Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
JCI Insight ; 8(5)2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36692956

RESUMEN

Bone metastases are a common complication of breast cancer. We have demonstrated that intermittent administration of parathyroid hormone (PTH[1-34]) reduces the incidence of bone metastases in murine models of breast cancer by acting on osteoblasts to alter the bone microenvironment. Here, we examined the role of signaling mediated by PTH 1 receptor (PTH1R) in both osteoblasts and breast cancer cells in influencing bone metastases. In mice with impaired PTH1R signaling in osteoblasts, intermittent PTH did not reduce bone metastasis. Intermittent PTH also did not reduce bone metastasis when expression of PTH1R was knocked down in 4T1 murine breast cancer cells by shRNA. In 4T1 breast cancer cells, PTH decreased expression of PTH-related protein (PTHrP), implicated in the vicious cycle of bone metastases. Knockdown of PTHrP in 4T1 cells significantly reduced migration toward MC3T3-E1 osteoblasts, and migration was further inhibited by treatment with intermittent PTH. Conversely, overexpression of PTHrP in 4T1 cells increased migration toward MC3T3-E1 osteoblasts, and this was not inhibited by PTH. In conclusion, PTH1R expression is crucial in both osteoblasts and breast cancer cells for PTH to reduce bone metastases, and in breast cancer cells, this may be mediated in part by suppression of PTHrP.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Animales , Ratones , Hormona Paratiroidea , Proteína Relacionada con la Hormona Paratiroidea/genética , Microambiente Tumoral , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Melanoma Cutáneo Maligno
2.
JBMR Plus ; 6(12): e10695, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36530190

RESUMEN

Teriparatide, recombinant parathyroid hormone (PTH[1-34]), and abaloparatide, an analogue of PTH related-peptide (PTHrP[1-34]), are both anabolic medications for osteoporosis that target the PTH receptor PTH1R. PTH1R is a G protein-coupled receptor, and the stimulatory Gs protein is an important mediator of the anabolic actions of PTH1R activation in bone. We have published that mice lacking the α subunit of Gs in osteoprogenitors do not increase bone mass in response to PTH(1-34). Unexpectedly, however, PTH(1-34) still increases osteoblast numbers and bone formation rate in male mice, suggesting that PTH1R may have both Gs-dependent and -independent actions in bone. Here we examine the role of Gs signaling in the anabolic actions of abaloparatide. We find that abaloparatide increases bone formation in male mice with postnatal deletion of Gsα in Osx-expressing osteoprogenitors (P-GsαOsxKO mice) but not in female P-GsαOsxKO mice. Therefore, abaloparatide has anabolic effects on bone in male but not female mice that appear to be independent of Gs-mediated signaling. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

3.
Bone Rep ; 14: 100734, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33665234

RESUMEN

Diaphyseal long bone cortical tissue from 30 patients with lethal perinatal Sillence II and progressively deforming Sillence III osteogenesis imperfecta (OI) has been studied at multiple levels of structural resolution. Interpretation in the context of woven to lamellar bone formation by mesenchymal osteoblasts (MOBLs) and surface osteoblasts (SOBLs) respectively demonstrates lamellar on woven bone synthesis as an obligate self-assembly mechanism and bone synthesis following the normal developmental pattern but showing variable delay in maturation caused by structurally abnormal or insufficient amounts of collagen matrix. The more severe the variant of OI is, the greater the persistence of woven bone and the more immature the structural pattern; the pattern shifts to a structurally stronger lamellar arrangement once a threshold accumulation for an adequate scaffold of woven bone has been reached. Woven bone alone characterizes lethal perinatal variants; variable amounts of woven and lamellar bone occur in progressively deforming variants; and lamellar bone increasingly forms rudimentary and then partially compacted osteons not reaching full compaction. At differing levels of microscopic resolution: lamellar bone is characterized by short, obliquely oriented lamellae with a mosaic appearance in progressively deforming forms; polarization defines tissue conformations and localizes initiation of lamellar formation; ultrastructure of bone forming cells shows markedly dilated rough endoplasmic reticulum (RER) and prominent Golgi bodies with disorganized cisternae and swollen dispersed tubules and vesicles, structural indications of storage disorder/stress responses and mitochondrial swelling in cells with massively dilated RER indicating apoptosis; ultrastructural matrix assessments in woven bone show randomly oriented individual fibrils but also short pericellular bundles of parallel oriented fibrils positioned obliquely and oriented randomly to one another and in lamellar bone show unidirectional fibrils that deviate at slight angles to adjacent bundles and obliquely oriented fibril groups consistent with twisted plywood fibril organization. Histomorphometric indices, designed specifically to document woven and lamellar conformations in normal and OI bone, establish ratios for: i) cell area/total area X 100 indicating the percentage of an area occupied by cells (cellularity index) and ii) total area/number of cells (pericellular matrix domains). Woven bone is more cellular than lamellar bone and OI bone is more cellular than normal bone, but these findings occur in a highly specific fashion with values (high to low) encompassing OI woven, normal woven, OI lamellar and normal lamellar conformations. Conversely, for the total area/number of cells ratio, pericellular matrix accumulations in OI woven are smallest and normal lamellar largest. Since genotype-phenotype correlation is not definitive, interposing histologic/structural analysis allowing for a genotype-histopathologic-phenotype correlation will greatly enhance understanding and clinical management of OI.

4.
Cell Stem Cell ; 27(1): 50-63.e5, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32619518

RESUMEN

Modulating signaling pathways including Wnt and Hippo can induce cardiomyocyte proliferation in vivo. Applying these signaling modulators to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro can expand CMs modestly (<5-fold). Here, we demonstrate massive expansion of hiPSC-CMs in vitro (i.e., 100- to 250-fold) by glycogen synthase kinase-3ß (GSK-3ß) inhibition using CHIR99021 and concurrent removal of cell-cell contact. We show that GSK-3ß inhibition suppresses CM maturation, while contact removal prevents CMs from cell cycle exit. Remarkably, contact removal enabled 10 to 25 times greater expansion beyond GSK-3ß inhibition alone. Mechanistically, persistent CM proliferation required both LEF/TCF activity and AKT phosphorylation but was independent from yes-associated protein (YAP) signaling. Engineered heart tissues from expanded hiPSC-CMs showed comparable contractility to those from unexpanded hiPSC-CMs, demonstrating uncompromised cellular functionality after expansion. In summary, we uncovered a molecular interplay that enables massive hiPSC-CM expansion for large-scale drug screening and tissue engineering applications.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3 beta , Humanos , Miocitos Cardíacos
5.
J Bone Miner Res ; 35(4): 698-713, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31793059

RESUMEN

Although induced pluripotent stem cells hold promise as a potential source of osteoblasts for skeletal regeneration, the induction of pluripotency followed by directed differentiation into osteoblasts is time consuming and low yield. In contrast, direct lineage reprogramming without an intervening stem/progenitor cell stage would be a more efficient approach to generate osteoblasts. We screened combinations of osteogenic transcription factors and identified four factors, Runx2, Osx, Dlx5, and ATF4, that rapidly and efficiently reprogram mouse fibroblasts derived from 2.3 kb type I collagen promoter-driven green fluorescent protein (Col2.3GFP) transgenic mice into induced osteoblast cells (iOBs). iOBs exhibit osteoblast morphology, form mineralized nodules, and express Col2.3GFP and gene markers of osteoblast differentiation. The global transcriptome profiles validated that iOBs resemble primary osteoblasts. Genomewide DNA methylation analysis demonstrates that within differentially methylated loci, the methylation status of iOBs more closely resembles primary osteoblasts than mouse fibroblasts. We further demonstrate that Col2.3GFP+ iOBs have transcriptome profiles similar to GFP+ cells harvested from Col2.3GFP mouse bone chips. Functionally, Col2.3GFP+ iOBs form mineralized bone structures after subcutaneous implantation in immunodeficient mice and contribute to bone healing in a tibia bone fracture model. These findings provide an approach to derive and study osteoblasts for skeletal regeneration. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Osteoblastos , Animales , Diferenciación Celular , Fibroblastos , Ratones , Ratones Transgénicos , Cráneo
6.
Biomaterials ; 196: 31-45, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29456164

RESUMEN

Appropriate and abundant sources of bone-forming osteoblasts are essential for bone tissue engineering. Pluripotent stem cells can self-renew and thereby offer a potentially unlimited supply of osteoblasts, a significant advantage over other cell sources. We generated mouse embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) from transgenic mice expressing rat 2.3 kb type I collagen promoter-driven green fluorescent protein (Col2.3GFP), a reporter of the osteoblast lineage. We demonstrated that Col2.3GFP ESCs and iPSCs can be successfully differentiated to osteoblast lineage cells that express Col2.3GFP in vitro. We harvested GFP+ osteoblasts differentiated from ESCs. Genome wide gene expression profiles validated that ESC- and iPSC-derived osteoblasts resemble calvarial osteoblasts, and that Col2.3GFP expression serves as a marker for mature osteoblasts. Our results confirm the cell identity of ESC- and iPSC-derived osteoblasts and highlight the potential of pluripotent stem cells as a source of osteoblasts for regenerative medicine.


Asunto(s)
Regeneración Ósea/fisiología , Osteoblastos/citología , Células Madre Pluripotentes/citología , Animales , Regeneración Ósea/genética , Diferenciación Celular/genética , Línea Celular , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Osteoblastos/metabolismo , Cráneo/citología , Transcriptoma/genética , Regulación hacia Arriba/genética
7.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878134

RESUMEN

Advanced breast cancer is frequently associated with skeletal metastases and accelerated bone loss. Recombinant parathyroid hormone [teriparatide, PTH(1-34)] is the first anabolic agent approved in the US for treatment of osteoporosis. While signaling through the PTH receptor in the osteoblast lineage regulates bone marrow hematopoietic niches, the effects of anabolic PTH on the skeletal metastatic niche are unknown. Here, we demonstrate, using orthotopic and intratibial models of 4T1 murine and MDA-MB-231 human breast cancer tumors, that anabolic PTH decreases both tumor engraftment and the incidence of spontaneous skeletal metastasis in mice. Microcomputed tomography and histomorphometric analyses revealed that PTH increases bone volume and reduces tumor engraftment and volume. Transwell migration assays with murine and human breast cancer cells revealed that PTH alters the gene expression profile of the metastatic niche, in particular VCAM-1, to inhibit recruitment of cancer cells. While PTH did not affect growth or migration of the primary tumor, it elicited several changes in the tumor gene expression profile resulting in a less metastatic phenotype. In conclusion, PTH treatment in mice alters the bone microenvironment, resulting in decreased cancer cell engraftment, reduced incidence of metastases, preservation of bone microarchitecture and prolonged survival.


Asunto(s)
Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Hormona Paratiroidea/uso terapéutico , Animales , Neoplasias Óseas/diagnóstico por imagen , Neoplasias de la Mama/genética , Línea Celular Tumoral , Microambiente Celular , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Ratones , Neoplasias del Bazo/secundario , Análisis de Supervivencia , Microtomografía por Rayos X
8.
Endocrinology ; 157(4): 1341-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26934299

RESUMEN

Patients with breast cancer (BCa) frequently have preexisting vitamin D deficiency (low serum 25-hydroxyvitamin D) when their cancer develops. A number of epidemiological studies show an inverse association between BCa risk and vitamin D status in humans, although some studies have failed to find an association. In addition, several studies have reported that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic indicators. However, it is unknown whether this association is mechanistically causative and, if so, whether it results from systemic or tumor autonomous effects of vitamin D signaling. We found that ablation of vitamin D receptor expression within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We show that vitamin D signaling inhibits the expression of the tumor progression gene Id1, and this pathway is abrogated in vitamin D deficiency in vivo in 2 murine models of BCa. These findings are relevant to humans, because we discovered that the mechanism of VDR regulation of Inhibitor of differentiation 1 (ID1) is conserved in human BCa cells, and there is a negative correlation between serum 25-hydroxyvitamin D levels and the level of ID1 in primary tumors from patients with BCa.


Asunto(s)
Neoplasias Mamarias Experimentales/metabolismo , Receptores de Calcitriol/metabolismo , Transducción de Señal , Deficiencia de Vitamina D/metabolismo , Animales , Western Blotting , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Neoplasias Mamarias Experimentales/complicaciones , Neoplasias Mamarias Experimentales/genética , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Receptores de Calcitriol/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vitamina D/análogos & derivados , Vitamina D/sangre , Vitamina D/metabolismo , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/genética
9.
Endocr Relat Cancer ; 23(4): 251-64, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26817629

RESUMEN

Obesity is an established risk factor for postmenopausal breast cancer (BCa), insulin resistance, and vitamin D deficiency, and all contribute to increased synthesis of mammary estrogens, the drivers of estrogen receptor-positive (ER+) BCa growth. As both dietary vitamin D and calcitriol treatments inhibit breast estrogen synthesis and signaling, we hypothesized that vitamin D would be especially beneficial in mitigating the adverse effects of obesity on ER+BCa. To assess whether obesity exerted adverse effects on BCa growth and whether vitamin D compounds could reduce these unfavorable effects, we employed a diet-induced obesity (DIO) model in ovariectomized C57BL/6 mice. Breast tumor cells originally from syngeneic Mmtv-Wnt1 transgenic mice were then implanted into the mammary fat pads of lean and obese mice. DIO accelerated the initiation and progression of the mammary tumors. Treatments with either calcitriol or dietary vitamin D reduced the adverse effects of obesity causing a delay in tumor appearance and inhibiting continued tumor growth. Beneficial actions of treatments with vitamin D or calcitriol on BCa and surrounding adipose tissue included repressed Esr1, aromatase, and Cox2 expression; decreased tumor-derived estrogen and PGE2; reduced expression of leptin receptors; and increased adiponectin receptors. We demonstrate that vitamin D treatments decreased insulin resistance, reduced leptin, and increased adiponectin signaling and also regulated the LKB1/AMPK pathway contributing to an overall decrease in local estrogen synthesis in the obese mice. We conclude that calcitriol and dietary vitamin D, acting by multiple interrelated pathways, mitigate obesity-enhanced BCa growth in a postmenopausal setting.


Asunto(s)
Suplementos Dietéticos , Neoplasias Mamarias Experimentales/metabolismo , Obesidad/metabolismo , Vitamina D/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/sangre , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Aromatasa/genética , Calcio/sangre , Ciclooxigenasa 2/genética , Dieta Alta en Grasa , Dinoprostona/metabolismo , Estradiol/metabolismo , Estrógenos/metabolismo , Estrona/metabolismo , Femenino , Humanos , Leptina/sangre , Células MCF-7 , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/complicaciones , Obesidad/patología , Ovariectomía , ARN Mensajero/metabolismo , Carga Tumoral , Vitamina D/sangre
10.
J Biol Chem ; 291(4): 1631-1642, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26598522

RESUMEN

Parathyroid hormone (PTH) is an important regulator of osteoblast function and is the only anabolic therapy currently approved for treatment of osteoporosis. The PTH receptor (PTH1R) is a G protein-coupled receptor that signals via multiple G proteins including Gsα. Mice expressing a constitutively active mutant PTH1R exhibited a dramatic increase in trabecular bone that was dependent upon expression of Gsα in the osteoblast lineage. Postnatal removal of Gsα in the osteoblast lineage (P-Gsα(OsxKO) mice) yielded markedly reduced trabecular and cortical bone mass. Treatment with anabolic PTH(1-34) (80 µg/kg/day) for 4 weeks failed to increase trabecular bone volume or cortical thickness in male and female P-Gsα(OsxKO) mice. Surprisingly, in both male and female mice, PTH administration significantly increased osteoblast numbers and bone formation rate in both control and P-Gsα(OsxKO) mice. In mice that express a mutated PTH1R that activates adenylyl cyclase and protein kinase A (PKA) via Gsα but not phospholipase C via Gq/11 (D/D mice), PTH significantly enhanced bone formation, indicating that phospholipase C activation is not required for increased bone turnover in response to PTH. Therefore, although the anabolic effect of intermittent PTH treatment on trabecular bone volume is blunted by deletion of Gsα in osteoblasts, PTH can stimulate osteoblast differentiation and bone formation. Together these findings suggest that alternative signaling pathways beyond Gsα and Gq/11 act downstream of PTH on osteoblast differentiation.


Asunto(s)
Anabolizantes/administración & dosificación , Desarrollo Óseo/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gs/deficiencia , Terapia de Reemplazo de Hormonas , Osteoporosis/tratamiento farmacológico , Osteoporosis/enzimología , Hormona Paratiroidea/administración & dosificación , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Diferenciación Celular/efectos de los fármacos , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Masculino , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Osteoblastos/metabolismo , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
11.
Mol Cancer Ther ; 14(8): 1951-61, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25934710

RESUMEN

The anticancer actions of vitamin D and its hormonally active form, calcitriol, have been extensively documented in clinical and preclinical studies. However, the mechanisms underlying these actions have not been completely elucidated. Here, we examined the effect of dietary vitamin D and calcitriol on mouse breast tumor-initiating cells (TICs, also known as cancer stem cells). We focused on MMTV-Wnt1 mammary tumors, for which markers for isolating TICs have previously been validated. We confirmed that these tumors expressed functional vitamin D receptors and estrogen receptors (ER) and exhibited calcitriol-induced molecular responses including ER downregulation. Following orthotopic implantation of MMTV-Wnt1 mammary tumor cells into mice, calcitriol injections or a vitamin D-supplemented diet caused a striking delay in tumor appearance and growth, whereas a vitamin D-deficient diet accelerated tumor appearance and growth. Calcitriol inhibited TIC tumor spheroid formation in a dose-dependent manner in primary cultures and inhibited TIC self-renewal in secondary passages. A combination of calcitriol and ionizing radiation inhibited spheroid formation more than either treatment alone. Further, calcitriol significantly decreased TIC frequency as evaluated by in vivo limiting dilution analyses. Calcitriol inhibition of TIC spheroid formation could be overcome by the overexpression of ß-catenin, suggesting that the inhibition of Wnt/ß-catenin pathway is an important mechanism mediating the TIC inhibitory activity of calcitriol in this tumor model. Our findings indicate that vitamin D compounds target breast TICs reducing tumor-initiating activity. Our data also suggest that combining vitamin D compounds with standard therapies may enhance anticancer activity and improve therapeutic outcomes.


Asunto(s)
Calcitriol/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Vitamina D/farmacología , Animales , Peso Corporal , Calcio/sangre , Línea Celular Tumoral , Estrógenos/metabolismo , Femenino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Células Madre Neoplásicas/metabolismo , Receptores de Calcitriol/metabolismo , Receptores de Estrógenos/metabolismo , Carga Tumoral , Vitamina D/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
12.
Nat Rev Cancer ; 14(5): 342-57, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24705652

RESUMEN

Vitamin D is not really a vitamin but the precursor to the potent steroid hormone calcitriol, which has widespread actions throughout the body. Calcitriol regulates numerous cellular pathways that could have a role in determining cancer risk and prognosis. Although epidemiological and early clinical trials are inconsistent, and randomized control trials in humans do not yet exist to conclusively support a beneficial role for vitamin D, accumulating results from preclinical and some clinical studies strongly suggest that vitamin D deficiency increases the risk of developing cancer and that avoiding deficiency and adding vitamin D supplements might be an economical and safe way to reduce cancer incidence and improve cancer prognosis and outcome.


Asunto(s)
Calcitriol/fisiología , Neoplasias/patología , Neoplasias/prevención & control , Vitamina D/fisiología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Neoplasias de la Mama/metabolismo , Colecalciferol/fisiología , Neoplasias del Colon/metabolismo , Progresión de la Enfermedad , Sistema Endocrino , Femenino , Humanos , Masculino , Células Madre Neoplásicas/citología , Polimorfismo Genético , Pronóstico , Neoplasias de la Próstata/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Riesgo , Transducción de Señal , Esteroide Hidroxilasas/metabolismo , Deficiencia de Vitamina D/complicaciones , Vitamina D3 24-Hidroxilasa
13.
Endocr Relat Cancer ; 20(4): 565-77, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23744764

RESUMEN

Calcitriol (1,25-dihydroxyvitamin D3), the hormonally active metabolite of vitamin D, exerts its anti-proliferative activity in breast cancer (BCa) cells by multiple mechanisms including the downregulation of the expression of estrogen receptor α (ER). We analyzed an ∼3.5 kb ER promoter sequence and demonstrated the presence of two potential negative vitamin D response elements (nVDREs), a newly identified putative nVDRE upstream at -2488 to -2473 bp (distal nVDRE) and a previously published sequence (proximal nVDRE) at -94 to -70 bp proximal to the P1 start site. Transactivation analysis using ER promoter deletion constructs and heterologous promoter-reporter constructs revealed that both nVDREs functioned to mediate calcitriol transrepression. In the electrophoretic mobility shift assay, the vitamin D receptor (VDR) showed strong binding to both nVDREs in the presence of calcitriol, and the chromatin immunoprecipitation assay demonstrated the recruitment of the VDR to the distal nVDRE site. Mutations in the 5' hexameric DNA sequence of the distal nVDRE resulted in the loss of calcitriol-mediated transrepression and the inhibition of protein-DNA complex formation, demonstrating the importance of these nucleotides in VDR DNA binding and transrepression. A putative nuclear factor-Y (NFY) binding site, identified within the distal nVDRE, led to the findings that NFY bound to the distal nVDRE site interfered with the binding of the VDR at the site and reduced calcitriol-mediated transrepression. In conclusion, the ER promoter region contains two negative VDREs that act in concert to bind to the VDR and both nVDREs are required for the maximal inhibition of ER expression by calcitriol. The suppression of ER expression and estrogen-mediated signaling by calcitriol in BCa cells suggests that vitamin D may be useful in the treatment of ER+ BCa.


Asunto(s)
Neoplasias de la Mama/genética , Calcitriol/farmacología , Receptores de Estrógenos/genética , Elemento de Respuesta a la Vitamina D/genética , Vitaminas/farmacología , Neoplasias de la Mama/metabolismo , Factor de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Regiones Promotoras Genéticas , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Receptores X Retinoide/metabolismo
14.
J Steroid Biochem Mol Biol ; 136: 289-95, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22939886

RESUMEN

Calcitriol [1,25(OH)2D3], the hormonally active form of vitamin D exerts anti-proliferative, pro-apoptotic, anti-inflammatory effects and other anticancer actions in breast cancer (BCa) cell cultures and animal models of BCa. Our research is focused on investigating the potential beneficial effects of dietary vitamin D3 compared to calcitriol and the underlying mechanisms in BCa treatment and chemoprevention. We recently found that dietary vitamin D3 exhibits significant tumor inhibitory effects in xenograft models of BCa that are equivalent to those elicited by the administration of the active hormone calcitriol. At the easily achievable dose tested in our studies, dietary vitamin D3 exhibited substantial tumor inhibitory activity and, unlike calcitriol, did not cause hypercalcemia demonstrating its relative safety. We found elevations in circulating calcitriol as well as increased CYP27B1 expression in the tumor and the intestine in tumor-bearing mice ingesting a vitamin D3-supplemented diet. We hypothesize that the elevation in circulating 25(OH)D induced by dietary vitamin D3 supplements stimulates local synthesis of calcitriol in the mammary tumor microenvironment and the ensuing paracrine/autocrine actions play a major role in the anticancer activity of dietary vitamin D3. Our findings suggest that the endocrine activity of calcitriol derived from tumor and other extra-renal sources such as the intestine, probably also plays a role in mediating the anticancer effects of dietary vitamin D3. Thus it appears that multiple sites of 1α-hydroxylation contribute to the anticancer effects of dietary vitamin D3. Our data strongly suggest that dietary vitamin D will be useful in the chemoprevention and treatment of BCa since it is a safe, economical and easily available nutritional agent that is equivalent to calcitriol in exerting anticancer effects, at least in mouse models. Furthermore, adequate vitamin D nutrition and avoidance of vitamin D deficiency appear to be important in reducing BCa risk. These findings warrant clinical trials in BCa patients and in women at high risk for BCa to evaluate the benefits of dietary vitamin D3 supplementation. This article is part of a Special Issue entitled 'Vitamin D Workshop'.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/fisiología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/uso terapéutico , Calcitriol/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/prevención & control , Vitamina D/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Calcitriol/uso terapéutico , Suplementos Dietéticos , Femenino , Humanos , Neoplasias Mamarias Experimentales/enzimología , Ratones , Equivalencia Terapéutica , Microambiente Tumoral , Vitamina D/uso terapéutico
15.
Steroids ; 77(11): 1107-12, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22801352

RESUMEN

Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active form of vitamin D, inhibits the growth of many malignant cells including breast cancer (BCa) cells. The mechanisms of calcitriol anticancer actions include cell cycle arrest, stimulation of apoptosis and inhibition of invasion, metastasis and angiogenesis. In addition we have discovered new pathways of calcitriol action that are especially relevant in inhibiting the growth of estrogen receptor positive (ER+) BCa cells. Calcitriol suppresses COX-2 expression and increases that of 15-PGDH thereby reducing the levels of inflammatory prostaglandins (PGs). Our in vitro and in vivo studies show that calcitriol decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis selectively in BCa cells and in the mammary adipose tissue surrounding BCa, by a direct repression of aromatase transcription via promoter II as well as an indirect effect due to the reduction in the levels of PGs, which are major stimulator of aromatase transcription through promoter II. Calcitriol down-regulates the expression of ERα and thereby attenuates estrogen signaling in BCa cells including the proliferative stimulus provided by estrogens. Thus the inhibition of estrogen synthesis and signaling by calcitriol and its anti-inflammatory actions will play an important role in inhibiting ER+BCa. We hypothesize that dietary vitamin D would exhibit similar anticancer activity due to the presence of the enzyme 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1) in breast cells ensuring conversion of circulating 25-hydroxyvitamin D to calcitriol locally within the breast micro-environment where it can act in a paracrine manner to inhibit BCa growth. Cell culture and in vivo data in mice strongly suggest that calcitriol and dietary vitamin D would play a beneficial role in the prevention and/or treatment of ER+BCa in women.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Receptores de Estrógenos/metabolismo , Vitamina D/farmacología , Vitaminas/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/prevención & control , Calcitriol/farmacología , Calcitriol/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Suplementos Dietéticos , Estrógenos/metabolismo , Femenino , Humanos , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/prevención & control , Vitamina D/administración & dosificación , Vitamina D/uso terapéutico , Vitaminas/administración & dosificación , Vitaminas/uso terapéutico
16.
Prostate ; 72(15): 1628-37, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22457201

RESUMEN

BACKGROUND: The potential role of vitamin D and soy in prostate cancer (PCa) prevention/treatment has gained much attention in recent years. In this study, we evaluated the anticancer activity of calcitriol, the active form of vitamin D, dietary soy, and their combinations in a mouse model of PCa. METHODS: Athymic male nude mice bearing PC-3 human PCa xenografts received diets containing 10 or 20 kcal% soy, calcitriol injections, or a combination of dietary soy and calcitriol. Changes in tumor growth, serum levels of 1,25(OH)(2)D and calcium, and regulation of tumor gene expression were examined. RESULTS: The combination treatments resulted in substantially greater inhibition of tumor growth than either agent alone. Soy diets alone caused a modest elevation in serum 1,25(OH)(2)D, whereas the calcitriol-soy combinations led to substantially elevated serum 1,25(OH)(2) D, hypercalcemia, and in some cases lethal toxicity. The combinations enhanced calcitriol activity in regulating target gene expression, including greater up-regulation of anti-proliferative (p21, IGFBP-3) and pro-apoptotic (Bax) genes, increased inhibition of anti-apoptotic (Bcl-2) and cell cycle promoting (cyclin D1) genes, and suppression of prostaglandin (PG) synthesis and signaling (COX-2, 15-PGDH, PG receptors). Increases in serum calcium were accompanied by elevated expression of intestinal calcium absorption genes (TRPV6, calbindin-9k). CONCLUSIONS: Soy increases the bioavailability of endogenous and administered calcitriol, thereby enhancing its anticancer effects and risk of hypercalcemia. Since both agents are easily available as dietary supplements, the increased potential for hypercalcemic toxicity becomes an important factor when considering the combined use of vitamin D and soy in PCa therapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Calcitriol/uso terapéutico , Hipercalcemia/inducido químicamente , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas de Soja/administración & dosificación , Vitaminas/uso terapéutico , Adenocarcinoma/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Apoptosis/genética , Calcitriol/efectos adversos , Calcitriol/sangre , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Suplementos Dietéticos , Quimioterapia Combinada , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Hipercalcemia/patología , Masculino , Ratones , Ratones Desnudos , Prostaglandinas/biosíntesis , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Proteínas de Soja/efectos adversos , Vitaminas/efectos adversos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Endocrinology ; 153(6): 2576-87, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22454149

RESUMEN

1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3) or calcitriol], the hormonally active vitamin D metabolite, exhibits anticancer actions in models of breast cancer and prostate cancer. Because CYP27B1 (1α-hydroxylase), the enzyme catalyzing 1,25(OH)(2)D(3) formation in the kidney, is also expressed in extrarenal tissues, we hypothesize that dietary vitamin D(3) will be converted to 25(OH)D(3) in the body and then to 1,25(OH)(2)D(3) locally in the cancer microenvironment in which it will exert autocrine/paracrine anticancer actions. Immunocompromised mice bearing MCF-7 breast cancer xenografts showed significant tumor shrinkage (>50%) after ingestion of a vitamin D(3)-supplemented diet (5000 IU/kg) compared with a control diet (1000 IU/kg). Dietary vitamin D(3) inhibition of tumor growth was equivalent to administered calcitriol (0.025, 0.05, or 0.1 µg/mouse, three times a week). Both treatments equivalently inhibited PC-3 prostate cancer xenograft growth but to a lesser extent than the MCF-7 tumors. Calcitriol at 0.05 µg and 0.1 µg caused modest but statistically significant increases in serum calcium levels indicating that the dietary vitamin D(3) comparison was to a maximally safe calcitriol dose. Dietary vitamin D(3) did not increase serum calcium, demonstrating its safety at the concentration tested. The vitamin D(3) diet raised circulating 1,25 dihydroxyvitamin D levels and did not alter CYP27B1 mRNA in the kidney but increased it in the tumors, suggesting that extrarenal sources including the tumors contributed to the elevated circulating 1,25 dihydroxyvitamin D(3). Both calcitriol and dietary vitamin D(3) were equipotent in suppressing estrogen synthesis and signaling and other proinflammatory and growth signaling pathways. These preclinical data demonstrate the potential utility of dietary vitamin D(3) supplementation in cancer prevention and therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Calcitriol/farmacología , Colecalciferol/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Animales , Peso Corporal/efectos de los fármacos , Neoplasias de la Mama/patología , Calcitriol/administración & dosificación , Calcio/sangre , Línea Celular Tumoral , Colecalciferol/administración & dosificación , Suplementos Dietéticos , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Ovariectomía , Neoplasias de la Próstata/patología , Receptores de Calcitriol/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esteroide Hidroxilasas/genética , Carga Tumoral/efectos de los fármacos , Vitamina D3 24-Hidroxilasa , Vitaminas/farmacología
18.
Horm Cancer ; 2(3): 190-202, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21686077

RESUMEN

Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active metabolite of vitamin D, exerts many anticancer effects in breast cancer (BCa) cells. We have previously shown using cell culture models that calcitriol acts as a selective aromatase modulator (SAM) and inhibits estrogen synthesis and signaling in BCa cells. We have now examined calcitriol effects in vivo on aromatase expression, estrogen signaling, and tumor growth when used alone and in combination with aromatase inhibitors (AIs). In immunocompromised mice bearing MCF-7 xenografts, increasing doses of calcitriol exhibited significant tumor inhibitory effects (~50% to 70% decrease in tumor volume). At the suboptimal doses tested, anastrozole and letrozole also caused significant tumor shrinkage when used individually. Although the combinations of calcitriol and the AIs caused a statistically significant increase in tumor inhibition in comparison to the single agents, the cooperative interaction between these agents appeared to be minimal at the doses tested. Calcitriol decreased aromatase expression in the xenograft tumors. Importantly, calcitriol also acted as a SAM in the mouse, decreasing aromatase expression in the mammary adipose tissue, while increasing it in bone marrow cells and not altering it in the ovaries and uteri. As a result, calcitriol significantly reduced estrogen levels in the xenograft tumors and surrounding breast adipose tissue. In addition, calcitriol inhibited estrogen signaling by decreasing tumor ERα levels. Changes in tumor gene expression revealed the suppressive effects of calcitriol on inflammatory and growth signaling pathways and demonstrated cooperative interactions between calcitriol and AIs to modulate gene expression. We hypothesize that cumulatively these calcitriol actions would contribute to a beneficial effect when calcitriol is combined with an AI in the treatment of BCa.


Asunto(s)
Inhibidores de la Aromatasa/farmacología , Aromatasa/biosíntesis , Calcitriol/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Vitaminas/farmacología , Anastrozol , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estrógenos/metabolismo , Femenino , Humanos , Inmunohistoquímica , Letrozol , Neoplasias Mamarias Experimentales/enzimología , Ratones , Ratones Desnudos , Nitrilos/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Cell Endocrinol ; 347(1-2): 61-9, 2011 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-21664249

RESUMEN

Prostate cancer (PCa) is the second most common cancer in men worldwide. Epidemiological, molecular, and cellular studies have implicated vitamin D deficiency as a risk factor for the development and/or progression of PCa. Studies using cell culture systems and animal models suggest that vitamin D acts to reduce the growth of PCa through regulation of cellular proliferation and differentiation. However, although preclinical studies provide a strong indication for anti-cancer activity, proof of therapeutic benefits in men is still lacking. The anti-proliferative and pro-differentiating properties of vitamin D have been attributed to calcitriol [1,25(OH)(2)D(3)], the hormonally active form of vitamin D, acting through the vitamin D receptor (VDR). Metabolism of vitamin D in target tissues is mediated by two key enzymes: 1α-hydroxylase (CYP27B1), which catalyzes the synthesis of calcitriol from 25(OH)D and 24-hydroxylase (CYP24), which catalyzes the initial step in the conversion of calcitriol to less active metabolites. Many factors affect the balance of calcitriol synthesis and catabolism and several maneuvers, like combination therapy of calcitriol with other drugs, have been explored to treat PCa and reduce its risk. The current paper is an overview addressing some of the key factors that influence the biological actions of vitamin D and its metabolites in the treatment and/or prevention of PCa.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Deficiencia de Vitamina D/complicaciones , Vitamina D/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/fisiología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Calcitriol/administración & dosificación , Calcitriol/uso terapéutico , Colecalciferol/administración & dosificación , Colecalciferol/uso terapéutico , Ensayos Clínicos como Asunto , Quimioterapia Combinada , Humanos , Isoflavonas/administración & dosificación , Isoflavonas/uso terapéutico , Masculino , Ratones , Neoplasias de la Próstata/complicaciones , Neoplasias de la Próstata/prevención & control , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Receptores de Calcitriol/fisiología , Factores de Riesgo , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/metabolismo , Esteroide Hidroxilasas/fisiología , Vitamina D/fisiología , Vitamina D3 24-Hidroxilasa
20.
J Steroid Biochem Mol Biol ; 121(1-2): 343-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20156557

RESUMEN

Calcitriol (1,25-dihydroxyvitamin D3), the hormonally active metabolite of vitamin D, inhibits the growth and induces the differentiation of many malignant cells including breast cancer (BCa) cells. Calcitriol exerts its anti-proliferative activity in BCa cells by inducing cell cycle arrest and stimulating apoptosis. Calcitriol also inhibits invasion, metastasis and tumor angiogenesis in experimental models of BCa. Our recent studies show additional newly discovered pathways of calcitriol action to inhibit the growth of BCa cells. Calcitriol suppresses COX-2 expression and increases that of 15-PGDH thereby reducing the levels and biological activity of prostaglandins (PGs). Calcitriol decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis selectively in BCa cells and the breast adipose tissue surrounding BCa, by a direct repression of aromatase transcription via promoter II as well as an indirect effect due to the reduction in the levels and biological activity of PGE2, which is a major stimulator of aromatase transcription through promoter II in BCa. Calcitriol down-regulates the expression of estrogen receptor alpha and thereby attenuates estrogen signaling in BCa cells including the proliferative stimulus provided by estrogens. We hypothesize that the inhibition of estrogen synthesis and signaling by calcitriol and its anti-inflammatory actions will play an important role in the use of calcitriol for the prevention and/or treatment of BCa.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antagonistas de Estrógenos/metabolismo , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Vitamina D/metabolismo , Tejido Adiposo/metabolismo , Animales , Apoptosis , Mama/patología , Calcitriol/metabolismo , Ciclooxigenasa 2/metabolismo , Femenino , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Metástasis de la Neoplasia , Neovascularización Patológica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA