specific breast cancer molecular subtypes, and (iii) influences patient outcomes. Analysis of epigenetic biomarker genes (CDH1, CEACAM6, CST6, ESR1, GNA11, MUC1, MYB, SCNN1A, and TFF3) identified a gene expression signature characterized by reduced expression levels or loss of expression among a cohort of primary breast cancers. The breast cancers that express this gene expression signature are enriched for triple-negative subtypes - basal-like and claudin-low breast cancers. Methylation analysis of primary breast cancers showed extensive promoter hypermethylation of epigenetic biomarker genes among triple-negative breast cancers, compared to other breast cancer subclasses where promoter hypermethylation events were less frequent. Furthermore, triple-negative breast cancers either did not express or expressed significantly reduced levels of protein corresponding to methylation-sensitive biomarker gene products. Together, these findings suggest strongly that loss of epigenetic biomarker gene expression is frequently associated with gene promoter hypermethylation events. We propose that aberrant DNA hypermethylation is a common characteristic of triple-negative breast cancers and may represent a fundamental biological property of basal-like and claudin-low breast cancers. Kaplan-Meier analysis of relapse-free survival revealed a survival disadvantage for patients with breast cancers that exhibit aberrant DNA hypermethylation. Identification of this distinguishing trait among triple-negative breast cancers forms the basis for development of new rational therapies that target the epigenome in patients with basal-like and claudin-low breast cancers.


Asunto(s)
Biomarcadores de Tumor/genética , Mama/patología , Metilación de ADN , Epigenómica , Recurrencia Local de Neoplasia/genética , Regiones Promotoras Genéticas/genética , Neoplasias de la Mama Triple Negativas/genética , Mama/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Recurrencia Local de Neoplasia/mortalidad , Pronóstico , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Tasa de Supervivencia , Neoplasias de la Mama Triple Negativas/clasificación , Neoplasias de la Mama Triple Negativas/mortalidad
13.
Exp Mol Pathol ; 93(3): 391-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23142414

RESUMEN

The natural history of breast cancer unfolds with the development of ductal carcinoma in situ (DCIS) in normal breast tissue, and evolution of this pre-invasive neoplasm into invasive cancer. The mechanisms that drive these processes are poorly understood, but evidence from the literature suggests that mammary carcinogenesis may occur through the process of field cancerization. Clinical observations are consistent with the idea that (i) DCIS may arise in a field of altered breast epithelium, (ii) narrow surgical margins do not remove the entire altered field (contributing to recurrence and/or disease progression), and (iii) whole-breast radiation therapy is effective in elimination of the residual field of altered cells adjacent to the resected DCIS. Molecular studies suggest that the field of altered breast epithelial cells may carry cancer-promoting genetic mutations (or other molecular alterations) or cancer promoting epimutations (oncogenic alterations in the epigenome). In fact, most breast cancers develop through a succession of molecular events involving both genetic mutations and epimutations. Hence, in hereditary forms of breast cancer, the altered field reflects the entire breast tissue which is composed of cells with a predisposing molecular lesion (such as a BRCA1 mutation). In the example of a BRCA1-mutant patient, it is evident that local resection of a DCIS lesion or localized but invasive cancer will not result in elimination of the altered field. In sporadic breast cancer patients, the mechanistic basis for the altered field may not be so easily recognized. Nonetheless, identification of the nature of field cancerization in a given patient may guide clinical intervention. Thus, patients with DCIS that develops in response to an epigenetic lesion (such as a hypermethylation defect affecting the expression of tumor suppressor genes) might be treated with epigenetic therapy to normalize the altered field and reduce the risk of secondary occurrence of DCIS or progression to invasive cancer.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Transformación Celular Neoplásica/genética , Lesiones Precancerosas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/terapia , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/terapia , Terapia Combinada , Progresión de la Enfermedad , Epigénesis Genética/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Mutación , Recurrencia Local de Neoplasia , Lesiones Precancerosas/genética , Lesiones Precancerosas/terapia , Ubiquitina-Proteína Ligasas/genética
14.
Int J Oncol ; 41(2): 721-32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22664488

RESUMEN

A hypermethylation defect associated with DNMT hyperactivity and DNMT3b overexpression characterizes a subset of breast cancers and breast cancer cell lines. We analyzed breast cancer cell lines for differential expression of regulatory miRs to determine if loss of miR-mediated post-transcriptional regulation of DNMT3b represents the molecular mechanism that governs the overexpression of DNMT3b that drives the hypermethylation defect in breast cancer. MicroRNAs (miRs) that regulate (miR-29a, miR-29b, miR-29c, miR-148a, miR-148b) or are predicted (miR-26a, miR-26b, miR-203, miR-222) to regulate DNMT3b were examined among 10 hypermethylator and 6 non-hypermethylator breast cancer cell lines. Hypermethylator cell lines express diminished levels of miR-29c, miR-148a, miR-148b, miR-26a, miR-26b, and miR-203 compared to non-hypermethylator cell lines. miR expression patterns correlate inversely with methylation-sensitive gene expression (r=-0.66, p=0.0056) and directly with the methylation status of these genes (r=0.72, p=0.002). To determine the mechanistic role of specific miRs in the dysregulation of DNMT3b among breast cancer cell lines, miR levels were modulated by transfection of pre-miR precursors for miR-148b, miR-26b, and miR-29c into hypermethylator cell lines (Hs578T, HCC1937, SUM185) and transfection of antagomirs directed against miR-148b, miR-26b, and miR-29c into non-hypermethylator cell lines (BT20, MDA-MB-415, MDA-MB-468). Antagomir-mediated knock-down of miR-148b, miR-29c, and miR-26b significantly increased DNMT3b mRNA in non-hypermethylator cell lines, and re-expression of miR-148b, miR-29c, and miR-26b following transfection of pre-miR precursors significantly reduced DNMT3b mRNA in hypermethylator cell lines. These findings strongly suggest that: i) post-transcriptional regulation of DNMT3b is combinatorial, ii) diminished expression of regulatory miRs contributes to DNMT3b overexpression, iii) re-expression of regulatory miRs reduces DNMT3b mRNA levels in hypermethylator breast cancer cell lines, and iv) down-regulation of regulatory miRs increases DNMT3b mRNA levels in non-hypermethylator breast cancer cell lines. In conlcusion, the molecular mechanism governing the DNMT3b-mediated hypermethylation defect in breast cancer cell lines involves the loss of post-transcriptional regulation of DNMT3b by regulatory miRs.


Asunto(s)
Neoplasias de la Mama/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/fisiología , Regiones Promotoras Genéticas , Interferencia de ARN , Transcripción Genética , ADN Metiltransferasa 3B
15.
Breast Cancer Res Treat ; 131(2): 385-99, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21359954

RESUMEN

A subset of primary breast cancers and breast cancer cell lines express a hypermethylation defect (characterized by DNMT hyperactivity and DNMT3b overexpression) which contributes to chemotherapy resistance and provides a target for development of new treatment strategies. The objective of the current study was to determine if targeting the epigenome enhances the sensitivity of breast cancer cells to cytotoxic chemotherapy. Hypermethylator breast cancer cell lines (MDA-MB-453, BT549, and Hs578T) were treated with 250 or 500 nM 5-aza-2'-deoxycytidine (5-aza) and/or were subjected to RNAi-mediated DNMT3b knockdown (KD), and then tested for sensitivity to doxorubicin hydrochloride (DOX), paclitaxel (PAX), and 5-fluorouracil (5-FU). In MDA-MB-453 cells, DNMT3b KD reduces the IC(50) for DOX from 0.086 to 0.048 µM (44% reduction), for PAX from 0.497 to 0.376 nM (24%), and for 5-FU from 0.817 to 0.145 mM (82%). Treatment with 250 nM 5-aza for 7 days did not increase the efficacy of DOX, PAX, or 5-FU, but 7-day treatment with 500 nM 5-aza sensitized cells, reducing the IC(50) for DOX to 0.035 µM (60%), PAX to 0.311 nM (37%), and 5-FU to 0.065 mM (92%). 5-aza treatment of DNMT3b KD cells reduced the IC(50) for DOX to 0.036 µM (59%), for PAX to 0.313 nM (37%) and for 5-FU to 0.067 (92%). Similar trends of enhancement of cell kill were seen in BT549 (13-60%) and Hs578T (29-70%) cells after RNAi-mediated DNMT3b KD and/or treatment with 5-aza. The effectiveness of DOX, PAX, and 5-FU is enhanced through targeted and/or pharmacological inhibition of DNMT3b, strongly suggesting that combined epigenetic and cytotoxic treatment will improve the efficacy of breast cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , ARN Interferente Pequeño/metabolismo , ADN Metiltransferasa 3B
16.
Vitam Horm ; 87: 93-109, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22127239

RESUMEN

The mammalian liver possesses an extraordinary capacity for regeneration of tissue mass and cell numbers following loss of hepatocytes due to partial tissue loss (surgical resection) or hepatotoxic injury (necrosis). Restoration of liver mass can be obtained through the outgrowth and expansion of a number of different cell types depending upon the nature and extent of injury and/or tissue deficit. In an otherwise healthy liver, the replacement of hepatocytes (and tissue mass) is achieved through the proliferation of differentiated, normally quiescent hepatocytes contained in the residual (viable) tissue. However, in certain forms of liver injury, the capacity of differentiated hepatocytes to proliferate in response to liver tissue deficit is significantly impaired. When this occurs, reserve stem-like progenitor cells are activated to proliferate and replace lost hepatocytes. In this review, we will discuss the currently available information regarding the activation and outgrowth of each of these liver progenitor cell populations.


Asunto(s)
Hígado/citología , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Hígado/fisiología , Regeneración Hepática , Células Madre/metabolismo
17.
Mol Cancer Ther ; 10(3): 427-36, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21282354

RESUMEN

Many types of cancer cells require a supply of fatty acids (FA) for growth and survival, and interrupting de novo FA synthesis in model systems causes potent anticancer effects. We hypothesized that, in addition to synthesis, cancer cells may obtain preformed, diet-derived FA by uptake from the bloodstream. This would require hydrolytic release of FA from triglyceride in circulating lipoprotein particles by the secreted enzyme lipoprotein lipase (LPL), and the expression of CD36, the channel for cellular FA uptake. We find that selected breast cancer and sarcoma cells express and secrete active LPL, and all express CD36. We further show that LPL, in the presence of triglyceride-rich lipoproteins, accelerates the growth of these cells. Providing LPL to prostate cancer cells, which express low levels of the enzyme, did not augment growth, but did prevent the cytotoxic effect of FA synthesis inhibition. Moreover, LPL knockdown inhibited HeLa cell growth. In contrast to the cell lines, immunohistochemical analysis confirmed the presence of LPL and CD36 in the majority of breast, liposarcoma, and prostate tumor tissues examined (n = 181). These findings suggest that, in addition to de novo lipogenesis, cancer cells can use LPL and CD36 to acquire FA from the circulation by lipolysis, and this can fuel their growth. Interfering with dietary fat intake, lipolysis, and/or FA uptake will be necessary to target the requirement of cancer cells for FA.


Asunto(s)
Proliferación Celular , Grasas de la Dieta/metabolismo , Ácidos Grasos/metabolismo , Lipoproteína Lipasa/metabolismo , Neoplasias/metabolismo , Animales , Antígenos CD36/genética , Línea Celular Tumoral , Inhibidores de la Síntesis de Ácidos Grasos/farmacología , Ácidos Grasos/farmacología , Femenino , Humanos , Lipólisis , Liposarcoma/genética , Liposarcoma/metabolismo , Masculino , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética
18.
Exp Mol Pathol ; 89(3): 209-16, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20840848

RESUMEN

The molecular pathogenesis of hepatocellular carcinoma is well-studied but not completely understood. We utilized a microcell-hybrid model of tumor suppression in rat liver tumor cells to facilitate the identification of liver tumor suppressor genes located on human chromosome 11. These investigations confirmed a liver tumor suppressor locus at human 11p11.2, identified Wt1 as a potential effector of 11p11.2-mediated tumor suppression, and subsequently identified human SYT13 as a strong candidate for the 11p11.2 liver tumor suppressor gene. In the studies presented here, we introduced SYT13 into the GN6TF rat liver tumor cell line to characterize a functional role for SYT13 in this model system. Transfected clones expressing an SDS-resistant dimer form of the SYT13 protein displayed induction of Wt1 gene expression and a significant attenuation of the neoplastic phenotype exhibited by the parental tumor cell line. Saturation densities and anchorage-independent growth of SYT13 dimer-positive cell lines were reduced in vitro, and tumorigenicity was significantly decreased or ablated in syngeneic host rats in vivo. In addition, restoration of the contact-inhibited, epithelioid morphology observed in normal liver epithelial cells accompanied ectopic expression of the SYT13 protein dimer, suggesting that SYT13 may be mediating an epithelial differentiation coordinate with tumor suppression in these cells. Accordingly, the expression of E-cadherin (Cdh1) mRNA was increased >100-fold in SYT13-dimer-positive cell lines and the Cdh1 transcriptional repressor Snail was decreased >3-fold in these cells compared to the parental tumor cells. These studies combine to suggest that SYT13 is a liver tumor suppressor gene and that its function may be mediated through pathways implicated in mesenchymal to epithelial transition.


Asunto(s)
Cromosomas Humanos Par 11/genética , Transición Epitelial-Mesenquimal/genética , Genes Supresores de Tumor/fisiología , Neoplasias Hepáticas/genética , Sinaptotagminas/biosíntesis , Animales , Western Blotting , Diferenciación Celular/fisiología , Línea Celular Tumoral , Humanos , Células Híbridas , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Fenotipo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Sinaptotagminas/genética , Transfección
19.
Exp Mol Pathol ; 89(2): 92-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20599936

RESUMEN

Liver regeneration after partial hepatectomy (PH) in rats exposed to the pyrrolizidine alkaloid retrorsine is accomplished through the proliferation and differentiation of a population of small hepatocyte-like progenitor cells (SHPCs). The activation, emergence, and outgrowth of SHPCs in response to the liver deficit generated through surgical PH have been well characterized. However, the participation of these cells in the restoration of hepatocyte numbers and regeneration of liver tissue mass following necrotic injury has not been investigated. To investigate the capacity of SHPCs to respond to necrotizing liver injury, we combined retrorsine treatment with the centrilobular-specific toxin carbon tetrachloride (CCl(4)). Male Fischer 344 rats were treated with retrorsine (30 mg/kg ip) at 6 and 8 weeks of age, followed by CCl(4) treatment (1500 mg/kg ip) 5 weeks later. Liver tissues were harvested at 3, 7, 14, 21, and 30-days post-injection. The dose of CCl(4) employed resulted in the necrotic destruction of 59±2% of liver mass and elicited a regenerative response equivalent to that of surgical PH. Livers from retrorsine-exposed CCl(4)-treated rats exhibit SHPC proliferation similar to retrorsine-exposed rats subjected to PH (RP). SHPCs appear at 3-days post-injection, continue to expand at 7-days and 14-days post-injection, and completely regenerate/restore the liver mass and structure in these animals by 30-days post-injection. The magnitude of SHPC response observed in the undamaged periportal zone of the liver in these animals is unaffected (versus RP rats) by the loss of the centrilobular region. The results of this study show that SHPCs are capable of regenerating liver after exposure to necrotizing agents and suggest that the progenitor cell of origin of the SHPCs is not restricted to the centrilobular zone of the liver parenchyma.


Asunto(s)
Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas , Regeneración Hepática/fisiología , Células Madre/citología , Células Madre/fisiología , Animales , Hepatectomía , Hepatocitos/citología , Regeneración Hepática/efectos de los fármacos , Masculino , Necrosis/patología , Necrosis/cirugía , Alcaloides de Pirrolicidina/farmacología , Distribución Aleatoria , Ratas , Ratas Endogámicas F344
20.
Exp Mol Pathol ; 88(1): 7-14, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19874816

RESUMEN

Complete liver regeneration after partial hepatectomy (PH) in rats exposed to the pyrrolizidine alkaloid retrorsine is accomplished through the activation, expansion, and differentiation of a population of small hepatocyte-like progenitor cells (SHPCs). The mechanism(s) governing activation of SHPCs after PH in retrorsine-injured rats has not been investigated. We examined the possibility that SHPCs require cytokine priming prior to becoming growth factor responsive in this model of liver injury and regeneration. Male Fischer 344 rats were treated with retrorsine (30 mg/kg ip) at 6 and 8 weeks of age. Retrorsine-exposed and age-matched control rats were randomized into dexamethasone-treated and no DEX groups. DEX-treated animals were either given a single dose of DEX (2 mg/kg ip) at the time of PH or multiple DEX treatments (2 mg/kg ip each) at 24 and 1 h before PH and 1, 2, and 3 days post-PH. A subset of rats received 10 microg of recombinant IL6 protein, administered intravenously 30 min after PH. Liver tissues were harvested at 7, 14, 21, and 30 days post-PH. Treatment of retrorsine-exposed rats with the cytokine inhibitor dexamethasone (DEX) effectively blocked the emergence of SHPCs resulting in an inhibition of liver regeneration and producing significant short-term mortality. The livers of DEX-treated retrorsine-exposed rats displayed decreased numbers and smaller SHPC clusters compared to retrorsine-exposed rats in the absence of DEX treatment. Administration of recombinant IL6 to DEX-treated retrorsine-exposed rats restored the emergence of SHPCs and SHPC-mediated regenerative response. The livers of DEX-treated retrorsine-exposed rats that received IL6 displayed numbers of expanding SHPC clusters comparable to that of retrorsine-exposed rats in the absence of DEX treatment. These results combine to suggest that SHPC activation after PH in retrorsine-exposed rats is cytokine dependent and may specifically require IL6.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Citocinas/fisiología , Hepatocitos/efectos de los fármacos , Regeneración Hepática/efectos de los fármacos , Alcaloides de Pirrolicidina/toxicidad , Células Madre/efectos de los fármacos , Animales , Citocinas/antagonistas & inhibidores , Dexametasona/farmacología , Modelos Animales de Enfermedad , Antagonismo de Drogas , Quimioterapia Combinada , Glucocorticoides/farmacología , Hepatectomía , Hepatocitos/metabolismo , Hepatocitos/patología , Interleucina-6/farmacología , Hígado/efectos de los fármacos , Hígado/patología , Regeneración Hepática/fisiología , Longevidad/efectos de los fármacos , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes , Células Madre/metabolismo , Células Madre/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...