Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cell Physiol ; 235(7-8): 5679-5688, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31975377

RESUMEN

Deletion of TGFß inducible early gene-1 (TIEG) in mice results in an osteopenic phenotype that exists only in female animals. Molecular analyses on female TIEG knockout (KO) mouse bones identified increased expression of sclerostin, an effect that was confirmed at the protein level in serum. Sclerostin antibody (Scl-Ab) therapy has been shown to elicit bone beneficial effects in multiple animal model systems and human clinical trials. For these reasons, we hypothesized that Scl-Ab therapy would reverse the low bone mass phenotype of female TIEG KO mice. In this study, wildtype (WT) and TIEG KO female mice were randomized to either vehicle control (Veh, n = 12/group) or Scl-Ab therapy (10 mg/kg, 1×/wk, s.c.; n = 12/group) and treated for 6 weeks. Following treatment, bone imaging analyses revealed that Scl-Ab therapy significantly increased cancellous and cortical bone in the femur of both WT and TIEG KO mice. Similar effects also occurred in the vertebra of both WT and TIEG KO animals. Additionally, histomorphometric analyses revealed that Scl-Ab therapy resulted in increased osteoblast perimeter/bone perimeter in both WT and TIEG KO animals, with a concomitant increase in P1NP, a serum marker of bone formation. In contrast, osteoclast perimeter/bone perimeter and CTX-1 serum levels were unaffected by Scl-Ab therapy, irrespective of mouse genotype. Overall, our findings demonstrate that Scl-Ab therapy elicits potent bone-forming effects in both WT and TIEG KO mice and effectively increases bone mass in female TIEG KO mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Enfermedades Óseas Metabólicas/genética , Proteínas de Unión al ADN/genética , Osteogénesis/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/sangre , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Anticuerpos/farmacología , Densidad Ósea/genética , Desarrollo Óseo/genética , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/inmunología , Enfermedades Óseas Metabólicas/patología , Femenino , Fémur/crecimiento & desarrollo , Fémur/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Fenotipo
2.
Nucleic Acids Res ; 45(9): 5170-5182, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28201653

RESUMEN

We have previously demonstrated that TGFß Inducible Early Gene-1 (TIEG1), also known as KLF10, plays important roles in mediating skeletal development and homeostasis in mice. TIEG1 has also been identified in clinical studies as one of a handful of genes whose altered expression levels or allelic variations are associated with decreased bone mass and osteoporosis in humans. Here, we provide evidence for the first time that TIEG1 is involved in regulating the canonical Wnt signaling pathway in bone through multiple mechanisms of action. Decreased Wnt signaling in the absence of TIEG1 expression is shown to be in part due to impaired ß-catenin nuclear localization resulting from alterations in the activity of AKT and GSK-3ß. We also provide evidence that TIEG1 interacts with, and serves as a transcriptional co-activator for, Lef1 and ß-catenin. Changes in Wnt signaling in the setting of altered TIEG1 expression and/or activity may in part explain the observed osteopenic phenotype of TIEG1 KO mice as well as the known links between TIEG1 expression levels/allelic variations and patients with osteoporosis.


Asunto(s)
Huesos/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Huesos/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ligandos , Cloruro de Litio/farmacología , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Cráneo/citología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
3.
J Cell Physiol ; 233(4): 3540-3551, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29044507

RESUMEN

TIEG knockout (KO) mice exhibit a female-specific osteopenic phenotype and altered expression of TIEG in humans is associated with osteoporosis. Gene expression profiling studies identified sclerostin as one of the most highly up-regulated transcripts in the long bones of TIEG KO mice relative to WT littermates suggesting that TIEG may regulate SOST expression. TIEG was shown to substantially suppress SOST promoter activity and the regulatory elements through which TIEG functions were identified using promoter deletion and chromatin immunoprecipitation assays. Knockdown of TIEG in IDG-SW3 osteocyte cells using shRNA and CRISPR-Cas9 technology resulted in increased SOST expression and delayed mineralization, mimicking the results obtained from TIEG KO mouse bones. Given that TIEG is an estrogen regulated gene, and as changes in the hormonal milieu affect SOST expression, we performed ovariectomy (OVX) and estrogen replacement therapy (ERT) studies in WT and TIEG KO mice followed by miRNA and mRNA sequencing of cortical and trabecular compartments of femurs. SOST expression levels were considerably higher in cortical bone compared to trabecular bone. In cortical bone, SOST expression was increased following OVX only in WT mice and was suppressed following ERT in both genotypes. In contrast, SOST expression in trabecular bone was decreased following OVX and significantly increased following ERT. Interestingly, a number of miRNAs that are predicted to target sclerostin exhibited inverse expression levels in response to OVX and ERT. These data implicate important roles for TIEG and estrogen-regulated miRNAs in modulating SOST expression in bone.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Estrógenos/farmacología , Glicoproteínas/metabolismo , Osteocitos/efectos de los fármacos , Esqueleto/metabolismo , Factores de Transcripción/deficiencia , Proteínas Adaptadoras Transductoras de Señales , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Femenino , Marcadores Genéticos/fisiología , Péptidos y Proteínas de Señalización Intercelular , Ratones Noqueados , Osteocitos/metabolismo , Ovariectomía/métodos , Esqueleto/efectos de los fármacos
4.
Biochem Biophys Res Commun ; 470(3): 528-533, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26801561

RESUMEN

Deletion of TIEG1/KLF10 in mice results in an osteopenic skeletal phenotype with significant decreases in both bone mineral density and content throughout the skeleton. Calvarial osteoblasts isolated from TIEG1 knockout (KO) mice display numerous changes in gene expression and exhibit significant delays in their mineralization rates relative to wild-type (WT) controls. Here, we demonstrate that loss of TIEG1 expression in osteoblasts results in decreased levels of Osterix mRNA. Suppression of TIEG1 expression in WT osteoblasts leads to decreased Osterix expression while restoration of TIEG1 expression in TIEG1 KO osteoblasts results in increased levels of Osterix. Transient transfection and chromatin immunoprecipitation assays reveal that TIEG1 directly binds to and activates the Osterix promoter and demonstrate that the zinc finger-containing DNA binding domain of TIEG1 is necessary for this regulation. Furthermore, we reveal that TIEG1 expression is essential for the induction of Osterix expression by important bone-related cytokines such as TGFß and BMP2 in osteoblast cells. Taken together, these data implicate an important role for TIEG1 in regulating the expression of Osterix, a master regulator of osteoblast differentiation and bone formation, and suggest that decreased expression of Osterix, as well as impaired TGFß and BMP2 signaling, contribute to the observed osteopenic bone phenotype of TIEG1 KO mice.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Proteínas de Unión al ADN/metabolismo , Osteoblastos/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Enfermedades Óseas Metabólicas/patología , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Osteoblastos/patología , Transducción de Señal , Factor de Transcripción Sp7
5.
BMC Cancer ; 14: 749, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25288324

RESUMEN

BACKGROUND: The role and clinical value of ERß1 expression is controversial and recent data demonstrates that many ERß antibodies are insensitive and/or non-specific. Therefore, we sought to comprehensively characterize ERß1 expression across all sub-types of breast cancer using a validated antibody and determine the roles of this receptor in mediating response to multiple forms of endocrine therapy both in the presence and absence of ERα expression. METHODS: Nuclear and cytoplasmic expression patterns of ERß1 were analyzed in three patient cohorts, including a retrospective analysis of a prospective adjuvant tamoxifen study and a triple negative breast cancer cohort. To investigate the utility of therapeutically targeting ERß1, we generated multiple ERß1 expressing cell model systems and determined their proliferative responses following anti-estrogenic or ERß-specific agonist exposure. RESULTS: Nuclear ERß1 was shown to be expressed across all major sub-types of breast cancer, including 25% of triple negative breast cancers and 33% of ER-positive tumors, and was associated with significantly improved outcomes in ERα-positive tamoxifen-treated patients. In agreement with these observations, ERß1 expression sensitized ERα-positive breast cancer cells to the anti-cancer effects of selective estrogen receptor modulators (SERMs). However, in the absence of ERα expression, ERß-specific agonists potently inhibited cell proliferation rates while anti-estrogenic therapies were ineffective. CONCLUSIONS: Using a validated antibody, we have confirmed that nuclear ERß1 expression is commonly present in breast cancer and is prognostic in tamoxifen-treated patients. Using multiple breast cancer cell lines, ERß appears to be a novel therapeutic target. However, the efficacy of SERMs and ERß-specific agonists differ as a function of ERα expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antagonistas de Estrógenos/farmacología , Receptor beta de Estrógeno/metabolismo , Tamoxifeno/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Femenino , Humanos , Células MCF-7 , Persona de Mediana Edad
6.
Mol Ther Nucleic Acids ; 33: 28-41, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37359348

RESUMEN

Estrogen signaling is critical for the development and maintenance of healthy bone, and age-related decline in estrogen levels contributes to the development of post-menopausal osteoporosis. Most bones consist of a dense cortical shell and an internal mesh-like network of trabecular bone that respond differently to internal and external cues such as hormonal signaling. To date, no study has assessed the transcriptomic differences that occur specifically in cortical and trabecular bone compartments in response to hormonal changes. To investigate this, we employed a mouse model of post-menopausal osteoporosis (ovariectomy, OVX) and estrogen replacement therapy (ERT). mRNA and miR sequencing revealed distinct transcriptomic profiles between cortical and trabecular bone in the setting of OVX and ERT. Seven miRs were identified as likely contributors to the observed estrogen-mediated mRNA expression changes. Of these, four miRs were prioritized for further study and decreased predicted target gene expression in bone cells, enhanced the expression of osteoblast differentiation markers, and altered the mineralization capacity of primary osteoblasts. As such, candidate miRs and miR mimics may have therapeutic relevance for bone loss resulting from estrogen depletion without the unwanted side effects of hormone replacement therapy and therefore represent novel therapeutic approaches to combat diseases of bone loss.

7.
Exp Gerontol ; 142: 111113, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33065227

RESUMEN

Menopause is a natural physiological process in older women that is associated with reduced estrogen production and results in increased risk for obesity, diabetes, and osteoporosis. 17α-estradiol (17α-E2) treatment in males, but not females, reverses several metabolic conditions associated with advancing age, highlighting sexually dimorphic actions on age-related pathologies. In this study we sought to determine if 17α-E2 could prevent ovariectomy (OVX)-mediated detriments on adiposity and bone parameters in females. Eight-week-old female C57BL/6J mice were subjected to SHAM or OVX surgery and received dietary 17α-E2 during a six-week intervention period. We observed that 17α-E2 prevented OVX-induced increases in body weight and adiposity. Similarly, uterine weight and luminal cell thickness were decreased by OVX and prevented by 17α-E2 treatment. Interestingly, 17α-E2 prevented OVX-induced declines in tibial metaphysis cancellous bone. And similarly, 17α-E2 improved bone density parameters in both tibia and femur cancellous bone, primarily in OVX mice. In contrast, to the effects on cancellous bone, cortical bone parameters were largely unaffected by OVX or 17α-E2. In the non-weight bearing lumbar vertebrae, OVX reduced trabecular thickness but not spacing, while 17α-E2 increased trabecular thickness and reduced spacing. Despite this, 17α-E2 did improve bone volume/tissue volume in lumbar vertebrae. Overall, we found that 17α-E2 prevented OVX-induced increases in adiposity and changes in bone mass and architecture, with minimal effects in SHAM-operated mice. We also observed that 17α-E2 rescued uterine tissue mass and lining morphology to control levels without inducing hypertrophy, suggesting that 17α-E2 could be considered as an adjunct to traditional hormone replacement therapies.


Asunto(s)
Densidad Ósea , Estradiol , Anciano , Animales , Estradiol/farmacología , Estrógenos , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Obesidad , Tamaño de los Órganos , Ovariectomía , Ratas , Ratas Sprague-Dawley
8.
Sci Rep ; 8(1): 9445, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29930344

RESUMEN

Cervical cancer (CC) is associated with alterations in immune system balance, which is primarily due to a shift from Th1 to Th2 and the unbalance of Th17/Treg cells. Using in silico DNA copy number analysis, we have demonstrated that ~20% of CC samples exhibit gain of 8q22.3 and 19q13.31; the regions of the genome that encodes the KLF10 and PSG genes, respectively. Gene expression studies demonstrated that there were no alterations in KLF10 mRNA expression, whilst the PSG2 and -5 genes were up-regulated by 1.76 and 3.97-fold respectively in CC compared to normal tissue controls. siRNA and ChIP experiments in SiHa cells have demonstrated that KLF10 participates in immune response through regulation of IL6, IL25 and PSG2 and PSG5 genes. Using cervical tissues from KLF10-/- mice, we have identified down-regulation of PSG17, -21 and -23 and IL11. These results suggest that KLF10 may regulate immune system response genes in cervical cancer among other functions. KLF10 and PSG copy number variations and alterations in mRNA expression levels could represent novel molecular markers in CC.


Asunto(s)
Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Glicoproteínas beta 1 Específicas del Embarazo/genética , Neoplasias del Cuello Uterino/genética , Animales , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Femenino , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Glicoproteínas beta 1 Específicas del Embarazo/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Neoplasias del Cuello Uterino/inmunología
9.
Cancer Res ; 77(9): 2387-2400, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28249899

RESUMEN

TGFß-SMAD signaling exerts a contextual effect that suppresses malignant growth early in epithelial tumorigenesis but promotes metastasis at later stages. Longstanding challenges in resolving this functional dichotomy may uncover new strategies to treat advanced carcinomas. The Krüppel-like transcription factor, KLF10, is a pivotal effector of TGFß/SMAD signaling that mediates antiproliferative effects of TGFß. In this study, we show how KLF10 opposes the prometastatic effects of TGFß by limiting its ability to induce epithelial-to-mesenchymal transition (EMT). KLF10 depletion accentuated induction of EMT as assessed by multiple metrics. KLF10 occupied GC-rich sequences in the promoter region of the EMT-promoting transcription factor SLUG/SNAI2, repressing its transcription by recruiting HDAC1 and licensing the removal of activating histone acetylation marks. In clinical specimens of lung adenocarcinoma, low KLF10 expression associated with decreased patient survival, consistent with a pivotal role for KLF10 in distinguishing the antiproliferative versus prometastatic functions of TGFß. Our results establish that KLF10 functions to suppress TGFß-induced EMT, establishing a molecular basis for the dichotomy of TGFß function during tumor progression. Cancer Res; 77(9); 2387-400. ©2017 AACR.


Asunto(s)
Adenocarcinoma/genética , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Transición Epitelial-Mesenquimal/genética , Retroalimentación Fisiológica , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Pulmonares/genética , Factor de Crecimiento Transformador beta/genética , Células A549 , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Humanos , Neoplasias Pulmonares/patología , Ratones Noqueados , Pacientes , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética
10.
Endocrinology ; 158(10): 3354-3368, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977607

RESUMEN

Endoxifen, the primary active metabolite of tamoxifen, is currently being investigated as a novel endocrine therapy for the treatment of breast cancer. Tamoxifen is a selective estrogen receptor modulator that elicits potent anti-breast cancer effects. However, long-term use of tamoxifen also induces bone loss in premenopausal women and is associated with an increased risk of endometrial cancer in postmenopausal women. For these reasons, we have used a rat model system to comprehensively characterize the impact of endoxifen on the skeleton and uterus. Our results demonstrate that endoxifen elicits beneficial effects on bone in ovary-intact rats and protects against bone loss following ovariectomy. Endoxifen is also shown to reduce bone turnover in both ovary-intact and ovariectomized rats at the cellular and biochemical levels. With regard to the uterus, endoxifen decreased uterine weight but maintained luminal epithelial cell height in ovariectomized animals. Within luminal epithelial cells, endoxifen resulted in differential effects on the expression levels of estrogen receptors α and ß as well as multiple other genes previously implicated in regulating epithelial cell proliferation and hypertrophy. These studies analyze the impact of extended endoxifen exposure on both bone and uterus using a Food and Drug Administration-recommended animal model. Although endoxifen is a more potent breast cancer agent than tamoxifen, the results of the present study demonstrate that endoxifen does not induce bone loss in ovary-intact rats and that it elicits partial agonistic effects on the uterus and skeleton in ovariectomized animals.


Asunto(s)
Remodelación Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/análogos & derivados , Útero/efectos de los fármacos , Animales , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/patología , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Femenino , Tamaño de los Órganos , Osteoporosis/inducido químicamente , Ovariectomía , Ratas , Moduladores Selectivos de los Receptores de Estrógeno/efectos adversos , Tamoxifeno/efectos adversos , Tamoxifeno/farmacología , Útero/metabolismo , Útero/patología
11.
J Bone Miner Res ; 29(5): 1206-16, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24190163

RESUMEN

TGFß Inducible Early Gene-1 (TIEG1) knockout (KO) mice display a sex-specific osteopenic phenotype characterized by low bone mineral density, bone mineral content, and overall loss of bone strength in female mice. We, therefore, speculated that loss of TIEG1 expression would impair the actions of estrogen on bone in female mice. To test this hypothesis, we employed an ovariectomy (OVX) and estrogen replacement model system to comprehensively analyze the role of TIEG1 in mediating estrogen signaling in bone at the tissue, cell, and biochemical level. Dual-energy X-ray absorptiometry (DXA), peripheral quantitative computed tomography (pQCT), and micro-CT analyses revealed that loss of TIEG1 expression diminished the effects of estrogen throughout the skeleton and within multiple bone compartments. Estrogen exposure also led to reductions in bone formation rates and mineralizing perimeter in wild-type mice with little to no effects on these parameters in TIEG1 KO mice. Osteoclast perimeter per bone perimeter and resorptive activity as determined by serum levels of CTX-1 were differentially regulated after estrogen treatment in TIEG1 KO mice compared with wild-type littermates. No significant differences were detected in serum levels of P1NP between wild-type and TIEG1 KO mice. Taken together, these data implicate an important role for TIEG1 in mediating estrogen signaling throughout the mouse skeleton and suggest that defects in this pathway are likely to contribute to the sex-specific osteopenic phenotype observed in female TIEG1 KO mice.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Huesos/metabolismo , Proteínas de Unión al ADN/biosíntesis , Estrógenos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Transducción de Señal , Factores de Transcripción/biosíntesis , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Huesos/patología , Proteínas de Unión al ADN/genética , Estrógenos/farmacología , Femenino , Regulación de la Expresión Génica/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
12.
PLoS One ; 9(5): e98219, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24853369

RESUMEN

Endoxifen has recently been identified as the predominant active metabolite of tamoxifen and is currently being developed as a novel hormonal therapy for the treatment of endocrine sensitive breast cancer. Based on past studies in breast cancer cells and model systems, endoxifen classically functions as an anti-estrogenic compound. Since estrogen and estrogen receptors play critical roles in mediating bone homeostasis, and endoxifen is currently being implemented as a novel breast cancer therapy, we sought to comprehensively characterize the in vivo effects of endoxifen on the mouse skeleton. Two month old ovariectomized C57BL/6 mice were treated with vehicle or 50 mg/kg/day endoxifen hydrochloride via oral gavage for 45 days. Animals were analyzed by dual-energy x-ray absorptiometry, peripheral quantitative computed tomography, micro-computed tomography and histomorphometry. Serum from control and endoxifen treated mice was evaluated for bone resorption and bone formation markers. Gene expression changes were monitored in osteoblasts, osteoclasts and the cortical shells of long bones from endoxifen treated mice and in a human fetal osteoblast cell line. Endoxifen treatment led to significantly higher bone mineral density and bone mineral content throughout the skeleton relative to control animals. Endoxifen treatment also resulted in increased numbers of osteoblasts and osteoclasts per tissue area, which was corroborated by increased serum levels of bone formation and resorption markers. Finally, endoxifen induced the expression of osteoblast, osteoclast and osteocyte marker genes. These studies are the first to examine the in vivo and in vitro impacts of endoxifen on bone and our results demonstrate that endoxifen increases cancellous as well as cortical bone mass in ovariectomized mice, effects that may have implications for postmenopausal breast cancer patients.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Huesos/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Tamoxifeno/análogos & derivados , Animales , Antineoplásicos Hormonales/farmacología , Secuencia de Bases , Cartilla de ADN , Femenino , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Reacción en Cadena de la Polimerasa , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico
13.
PLoS One ; 8(1): e54613, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382923

RESUMEN

Endoxifen, a cytochrome P450 mediated tamoxifen metabolite, is being developed as a drug for the treatment of estrogen receptor (ER) positive breast cancer. Endoxifen is known to be a potent anti-estrogen and its mechanisms of action are still being elucidated. Here, we demonstrate that endoxifen-mediated recruitment of ERα to known target genes differs from that of 4-hydroxy-tamoxifen (4HT) and ICI-182,780 (ICI). Global gene expression profiling of MCF7 cells revealed substantial differences in the transcriptome following treatment with 4HT, endoxifen and ICI, both in the presence and absence of estrogen. Alterations in endoxifen concentrations also dramatically altered the gene expression profiles of MCF7 cells, even in the presence of clinically relevant concentrations of tamoxifen and its metabolites, 4HT and N-desmethyl-tamoxifen (NDT). Pathway analysis of differentially regulated genes revealed substantial differences related to endoxifen concentrations including significant induction of cell cycle arrest and markers of apoptosis following treatment with high, but not low, concentrations of endoxifen. Taken together, these data demonstrate that endoxifen's mechanism of action is different from that of 4HT and ICI and provide mechanistic insight into the potential importance of endoxifen in the suppression of breast cancer growth and progression.


Asunto(s)
Antagonistas de Estrógenos/metabolismo , Tamoxifeno/análogos & derivados , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Análisis por Conglomerados , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Unión Proteica , Reproducibilidad de los Resultados , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Tamoxifeno/metabolismo , Tamoxifeno/farmacología
14.
Nat Cell Biol ; 10(7): 825-36, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18516091

RESUMEN

Expression of p16(Ink4a) and p19(Arf) increases with age in both rodent and human tissues. However, whether these tumour suppressors are effectors of ageing remains unclear, mainly because knockout mice lacking p16(Ink4a) or p19(Arf) die early of tumours. Here, we show that skeletal muscle and fat, two tissues that develop early ageing-associated phenotypes in response to BubR1 insufficiency, have high levels of p16(Ink4a) and p19(Arf). Inactivation of p16(Ink4a) in BubR1-insufficient mice attenuates both cellular senescence and premature ageing in these tissues. Conversely, p19(Arf) inactivation exacerbates senescence and ageing in BubR1 mutant mice. Thus, we identify BubR1 insufficiency as a trigger for activation of the Cdkn2a locus in certain mouse tissues, and demonstrate that p16(Ink4a) is an effector and p19(Arf) an attenuator of senescence and ageing in these tissues.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Tejido Adiposo/anatomía & histología , Tejido Adiposo/metabolismo , Animales , Biomarcadores/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Ojo/anatomía & histología , Ojo/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/genética , Tasa de Supervivencia , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA