Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 269
Filtrar
1.
Oncogene ; 32(35): 4120-9, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23045280

RESUMEN

Our recent study showed critical roles of Dmp1 as a sensor of oncogenic Ras, HER2/neu signaling and activation of the Arf-p53 pathway. To elucidate the role of human DMP1 (hDMP1) in breast cancer, one hundred and ten pairs of human breast cancer specimen were studied for the alterations of the hDMP1-ARF-Hdm2-p53 pathway with follow up of clinical outcomes. Loss of heterozygosity (LOH) of the hDMP1 locus was found in 42% of human breast carcinomas, while that of INK4a/ARF and p53 were found in 20 and 34%, respectively. Hdm2 amplification was found in 13% of the same sample, which was found independently of LOH for hDMP1. Conversely, LOH for hDMP1 was found in mutually exclusive fashion with that of INK4a/ARF and p53, and was associated with low Ki67 index and diploid karyotype. Consistently, LOH for hDMP1 was associated with luminal A category and longer relapse-free survival, while that of p53 was associated with non-luminal A and shorter survival. Thus, loss of hDMP1 could define a new disease category associated with prognosis of breast cancer patients. Human breast epithelial cells/cancer cells with wild-type p53 were sensitive to growth inhibition by activated Dmp1:ER while those that delete p14(ARF) or p53, and/or Hdm2 amplification showed partial or nearly complete resistance, indicating that p53 is a critical target for hDMP1 to exhibit its biological activity.


Asunto(s)
Neoplasias de la Mama/mortalidad , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Factores de Transcripción/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Femenino , Humanos , Pérdida de Heterocigocidad , Invasividad Neoplásica , Pronóstico , Transducción de Señal
2.
Oncogene ; 31(16): 2007-16, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21909131

RESUMEN

Thyroid cancers are the most common malignancy of the endocrine system in humans. To understand the molecular genetic events underlying thyroid carcinogenesis, we have generated a mouse model that spontaneously develops follicular thyroid carcinoma similar to human thyroid cancer (Thrb(PV/PV) mouse). This mutant mouse harbors a dominant-negative mutated thyroid hormone receptor ß (denoted PV). The PV mutation was identified in a patient with resistance to thyroid hormone (TH). Thrb(PV/PV) mice exhibit highly elevated serum thyroid-stimulating hormone levels and increased TH. We have previously shown that thyroid-stimulating hormone is required, but not sufficient to induce metastatic follicular thyroid cancer in Thrb(PV/PV) mice. However, whether the elevated TH also contributes to the thyroid carcinogenesis of Thrb(PV/PV) mice was not elucidated. To understand the role of TH in thyroid carcinogenesis, we blocked the production of TH by treating Thrb(PV/PV) mice with propylthiouracil (Thrb(PV/PV)-PTU mice) and compared the development of thyroid cancer in Thrb(PV/PV)-PTU and untreated Thrb(PV/PV) mice. We found that thyroid tumor growth was reduced by ∼42% in Thrb(PV/PV)-PTU mice as compared with Thrb(PV/PV) mice. Analysis by bromodeoxyuridine-nuclear labeling showed decreased incorporation of bromodeoxyuridine in thyroid tumor cells of Thrb(PV/PV)-PTU mice, indicative of decreased tumor cell proliferation. However, cleaved-caspase 3 staining showed no apparent changes in apoptosis of tumor cells in Thrb(PV/PV)-PTU mice. Molecular studies identified a marked attenuation of the PI3K-AKT-ß-catenin signaling pathway that led to decreased protein levels of cyclin D2, thereby decreasing tumor cell proliferation in Thrb(PV/PV)-PTU mice. Furthermore, matrix metalloproteinase-2, a downstream target of ß-catenin and a key regulator during tumor invasion and metastasis, was also decreased. Thus, the present study uncovers a critical role of TH in promoting the thyroid carcinogenesis of Thrb(PV/PV) mice via membrane signaling events. Importantly, these findings suggest that anti-thyroid drugs could be considered as possible therapeutic agents of thyroid cancer.


Asunto(s)
Hormonas Tiroideas/fisiología , Neoplasias de la Tiroides/metabolismo , Adenocarcinoma Folicular , Animales , Antitiroideos/farmacología , Proliferación Celular/efectos de los fármacos , Ciclina D2/metabolismo , Modelos Animales de Enfermedad , Ratones , Propiltiouracilo/farmacología , Transducción de Señal , Receptores beta de Hormona Tiroidea/genética , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/prevención & control , Tirotropina/sangre
3.
Oncogene ; 30(30): 3381-90, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21399657

RESUMEN

Correlative data suggest that thyroid hormone receptor-ß (TRß) mutations could increase the risk of mammary tumor development, but unequivocal evidence is still lacking. To explore the role of TRß mutants in vivo in breast tumor development and progression, we took advantage of a knock-in mouse model harboring a mutation in the Thrb gene encoding TRß (Thrb(PV) mouse). Although in adult nulliparous females, a single ThrbPV allele did not contribute to mammary gland abnormalities, the presence of two ThrbPV alleles led to mammary hyperplasia in ∼36% Thrb(PV/PV) mice. The ThrbPV mutation further markedly augmented the risk of mammary hyperplasia in a mouse model with high susceptibility to mammary tumors (Pten(+/-) mouse), as demonstrated by the occurrence of mammary hyperplasia in ∼60% of Thrb(PV/+)Pten(+/-) and ∼77% of Thrb(PV/PV)Pten(+/-) mice versus ∼33% of Thrb(+/+)Pten(+/-) mice. The Thrb(PV) mutation increased the activity of signal transducer and activator of transcription (STAT5) to increase cell proliferation and the expression of the STAT5 target gene encoding ß-casein in the mammary gland. We next sought to understand the molecular mechanism underlying STAT5 overactivation by TRßPV. Cell-based studies with a breast cancer cell line (T47D cells) showed that thyroid hormone (T3) repressed STAT5 signaling in TRß-expressing cells through decreasing STAT5-mediated transcription activity and target gene expression, whereas sustained STAT5 signaling was observed in TRßPV-expressing cells. Collectively, these findings show for the first time that a TRß mutation promotes the development of mammary hyperplasia via aberrant activation of STAT5, thereby conferring a fertile genetic ground for tumorigenesis.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Mutación , Receptores beta de Hormona Tiroidea/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Hiperplasia/genética , Ligandos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/genética
4.
Oncogene ; 29(13): 1909-19, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20062085

RESUMEN

Aberrant expression and mutations of thyroid hormone receptor genes (TRs) are closely associated with several types of human cancers. To test the hypothesis that TRs could function as tumor suppressors, we took advantage of mice with deletion of all functional TRs (TRalpha1(-/-)TRbeta(-/-) mice). As these mice aged, they spontaneously developed follicular thyroid carcinoma with pathological progression from hyperplasia to capsular invasion, vascular invasion, anaplasia and metastasis to the lung, similar to human thyroid cancer. Detailed molecular analysis revealed that known tumor promoters such as pituitary tumor-transforming gene were activated and tumor suppressors such as peroxisome proliferator-activated receptor gamma and p53 were suppressed during carcinogenesis. In addition, consistent with the human cancer, AKT-mTOR-p70(S6K) signaling and vascular growth factor and its receptor were activated to facilitate tumor progression. This report presents in vivo evidence that functional loss of both TRalpha1 and TRbeta genes promotes tumor development and metastasis. Thus, TRs could function as tumor suppressors in a mouse model of metastatic follicular thyroid cancer.


Asunto(s)
Adenocarcinoma Folicular/genética , Regulación Neoplásica de la Expresión Génica , Ratones Transgénicos , Receptores de Hormona Tiroidea/genética , Neoplasias de la Tiroides/genética , Adenocarcinoma Folicular/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Mutación , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/genética , Neoplasias de la Tiroides/metabolismo
5.
Mol Endocrinol ; 23(3): 308-15, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19131509

RESUMEN

Thyroid hormone receptors (TRs) play critical roles in energy homeostasis. To understand the role of TRs in lipid homeostasis in vivo, we adopted the loss-of-function approach by creating knock-in mutant mice with targeted mutation in the TRalpha gene (TRalpha1PV mouse) or TRbeta gene (TRbetaPV mouse). The PV mutation, identified in a patient with resistance to thyroid hormone, exhibits potent dominant-negative activity. Here we show that in contrast to TRalpha1PV mouse, TRbetaPV mice exhibited no significant reduction in WAT but had significant increases in serum free fatty acids and total triglycerides. Moreover, the liver of TRbetaPV mice was markedly increased (33%) with excess lipid accumulation, but the liver mass of TRalpha1PV mouse was decreased (23%) with paucity of lipids. These results indicate that apo-TRbeta and apo-TRalpha1 exerted distinct abnormalities in lipid metabolism. Further biochemical analyses indicate that increased lipogenic enzyme expression, activated peroxisome proliferator-activated receptor gamma (Ppargamma) signaling, and decreased fatty acid beta-oxidation activity contributed to the adipogenic steatosis and lipid accumulation in the liver of TRbetaPV mice. In contrast, the expression of lipogenic enzymes and Ppargamma was decreased in the liver of TRalpha1PV mice. These results suggest that the regulation of genes critical for lipid metabolism by TRs in the liver is isoform dependent. These results indicate that apo-TRbeta and apo-TRalpha1 had different effects on lipid metabolism and that both TR isoforms contribute to the pathogenesis of lipid metabolism in hypothyroidism.


Asunto(s)
Metabolismo de los Lípidos/genética , Receptores de Hormona Tiroidea/fisiología , Adipogénesis/genética , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Hígado Graso/etiología , Hígado Graso/patología , Hepatomegalia/etiología , Hepatomegalia/patología , Hipotiroidismo/complicaciones , Hipotiroidismo/genética , Hipotiroidismo/metabolismo , Hipotiroidismo/patología , Ligandos , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Oxidación-Reducción , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Receptores de Hormona Tiroidea/agonistas , Receptores de Hormona Tiroidea/genética , Hormonas Tiroideas/farmacología , Activación Transcripcional
6.
Oncogene ; 28(4): 509-17, 2009 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18997818

RESUMEN

Inactivation and silencing of PTEN have been observed in multiple cancers, including follicular thyroid carcinoma. PTEN (phosphatase and tensin homologue deleted from chromosome 10) functions as a tumour suppressor by opposing the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signalling pathway. Despite correlative data, how deregulated PTEN signalling leads to thyroid carcinogenesis is not known. Mice harbouring a dominant-negative mutant thyroid hormone receptor beta (TRbeta(PV/PV) mice) spontaneously develop follicular thyroid carcinoma and distant metastases similar to human cancer. To elucidate the role of PTEN in thyroid carcinogenesis, we generated TRbeta(PV/PV) mice haploinsufficient for Pten (TRbeta(PV/PV)Pten(+/-) mouse). PTEN deficiency accelerated the progression of thyroid tumour and increased the occurrence of metastasis spread to the lung in TRbeta(PV/PV)Pten(+/-) mice, thereby significantly reducing their survival as compared with TRbeta(PV/PV)Pten(+/+) mice. AKT activation was further increased by two-fold in TRbeta(PV/PV)Pten(+/-) mice thyroids, leading to increased activity of the downstream mammalian target of rapamycin (mTOR)-p70S6K signalling and decreased activity of the forkhead family member FOXO3a. Consistently, cyclin D1 expression was increased. Apoptosis was decreased as indicated by increased expression of nuclear factor-kappaB (NF-kappaB) and decreased caspase-3 activity in the thyroids of TRbeta(PV/PV)Pten(+/-) mice. Our results indicate that PTEN deficiency resulted in increased cell proliferation and survival in the thyroids of TRbeta(PV/PV)Pten(+/-) mice. Altogether, our study provides direct evidence to indicate that in vivo, PTEN is a critical regulator in the follicular thyroid cancer progression and invasiveness.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Neoplasias de la Tiroides/metabolismo , Animales , Apoptosis/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Proliferación Celular , Supervivencia Celular/genética , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Mutantes , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
7.
Oncogene ; 27(6): 823-30, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-17653082

RESUMEN

The molecular genetic events underlying thyroid carcinogenesis are not well understood. Mice harboring a dominant-negative mutant thyroid hormone receptor-beta (TRbeta(PV/PV) mice) spontaneously develop follicular thyroid carcinoma similar to human cancer. The present study aimed to elucidate the role of the steroid receptor coactivator-3 (SRC-3) in thyroid carcinogenesis in vivo by using the offspring from the cross of TRbeta(PV/PV) and SRC-3(-/-) mice. TRbeta(PV/PV) mice deficient in SRC-3 (TRbeta(PV/PV)SRC-3(-/-) mice) had significantly increased survival, decreased thyroid tumor growth, delayed tumor progression and lower incidence of distant metastasis as compared with TRbeta(PV/PV) mice with SRC-3 (TRbeta(PV/PV)SRC-3(+/+) mice). Further, in vivo and in vitro analyses of multiple signaling pathways indicated that SRC-3 deficiency could lead to (1) inhibition of cell cycle progression at the G(1)/S transition via controlling the expression of cell cycle regulators, such as E2F1; (2) induction of apoptosis by controlling the expression of the Bcl-2 and caspase-3 genes and (3) suppression of neovascularization and metastasis, at least in part, through modulating the vascular endothelial growth factor gene expression. Taken together, SRC-3 could play important roles through regulating multiple target genes and signaling pathways during thyroid carcinogenesis, understanding of which should direct future therapeutic options for thyroid cancer.


Asunto(s)
Adenocarcinoma Folicular/genética , Regulación Neoplásica de la Expresión Génica , Histona Acetiltransferasas/fisiología , Neoplasias de la Tiroides/genética , Transactivadores/fisiología , Adenocarcinoma Folicular/irrigación sanguínea , Adenocarcinoma Folicular/patología , Animales , Apoptosis/genética , Caspasa 3/genética , Ciclo Celular/genética , Modelos Animales de Enfermedad , Histona Acetiltransferasas/genética , Ratones , Ratones Mutantes , Neovascularización Patológica/genética , Coactivador 3 de Receptor Nuclear , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptores beta de Hormona Tiroidea/genética , Neoplasias de la Tiroides/irrigación sanguínea , Neoplasias de la Tiroides/patología , Transactivadores/genética
8.
Oncogene ; 25(59): 7703-13, 2006 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-16878159

RESUMEN

Dmp1 is a Myb-like transcription factor that transmits oncogenic Ras-Raf signaling to the Arf-p53 pathway and induces cell cycle arrest. Immunohistochemical staining was performed to identify the pattern of Dmp1 expression in normal murine tissues compared with the proliferation marker, Ki67. In thymus, the nuclei of mature T lymphocytes in the medulla were strongly positive for Dmp1, whereas Ki67 was detected only in the cortex. In intestine, Dmp1 was detected in the nuclei of superficial layers of the villi, whereas Ki67-positive cells were confined to the lower one-third of the crypt. Double staining for Dmp1 and Ki67 revealed that these two proteins were expressed in mutually exclusive fashion in nearly all the tissues examined. Subsets of E2Fs were specifically bound to the Dmp1 promoter upon mitogenic signaling and E2Fs 1-4 inhibited the Dmp1 promoter in a reporter assay. The Dmp1 promoter was repressed when the cells entered the S to G2/M phase of the cell cycle when both Dmp1 and Arf expressions were downregulated. The Dmp1 mRNA was not downregulated by serum in E2F-DB(+) cells, suggesting that the Dmp1 promoter repression is E2F-dependent. This explains why the Dmp1 and Ki67-positive cells are stained in mutually exclusive fashion in normal tissues.


Asunto(s)
Factores de Transcripción E2F/fisiología , Factores de Transcripción/análisis , Células 3T3 , Animales , Secuencia de Bases , Western Blotting , Inmunohistoquímica , Antígeno Ki-67/análisis , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Factores de Transcripción/genética
9.
Oncogene ; 25(19): 2736-47, 2006 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-16314832

RESUMEN

The molecular genetic events underlying thyroid carcinogenesis are poorly understood. Mice harboring a knock-in dominantly negative mutant thyroid hormone receptor beta (TRbetaPV/PV mouse) spontaneously develop follicular thyroid carcinoma similar to human thyroid cancer. Using this mutant mouse, we tested the hypothesis that the peroxisome proliferator-activated receptor gamma (PPARgamma) could function as a tumor suppressor in thyroid cancer in vivo. Using the offspring from the cross of TRbetaPV/+ and PPARgamma+/- mice, we found that thyroid carcinogenesis progressed significantly faster in TRbetaPV/PV mice with PPARgamma insufficiency from increased cell proliferation and reduced apoptosis. Reduced PPARgamma protein abundance led to the activation of the nuclear factor-kappaB signaling pathway, resulting in the activation of cyclin D1 and repression of critical genes involved in apoptosis. Treatment of TRbetaPV/PV mice with a PPARgamma agonist, rosiglitazone, delayed the progression of thyroid carcinogenesis by decreasing cell proliferation and activation of apoptosis. These results suggest that PPARgamma is a critical modifier in thyroid carcinogenesis and could be tested as a therapeutic target in thyroid follicular carcinoma.


Asunto(s)
Transformación Celular Neoplásica , FN-kappa B/metabolismo , PPAR gamma/fisiología , Transducción de Señal , Receptores beta de Hormona Tiroidea/fisiología , Neoplasias de la Tiroides/metabolismo , Animales , Apoptosis , Proliferación Celular , Ciclina D1/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , FN-kappa B/genética , PPAR gamma/genética , Rosiglitazona , Tiazolidinedionas/farmacología , Receptores beta de Hormona Tiroidea/genética
10.
Mol Endocrinol ; 17(5): 895-907, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12576488

RESUMEN

Mutations in the thyroid hormone receptor (TR) beta gene result in resistance to thyroid hormone (RTH), characterized by reduced sensitivity of tissues to thyroid hormone. To understand which physiological TR pathways are affected by mutant receptors, we crossed mice with a dominantly negative TRbeta mutation (TRbetaPV) with mice carrying a TRbeta null mutation (TRbeta(-/-)) to determine the consequences of the TRbetaPV mutation in the absence of wild-type TRbeta. TRbeta(PV/-) mice are distinct from TRbeta(+/-) mice that did not show abnormalities in thyroid function tests. TRbeta(PV/-) mice are also distinct from TRbeta(PV/+) and TRbeta(-/-) mice in that the latter shows mild dysfunction in the pituitary-thyroid axis, whereas the former exhibit very severe abnormalities, including extensive papillary hyperplasia of the thyroid epithelium, indistinguishable from that observed in TRbeta(PV/PV) mice. Similar to TRbeta(PV/PV) mice, TRbeta(PV/-) mice exhibited impairment in weight gain. Moreover, the abnormal regulation patterns of T3-target genes in the tissues of TRbeta(PV/-) and TRbeta(PV/PV) mice were strikingly similar. Using TR isoforms and PV-specific antibodies in gel shift assays, we found that in vivo, PV competed with TRalpha1 for binding to thyroid hormone response elements in TRbeta(PV/-) mice as effectively as in TRbeta(PV/PV) mice. Thus, the actions of mutant TRbeta are markedly potentiated by the ablation of the second TRbeta allele, suggesting that interference with wild-type TRalpha1-mediated gene regulation by mutant TRbeta leads to severe RTH.


Asunto(s)
Mutación , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Alelos , Animales , Unión Competitiva , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Genes Dominantes , Hiperplasia/genética , Ratones , Ratones Mutantes , Hipófisis/fisiopatología , Elementos de Respuesta/genética , Glándula Tiroides/fisiopatología , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea , Síndrome de Resistencia a Hormonas Tiroideas/genética , Tirotropina/metabolismo , Triyodotironina/metabolismo , Aumento de Peso/genética
11.
Leukemia ; 17(1): 155-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12529673

RESUMEN

The novel fusion protein DT(388)IL3, composed of the catalytic and translocation domains of diphtheria toxin (DT(388)) fused with a Met-His linker to human interleukin 3 (IL-3), was tested for anti-leukemia efficacy in an in vivo model of differentiated human acute myeloid leukemia (AML). Six-week-old female SCID mice were irradiated with 350 cGy, inoculated 24 h later with 20 million (i.v., i.p., or s.c.) TF1 cells transfected with the v-SRC oncogene, and treated i.p., starting 24 h later, with up to five daily injections of saline, DT(388)IL3 (2 microg), DT(388)GMCSF (2 microg), DAB(389)IL2 (2 microg), or cytarabine (80 microg) or two weekly injections of anti-CD33-calicheamicin conjugate (5 microg). Animals were monitored twice daily, and moribund animals killed and necropsied. Control animals had a median disease-free survival (DFS) of 37 days (i.v., n = 45), 35 days (i.p., n = 20), and 21 days (s.c., n = 20), respectively. Only 5/49 (10%) of the DT(388)IL3 treated i.v. inoculated animals died with leukemia. Median DFS with i.v., i.p. and s.c. tumor inoculated animals was prolonged by fusion protein treatment to >120 days, 66 days and 31 days (P < 0.001, = 0.0003, and = 0.0006), respectively. Median DFS with s.c. tumor inoculated animals was also prolonged by other active anti-leukemia agents (DT(388)GMCSF, cytarabine and anti-CD33-calicheamicin) relative to controls by 67%, 172% and 47% (P < 0.0001, <0.0001, and =0.0004), respectively. In contrast, median DFS with s.c. tumor inoculated animals treated with DAB(389)IL2 non-significantly reduced by 13% relative to controls (P = 0.21). Thus, DT(388)IL3 fusion protein demonstrates in vivo anti-leukemia efficacy and warrants further preclinical development for treatment of chemo-resistant, IL-3 receptor positive AML patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Toxina Diftérica/uso terapéutico , Interleucina-3/uso terapéutico , Leucemia Mieloide/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Enfermedad Aguda , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Citarabina/administración & dosificación , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Huésped Inmunocomprometido , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Subcutáneas , Interleucina-2/administración & dosificación , Leucemia Mieloide/mortalidad , Leucemia Mieloide/patología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 98(26): 15095-100, 2001 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-11734632

RESUMEN

Mutations in the thyroid hormone receptor beta (TRbeta) gene result in resistance to thyroid hormone. However, it is unknown whether mutations in the TRalpha gene could lead to a similar disease. To address this question, we prepared mutant mice by targeting mutant thyroid hormone receptor kindred PV (PV) mutation to the TRalpha gene locus by means of homologous recombination (TRalpha1PV mice). The PV mutation was derived from a patient with severe resistance to thyroid hormone that has a frameshift of the C-terminal 14 aa of TRbeta1. We knocked in the same PV mutation to the corresponding TRalpha gene locus to compare the phenotypes of TRalpha1(PV/+) mice with those of TRbeta(PV/+) mice. TRalpha1(PV/+) mice were viable, indicating that the mutation of the TRalpha gene is not embryonic lethal. In drastic contrast to the TRbeta(PV/+) mice, which do not exhibit a growth abnormality, TRalpha1(PV/+) mice were dwarfs. These dwarfs exhibited increased mortality and reduced fertility. In contrast to TRbeta(PV/+) mice, which have a hyperactive thyroid, TRalpha1(PV/+) mice exhibited mild thyroid failure. The in vivo pattern of abnormal regulation of T3 target genes in TRalpha1(PV/+) mice was unique from those of TRbeta(PV/+) mice. The distinct phenotypes exhibited by TRalpha1(PV/+) and TRbeta(PV/+) mice indicate that the in vivo functions of TR mutants are isoform-dependent. The TRalpha1(PV/+) mice may be used as a tool to uncover human diseases associated with mutations in the TRalpha gene and, furthermore, to understand the molecular mechanisms by which TR isoforms exert their biological activities.


Asunto(s)
Enanismo/genética , Genes Dominantes , Infertilidad/genética , Mortalidad , Mutación , Receptores de Hormona Tiroidea/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Expresión Génica , Ratones , Receptores de Hormona Tiroidea/genética , Análisis de Supervivencia , Enfermedades de la Tiroides/genética , Activación Transcripcional
13.
J Virol ; 75(24): 12169-81, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11711608

RESUMEN

The induction of apoptosis in host cells is a prominent cytopathic effect of vesicular stomatitis virus (VSV) infection. The viral matrix (M) protein is responsible for several important cytopathic effects, including the inhibition of host gene expression and the induction of cell rounding in VSV-infected cells. This raises the question of whether M protein is also involved in the induction of apoptosis. HeLa or BHK cells were transfected with M mRNA to determine whether M protein induces apoptosis when expressed in the absence of other viral components. Expression of M protein induced apoptotic morphological changes and activated caspase-3 in both cell types, indicating that M protein induces apoptosis in the absence of other viral components. An M protein containing a point mutation that renders it defective in the inhibition of host gene expression (M51R mutation) activated little, if any, caspase-3, while a deletion mutant lacking amino acids 4 to 21 that is defective in the virus assembly function but fully functional in the inhibition of host gene expression was as effective as wild-type (wt) M protein in activating caspase-3. To determine whether M protein influences the induction of apoptosis in the context of a virus infection, the M51R M protein mutation was incorporated onto a wt background by using a recombinant infectious cDNA clone (rM51R-M virus). The timing of the induction of apoptosis by rM51R-M virus was compared to that by the corresponding recombinant wt (rwt) virus and to that by tsO82 virus, the mutant virus in which the M51R mutation was originally identified. In HeLa cells, rwt virus induced apoptosis faster than did rM51R-M virus, demonstrating a role for M protein in the induction of apoptosis. In contrast to the results obtained with HeLa cells, rwt virus induced apoptosis more slowly than did rM51R-M virus in BHK cells. This indicates that a viral component other than M protein contributes to induction of apoptosis in BHK cells and that wt M protein acts to delay induction of apoptosis by the other viral component. tsO82 virus induced apoptosis more rapidly than did rM51R-M virus in both HeLa and BHK cells. These two viruses contain the same point mutation in their M proteins, suggesting that sequence differences in genes other than that for M protein affect their rates of induction of apoptosis.


Asunto(s)
Apoptosis , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas de la Matriz Viral/fisiología , Animales , Caspasa 3 , Caspasas/fisiología , Cricetinae , Células HeLa , Humanos , ARN Mensajero/análisis , Proteínas de la Matriz Viral/genética
14.
Carcinogenesis ; 22(10): 1607-14, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11576999

RESUMEN

Iron is involved in essential biochemical reactions ranging from respiration to DNA synthesis. Consequently, iron deprivation has been proposed as a strategy for inhibition of tumor cell growth. We recently described a novel iron chelator, tachypyridine [N,N',N"-tris(2-pyridylmethyl)-cis,cis-1,3,5-triaminocyclohexane], and demonstrated that it not only inhibited growth of cultured tumor cells, but was actively cytotoxic. Here we explore the mechanisms underlying tachpyridine cytotoxicity. Using several criteria, including time-lapse video microscopy, DNA staining and TUNEL assays, tachpyridine was shown to specifically induce apoptotic cell death. Further, unlike numerous cytotoxic chemotherapeutic drugs which induce apoptosis by activating p53-dependent pathways, tachpyridine-mediated cell death did not require p53 activation. Although immunoblotting revealed rapid accumulation of p53 following treatment with tachpyridine, p21(WAF1) was not induced. Further, neither cytotoxicity nor apoptosis required p53. p53 null human lung cancer H1299 cells transfected with an ecdysone-inducible p53 exhibited equivalent sensitivity to tachpyridine in the presence and absence of p53, demonstrating the lack of requirement for p53 in an isogenic cell system. Further, time-lapse video microscopy and TUNEL assays demonstrated that both p53 null and p53 wild-type cells underwent apoptotic cell death in response to tachpyridine. In addition, in 55 human cancer cell lines the mean GI(50) of tachpyridine in cells with mutant p53 was virtually identical to the GI(50) in cells with wild-type p53. These results demonstrate that tachpyridine initiates an apoptotic mode of cell death that does not require functional p53. Since over 50% of human tumors contain a functionally defective p53 that reduces sensitivity to commonly used chemotherapeutic agents, such as etoposide and cisplatin, the ability of tachpyridine to induce apoptosis independently of p53 may offer an advantage in anti-tumor therapy.


Asunto(s)
Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Ciclohexilaminas/farmacología , Quelantes del Hierro/farmacología , Piridinas/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , ADN de Neoplasias/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Etiquetado Corte-Fin in Situ , Mutación , Transfección , Células Tumorales Cultivadas/metabolismo
15.
J Gerontol A Biol Sci Med Sci ; 56(8): B364-71, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11487595

RESUMEN

In this study, we examined the effects of age and growth hormone replacement on both coronary blood flow and capillary density. Blood flow was measured by using [(14)C]-iodoantipyrine in three groups of anesthetized Brown Norway x Fischer 344 rats: young vehicle-treated animals (6 months; n = 13), old vehicle treated animals (30 months; n = 9), and old animals treated with bovine growth hormone (200 microg/kg) twice a day for 30 days (30 months; n = 7). Capillary density was measured by color segmentation analysis of sections stained for platelet endothelial cell adhesion molecule-1. In all regions examined, coronary blood flow decreased with age, and growth hormone administration resulted in an increase in flow compared to vehicle-treated animals. Capillary density decreased with age in the apex and the left ventricular middle segment. In response to growth hormone administration, capillary density increased significantly in the apex but not in other regions of the heart. Our results demonstrate that growth hormone enhances regional myocardial blood flow in the aged heart and suggest that part of this effect could be due to an increase in capillary density.


Asunto(s)
Envejecimiento/fisiología , Capilares/patología , Permeabilidad Capilar/fisiología , Circulación Coronaria/fisiología , Hormona del Crecimiento/fisiología , Análisis de Varianza , Animales , Permeabilidad Capilar/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Hormona del Crecimiento/farmacología , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Modelos Animales , Análisis Multivariante , Probabilidad , Radioinmunoensayo , Ratas , Ratas Endogámicas F344 , Valores de Referencia
16.
Am J Physiol Heart Circ Physiol ; 281(2): H915-22, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11454598

RESUMEN

We tested the hypotheses that aging is associated with a reduction in overall cardiac contractility and myofilament force generation that could be reversed with growth hormone (GH) replacement. Three groups of male Brown-Norway rats were studied: young (Y(SAL): 8 mo old, n = 13), old (O(SAL): 28 mo old, n = 13), and old GH-treated (O(GH): 28 mo old, n = 12; 300 microg bovine GH, twice a day for 30 days). The left ventricular (LV) pressure-volume relation was derived in isolated hearts, after which isolated trabecular muscles from these hearts were permeabilized and maximal myofilament force generation (Fmax) was measured. LV developed pressures at a LV volume of 0.3 ml were significantly depressed with age: 84 +/- 6 vs. 71 +/- 6 mmHg (Y(SAL) vs. O(SAL), respectively, P = 0.001) and not restored by GH (69 +/- 4 mmHg). Fmax was reduced in the aged hearts: 47.5 +/- 3.12 vs. 35.9 +/- 3.03 mN/mm2 (Y(SAL) vs. O(SAL), respectively, P = 0.014) but was restored with GH replacement to 46.7 +/- 3.12 mN/mm2 (O(SAL) vs. O(GH), P = 0.021). Our results suggest that cellular myofilament contractility is reduced with aging and restored with GH replacement.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Hormona del Crecimiento/farmacología , Corazón/fisiología , Miocardio/patología , Animales , Masculino , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Ratas , Ratas Endogámicas BN
17.
J Histochem Cytochem ; 49(7): 821-32, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11410607

RESUMEN

In an earlier article from this laboratory, the current methods developed to detect apoptosis in cells and tissues were highlighted, along with the challenges in their interpretation. Recent discoveries concerning the underlying biochemical mechanisms of apoptotic effector pathways have made possible further assays that allow a more direct measure of the activation of the apoptotic machinery in cells. This article summarizes some of these newer methods and extends the interpretation of the more classical assays of apoptosis in a defined cell system. We present data in KB and PC3 cell model culture systems induced to undergo apoptosis by the plant toxin ricin. Using a modified in situ nick translation assay (ISNT) with either Bodipy or BUdR labeling, we confirm that most cells showing altered nuclear morphology do not show reactivity with this assay until very late in the apoptotic process. We also show that only a minority of cells label with fluorescent annexin V during apoptosis but that apoptotic cells continue to internalize material from the cell surface through endocytosis after becoming reactive with annexin V. In addition, we describe the utility of a prototype of new assays for caspase substrate cleavage products, the detection of cleaved cytokeratin 18. It is these newer cleavage product assays that perhaps hold the greatest promise for specific detection of apoptosis in cells either in cell culture or in intact tissues. (J Histochem Cytochem 49:821-832, 2001)


Asunto(s)
Apoptosis , Anexina A5 , Compuestos de Boro , Bromodesoxiuridina , Caspasas/metabolismo , Colorantes Fluorescentes , Humanos , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ , Queratinas/metabolismo , Ricina/farmacología , Células Tumorales Cultivadas
18.
Leukemia ; 15(5): 814-8, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11368443

RESUMEN

We have developed an in vivo model of differentiated human acute myeloid leukemia (AML) by retroviral infection of the cytokine-dependent AML cell line TF-1 with the v-Src oncogene. When injected either intravenously or intraperitoneally into 300 cGy irradiated SCID mice, animals formed multiple granulocytic sarcomas involving the adrenals, kidneys, lymph nodes and other organs. The mean survival time was 34+/-10 days (n = 40) after intravenous injection and 24+/-3 days (n = 5) after intraperitoneal injection of 20 million cells. The cells recovered from leukemic animals continued to express interleukin-3 receptors and remained sensitive to the diphtheria fusion protein DT388IL3. Further, these granulocytic sarcoma-derived cells grew again in irradiated SCID mice (n = 10). The cytogenetic abnormalities observed prior to inoculation in mice were stably present after in vivo passage. Similar to the results with v-Src transfected TF-1 cells, in vivo leukemic growth was observed with TF-1 cells transfected with the human granulocyte-macrophage colony-stimulating factor gene (n = 5) and with TF-1 cells recovered from subcutaneous tumors in nude mice (n = 5). In contrast, TF-1 cells expressing v-Ha-Ras (n = 5), BCR-ABL (n = 5), or activated Raf-1 (n = 44) did not grow in irradiated SCID mice. This is a unique, reproducible model for in vivo growth of a differentiated human acute myeloid leukemia and may be useful in the assessment of anti-leukemic therapeutics which have human-specific molecular targets such as the interleukin-3 receptor.


Asunto(s)
Genes src/fisiología , Leucemia Mieloide Aguda/patología , Animales , Aberraciones Cromosómicas , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Receptores de Interleucina-3/análisis , Células Tumorales Cultivadas
19.
J Lipid Res ; 41(12): 1991-2001, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11108732

RESUMEN

Two closely related enzymes with more than 50% sequence identity have been identified that catalyze the esterification of cholesterol using acyl-CoA substrates, namely acyl-CoA:cholesterol acyltransferase 1 (ACAT1) and ACAT2. Both are membrane-spanning proteins believed to reside in the endoplasmic reticulum of cells. ACAT2 has been hypothesized to be associated with lipoprotein particle secretion whereas ACAT1 is ubiquitous and may serve a more general role in cellular cholesterol homeostasis. We have prepared and affinity purified rabbit polyclonal antibodies unique to either ACAT enzyme to identify their cellular localization in liver and intestine, the two main lipoprotein-secreting tissues of the body, and for comparison, kidney and adrenal. In the liver, ACAT2 was identified in the rough endoplasmic reticulum of essentially all hepatocytes whereas ACAT1 was confined to cells lining the intercellular spaces among hepatocytes in a pattern typical of Kupffer cells. In the intestine, ACAT2 signal was strongly present in the apical third of the mucosal cells, whereas ACAT1 staining was diffuse throughout the mucosal cell, but with strong signal in goblet cells, Paneth cells, and villus macrophages. In the kidney, ACAT1 immunostaining was specific for the distal tubules and podocytes within the glomerulus. In the adrenal, ACAT1 signal was strongly present in the cells of the cortex, and absent from other adrenal cell types. No ACAT2 signal was identified in the kidney or adrenal. We conclude that only the cells of the liver and intestine that secrete apolipoprotein B-containing lipoproteins contain ACAT2, whereas ACAT1 is present in numerous other cell types. The data clearly suggest separate functions for these two closely related enzymes, with ACAT2 being most closely associated with plasma cholesterol levels.


Asunto(s)
Glándulas Suprarrenales/enzimología , Intestinos/enzimología , Isoenzimas/metabolismo , Riñón/enzimología , Hígado/enzimología , Esterol O-Aciltransferasa/metabolismo , Animales , Células CHO , Chlorocebus aethiops , Cricetinae , Inmunohistoquímica , Masculino
20.
Proc Natl Acad Sci U S A ; 97(24): 13209-14, 2000 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-11069286

RESUMEN

Patients with mutations in the thyroid hormone receptor beta (TRbeta) gene manifest resistance to thyroid hormone (RTH), resulting in a constellation of variable phenotypic abnormalities. To understand the molecular basis underlying the action of mutant TRbeta in vivo, we generated mice with a targeted mutation in the TRbeta gene (TRbetaPV; PV, mutant thyroid hormone receptor kindred PV) by using homologous recombination and the Cre/loxP system. Mice expressing a single PV allele showed the typical abnormalities of thyroid function found in heterozygous humans with RTH. Homozygous PV mice exhibit severe dysfunction of the pituitary-thyroid axis, impaired weight gains, and abnormal bone development. This phenotype is distinct from that seen in mice with a null mutation in the TRbeta gene. Importantly, we identified abnormal expression patterns of several genes in tissues of TRbetaPV mice, demonstrating the interference of the mutant TR with the gene regulatory functions of the wild-type TR in vivo. These results show that the actions of mutant and wild-type TRbeta in vivo are distinct. This model allows further study of the molecular action of mutant TR in vivo, which could lead to better treatment for RTH patients.


Asunto(s)
Desarrollo Óseo/genética , Crecimiento/genética , Hipófisis/fisiología , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/fisiología , Glándula Tiroides/fisiología , Alelos , Animales , Regulación del Desarrollo de la Expresión Génica , Trastornos del Crecimiento/genética , Homocigoto , Ratones , Ratones Transgénicos , Hipófisis/crecimiento & desarrollo , Recombinación Genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timidina Quinasa/genética , Glándula Tiroides/crecimiento & desarrollo , Aumento de Peso , beta-Galactosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA