Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Eur Urol Oncol ; 5(2): 164-175, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34774481

RESUMEN

BACKGROUND: No curative therapy is currently available for metastatic prostate cancer (PCa). The diverse mechanisms of progression include fibroblast growth factor (FGF) axis activation. OBJECTIVE: To investigate the molecular and clinical implications of fibroblast growth factor receptor 1 (FGFR1) and its isoforms (α/ß) in the pathogenesis of PCa bone metastases. DESIGN, SETTING, AND PARTICIPANTS: In silico, in vitro, and in vivo preclinical approaches were used. RNA-sequencing and immunohistochemical (IHC) studies in human samples were conducted. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: In mice, bone metastases (chi-square/Fisher's test) and survival (Mantel-Cox) were assessed. In human samples, FGFR1 and ladinin 1 (LAD1) analysis associated with PCa progression were evaluated (IHC studies, Fisher's test). RESULTS AND LIMITATIONS: FGFR1 isoform expression varied among PCa subtypes. Intracardiac injection of mice with FGFR1-expressing PC3 cells reduced mouse survival (α, p < 0.0001; ß, p = 0.032) and increased the incidence of bone metastases (α, p < 0.0001; ß, p = 0.02). Accordingly, IHC studies of human castration-resistant PCa (CRPC) bone metastases revealed significant enrichment of FGFR1 expression compared with treatment-naïve, nonmetastatic primary tumors (p = 0.0007). Expression of anchoring filament protein LAD1 increased in FGFR1-expressing PC3 cells and was enriched in human CRPC bone metastases (p = 0.005). CONCLUSIONS: FGFR1 expression induces bone metastases experimentally and is significantly enriched in human CRPC bone metastases, supporting its prometastatic effect in PCa. LAD1 expression, found in the prometastatic PCa cells expressing FGFR1, was also enriched in CRPC bone metastases. Our studies support and provide a roadmap for the development of FGFR blockade for advanced PCa. PATIENT SUMMARY: We studied the role of fibroblast growth factor receptor 1 (FGFR1) in prostate cancer (PCa) progression. We found that PCa cells with high FGFR1 expression increase metastases and that FGFR1 expression is increased in human PCa bone metastases, and identified genes that could participate in the metastases induced by FGFR1. These studies will help pinpoint PCa patients who use fibroblast growth factor to progress and will benefit by the inhibition of this pathway.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Próstata Resistentes a la Castración , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Factores de Crecimiento de Fibroblastos , Humanos , Masculino , Ratones , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
2.
Cell Metab ; 4(6): 441-51, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17141628

RESUMEN

The transcription factor ATF4 enhances bone formation by favoring amino acid import and collagen synthesis in osteoblasts, a function requiring its phosphorylation by RSK2, the kinase inactivated in Coffin-Lowry Syndrome. Here, we show that in contrast, RSK2 activity, ATF4-dependent collagen synthesis, and bone formation are increased in mice lacking neurofibromin in osteoblasts (Nf1(ob)(-/-) mice). Independently of RSK2, ATF4 phosphorylation by PKA is enhanced in Nf1(ob)(-/-) mice, thereby increasing Rankl expression, osteoclast differentiation, and bone resorption. In agreement with ATF4 function in amino acid transport, a low-protein diet decreased bone protein synthesis and normalized bone formation and bone mass in Nf1(ob)(-/-) mice without affecting other organ weight, while a high-protein diet overcame Atf4(-/-) and Rsk2(-/-) mice developmental defects, perinatal lethality, and low bone mass. By showing that ATF4-dependent skeletal dysplasiae are treatable by dietary manipulations, this study reveals a molecular connection between nutrition and skeletal development.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Enfermedades del Desarrollo Óseo/dietoterapia , Enfermedades del Desarrollo Óseo/metabolismo , Proteínas en la Dieta/uso terapéutico , Neurofibromina 1/metabolismo , Osteoblastos/metabolismo , Aminoácidos/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/genética , Enfermedades del Desarrollo Óseo/congénito , Enfermedades del Desarrollo Óseo/patología , Resorción Ósea/dietoterapia , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Síndrome de Coffin-Lowry/genética , Síndrome de Coffin-Lowry/metabolismo , Síndrome de Coffin-Lowry/patología , Colágeno/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Ratones Noqueados , Neurofibromina 1/deficiencia , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Osteogénesis/genética , Ligando RANK/biosíntesis , Ligando RANK/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/deficiencia , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
3.
Dev Cell ; 10(6): 771-82, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16740479

RESUMEN

Development and repair of the vertebrate skeleton requires the precise coordination of bone-forming osteoblasts and bone-resorbing osteoclasts. In diseases such as osteoporosis, bone resorption dominates over bone formation, suggesting a failure to harmonize osteoclast and osteoblast function. Here, we show that mice expressing a constitutively nuclear NFATc1 variant (NFATc1(nuc)) in osteoblasts develop high bone mass. NFATc1(nuc) mice have massive osteoblast overgrowth, enhanced osteoblast proliferation, and coordinated changes in the expression of Wnt signaling components. In contrast, viable NFATc1-deficient mice have defects in skull bone formation in addition to impaired osteoclast development. NFATc1(nuc) mice have increased osteoclastogenesis despite normal levels of RANKL and OPG, indicating that an additional NFAT-regulated mechanism influences osteoclastogenesis in vivo. Calcineurin/NFATc signaling in osteoblasts controls the expression of chemoattractants that attract monocytic osteoclast precursors, thereby coupling bone formation and bone resorption. Our results indicate that NFATc1 regulates bone mass by functioning in both osteoblasts and osteoclasts.


Asunto(s)
Densidad Ósea , Calcineurina/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoblastos/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Modelos Biológicos , Factores de Transcripción NFATC/genética , Osteoblastos/citología , Osteoblastos/fisiología , Cráneo/citología
4.
J Clin Invest ; 118(8): 2697-710, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18618013

RESUMEN

In prostate cancer, androgen blockade strategies are commonly used to treat osteoblastic bone metastases. However, responses to these therapies are typically brief, and the mechanism underlying androgen-independent progression is not clear. Here, we established what we believe to be the first human androgen receptor-negative prostate cancer xenografts whose cells induced an osteoblastic reaction in bone and in the subcutis of immunodeficient mice. Accordingly, these cells grew in castrated as well as intact male mice. We identified FGF9 as being overexpressed in the xenografts relative to other bone-derived prostate cancer cells and discovered that FGF9 induced osteoblast proliferation and new bone formation in a bone organ assay. Mice treated with FGF9-neutralizing antibody developed smaller bone tumors and reduced bone formation. Finally, we found positive FGF9 immunostaining in prostate cancer cells in 24 of 56 primary tumors derived from human organ-confined prostate cancer and in 25 of 25 bone metastasis cases studied. Collectively, these results suggest that FGF9 contributes to prostate cancer-induced new bone formation and may participate in the osteoblastic progression of prostate cancer in bone. Androgen receptor-null cells may contribute to the castration-resistant osteoblastic progression of prostate cancer cells in bone and provide a preclinical model for studying therapies that target these cells.


Asunto(s)
Neoplasias Óseas/secundario , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Osteogénesis/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Cariotipificación , Queratinas/metabolismo , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Metástasis de la Neoplasia , Trasplante de Neoplasias , Orquiectomía , Técnicas de Cultivo de Órganos , Osteoblastos/metabolismo , Neoplasias de la Próstata/genética , Trasplante Heterólogo , Vimentina/metabolismo
5.
Nature ; 434(7032): 514-20, 2005 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-15724149

RESUMEN

Bone remodelling, the mechanism by which vertebrates regulate bone mass, comprises two phases, namely resorption by osteoclasts and formation by osteoblasts; osteoblasts are multifunctional cells also controlling osteoclast differentiation. Sympathetic signalling via beta2-adrenergic receptors (Adrb2) present on osteoblasts controls bone formation downstream of leptin. Here we show, by analysing Adrb2-deficient mice, that the sympathetic nervous system favours bone resorption by increasing expression in osteoblast progenitor cells of the osteoclast differentiation factor Rankl. This sympathetic function requires phosphorylation (by protein kinase A) of ATF4, a cell-specific CREB-related transcription factor essential for osteoblast differentiation and function. That bone resorption cannot increase in gonadectomized Adrb2-deficient mice highlights the biological importance of this regulation, but also contrasts sharply with the increase in bone resorption characterizing another hypogonadic mouse with low sympathetic tone, the ob/ob mouse. This discrepancy is explained, in part, by the fact that CART ('cocaine amphetamine regulated transcript'), a neuropeptide whose expression is controlled by leptin and nearly abolished in ob/ob mice, inhibits bone resorption by modulating Rankl expression. Our study establishes that leptin-regulated neural pathways control both aspects of bone remodelling, and demonstrates that integrity of sympathetic signalling is necessary for the increase in bone resorption caused by gonadal failure.


Asunto(s)
Resorción Ósea , Leptina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso Simpático/metabolismo , Factor de Transcripción Activador 4 , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Niño , Preescolar , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Eliminación de Gen , Humanos , Leptina/genética , Leptina/farmacología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Modelos Neurológicos , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Osteoblastos/metabolismo , Osteogénesis , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética , Receptores Adrenérgicos beta 2/deficiencia , Receptores Adrenérgicos beta 2/genética , Receptores de Leptina , Elementos de Respuesta/genética , Transducción de Señal/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Cancers (Basel) ; 13(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34885132

RESUMEN

Renal medullary carcinoma (RMC) is a lethal malignancy affecting individuals with sickle hemoglobinopathies. Currently, no modifiable risk factors are known. We aimed to determine whether high-intensity exercise is a risk factor for RMC in individuals with sickle cell trait (SCT). We used multiple approaches to triangulate our conclusion. First, a case-control study was conducted at a single tertiary-care facility. Consecutive patients with RMC were compared to matched controls with similarly advanced genitourinary malignancies in a 1:2 ratio and compared on rates of physical activity and anthropometric measures, including skeletal muscle surface area. Next, we compared the rate of military service among our RMC patients to a similarly aged population of black individuals with SCT in the U.S. Further, we used genetically engineered mouse models of SCT to study the impact of exercise on renal medullary hypoxia. Compared with matched controls, patients with RMC reported higher physical activity and had higher skeletal muscle surface area. A higher proportion of patients with RMC reported military service than expected compared to the similarly-aged population of black individuals with SCT. When exposed to high-intensity exercise, mice with SCT demonstrated significantly higher renal medulla hypoxia compared to wild-type controls. These data suggest high-intensity exercise is the first modifiable risk factor for RMC in individuals with SCT.

7.
Dev Cell ; 8(5): 751-64, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15866165

RESUMEN

Inactivation of beta-catenin in mesenchymal progenitors prevents osteoblast differentiation; inactivation of Lrp5, a gene encoding a likely Wnt coreceptor, results in low bone mass (osteopenia) by decreasing bone formation. These observations indicate that Wnt signaling controls osteoblast differentiation and suggest that it may regulate bone formation in differentiated osteoblasts. Here, we study later events and find that stabilization of beta-catenin in differentiated osteoblasts results in high bone mass, while its deletion from differentiated osteoblasts leads to osteopenia. Surprisingly, histological analysis showed that these mutations primarily affect bone resorption rather than bone formation. Cellular and molecular studies showed that beta-catenin together with TCF proteins regulates osteoblast expression of Osteoprotegerin, a major inhibitor of osteoclast differentiation. These findings demonstrate that beta-catenin, and presumably Wnt signaling, promote the ability of differentiated osteoblasts to inhibit osteoclast differentiation; thus, they broaden our knowledge of the functions Wnt proteins have at various stages of skeletogenesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Animales , Desarrollo Óseo , Diferenciación Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas Relacionadas con Receptor de LDL , Operón Lac , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Osteogénesis , Osteopetrosis/etiología , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Wnt , beta Catenina
8.
Antioxid Redox Signal ; 32(17): 1243-1258, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31861963

RESUMEN

Aims: Bone is the most frequent site of prostate cancer (PCa) metastasis. Tumor cells interact with the bone microenvironment interrupting tissue balance. Heme oxygenase-1 (HO-1; encoded by Hmox1) appears as a potential target in PCa maintaining the cellular homeostasis. Our hypothesis is that HO-1 is implicated in bone physiology and modulates the communication with PCa cells. Here we aimed at (i) assessing the physiological impact of Hmox1 gene knockout (KO) on bone metabolism in vivo and (ii) determining the alterations of the transcriptional landscape associated with tumorigenesis and bone remodeling in cells growing in coculture (PCa cells with primary mouse osteoblasts [PMOs] from BALB/c Hmox1+/+, Hmox1+/-, and Hmox1-/- mice). Results: Histomorphometric analysis of Hmox1-/- mice bones exhibited significantly decreased bone density with reduced remodeling parameters. A positive correlation between Hmox1 expression and Runx2, Col1a1, Csf1, and Opg genes was observed in PMOs. Flow cytometry studies revealed two populations of PMOs with different reactive oxygen species (ROS) levels. The high ROS population was increased in PMOs Hmox1+/- compared with Hmox1+/+, but was significantly reduced in PMOs Hmox1-/-, suggesting restrained ROS tolerance in KO cells. Gene expression was altered in PMOs upon coculture with PCa cells, showing a pro-osteoclastic profile. Moreover, HO-1 induction in PCa cells growing in coculture with PMOs resulted in a significant modulation of key bone markers such as PTHrP and OPG. Innovation and Conclusion: We here demonstrate the direct implications of HO-1 expression in bone remodeling and how it participates in the alterations in the communication between bone and prostate tumor cells.


Asunto(s)
Neoplasias Óseas/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Neoplasias Óseas/secundario , Regeneración Ósea , Remodelación Ósea , Hemo-Oxigenasa 1/deficiencia , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
9.
Sci Transl Med ; 11(494)2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142679

RESUMEN

Osteolytic lesions in multiple myeloma are caused by osteoclast-mediated bone resorption and reduced bone formation. A unique feature of myeloma is a failure of bone healing after successful treatment. We observed adipocytes on trabecular bone near the resorbed area in successfully treated patients. Normal marrow adipocytes, when cocultured with myeloma cells, were reprogrammed and produced adipokines that activate osteoclastogenesis and suppress osteoblastogenesis. These adipocytes have reduced expression of peroxisome proliferator-activated receptor γ (PPARγ) mediated by recruitment of polycomb repressive complex 2 (PRC2), which modifies PPARγ promoter methylation at trimethyl lysine-27 histone H3. We confirmed the importance of methylation in the PPARγ promoter by demonstrating that adipocyte-specific knockout of EZH2, a member of the PRC2, prevents adipocyte reprogramming and reverses bone changes in a mouse model. We validated the strong correlation between the frequency of bone lesions and the expression of EZH2 in marrow adipocytes from patients in remission. These results define a role for adipocytes in genesis of myeloma-associated bone disease and that reversal of adipocyte reprogramming has therapeutic implications.


Asunto(s)
Adipocitos/patología , Enfermedades Óseas/patología , Médula Ósea/patología , Reprogramación Celular , Mieloma Múltiple/patología , Adipoquinas/metabolismo , Animales , Resorción Ósea/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/metabolismo , Humanos , Metilación , Ratones , Osteoblastos/patología , Osteogénesis , PPAR gamma/genética , PPAR gamma/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas/genética , Inducción de Remisión , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba/genética
10.
Sci Transl Med ; 10(452)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068572

RESUMEN

Intravital multiphoton microscopy (iMPM) in mice provides access to cellular and molecular mechanisms of metastatic progression of cancers and the underlying interactions with the tumor stroma. Whereas iMPM of malignant disease has been performed for soft tissues, noninvasive iMPM of solid tumor in the bone is lacking. We combined miniaturized tissue-engineered bone constructs in nude mice with a skin window to noninvasively and repetitively monitor prostate cancer lesions by three-dimensional iMPM. In vivo ossicles developed large central cavities containing mature bone marrow surrounded by a thin cortex and enabled tumor implantation and longitudinal iMPM over weeks. Tumors grew inside the bone cavity and along the cortical bone interface and induced niches of osteoclast activation (focal osteolysis). Interventional bisphosphonate therapy reduced osteoclast kinetics and osteolysis without perturbing tumor growth, indicating dissociation of the tumor-stroma axis. The ossicle window, with its high cavity-to-cortex ratio and long-term functionality, thus allows for the mechanistic dissection of reciprocal epithelial tumor-bone interactions and therapy response.


Asunto(s)
Neoplasias Óseas/terapia , Progresión de la Enfermedad , Microscopía Intravital/métodos , Osteólisis/patología , Animales , Médula Ósea/irrigación sanguínea , Médula Ósea/patología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Catepsina K/metabolismo , Línea Celular Tumoral , Difosfonatos/farmacología , Difosfonatos/uso terapéutico , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Miniaturización , Células del Estroma/patología , Ingeniería de Tejidos , Andamios del Tejido/química , Resultado del Tratamiento , Ácido Zoledrónico/farmacología , Ácido Zoledrónico/uso terapéutico
11.
Sci Transl Med ; 8(353): 353ra113, 2016 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-27559096

RESUMEN

Myelomatous bone disease is characterized by the development of lytic bone lesions and a concomitant reduction in bone formation, leading to chronic bone pain and fractures. To understand the underlying mechanism, we investigated the contribution of myeloma-expressed thymidine phosphorylase (TP) to bone lesions. In osteoblast progenitors, TP up-regulated the methylation of RUNX2 and osterix, leading to decreased bone formation. In osteoclast progenitors, TP up-regulated the methylation of IRF8 and thereby enhanced expression of NFATc1 (nuclear factor of activated T cells, cytoplasmic 1 protein), leading to increased bone resorption. TP reversibly catalyzes thymidine into thymine and 2-deoxy-d-ribose (2DDR). Myeloma-secreted 2DDR bound to integrin αVß3/α5ß1 in the progenitors, activated PI3K (phosphoinositide 3-kinase)/Akt signaling, and increased DNMT3A (DNA methyltransferase 3A) expression, resulting in hypermethylation of RUNX2, osterix, and IRF8 This study elucidates an important mechanism for myeloma-induced bone lesions, suggesting that targeting TP may be a viable approach to healing resorbed bone in patients. Because TP overexpression is common in bone-metastatic tumors, our findings could have additional mechanistic implications.


Asunto(s)
Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Resorción Ósea/enzimología , Resorción Ósea/patología , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Osteogénesis/fisiología , Timidina Fosforilasa/metabolismo , Neoplasias Óseas/fisiopatología , Resorción Ósea/fisiopatología , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Regulación hacia Abajo , Humanos , Factores Reguladores del Interferón/genética , Mieloma Múltiple/fisiopatología , Osteoblastos/patología , Osteoblastos/fisiología , Osteoclastos/patología , Osteoclastos/fisiología , Osteólisis/enzimología , Osteólisis/patología , Osteólisis/prevención & control , Ligando RANK/metabolismo , Factor de Transcripción Sp7/genética , Timidina Fosforilasa/antagonistas & inhibidores , Regulación hacia Arriba
12.
Sci Transl Med ; 6(252): 252ra122, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25186177

RESUMEN

Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell-bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Quinolonas/uso terapéutico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bencimidazoles/farmacología , Neoplasias Óseas/patología , Huesos/efectos de los fármacos , Huesos/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Quinolonas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Bone ; 50(3): 695-703, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22173053

RESUMEN

Transforming growth factor beta 1 (TGF-ß1) has been implicated in the pathogenesis of prostate cancer (PCa) bone metastasis. In this study, we tested the antitumor efficacy of a selective TGF-ß receptor I kinase inhibitor, LY2109761, in preclinical models. The effect of LY2109761 on the growth of MDA PCa 2b and PC-3 human PCa cells and primary mouse osteoblasts (PMOs) was assessed in vitro by measuring radiolabeled thymidine incorporation into DNA. In vivo, the right femurs of male SCID mice were injected with PCa cells. We monitored the tumor burden in control- and LY2109761-treated mice with MRI analysis and the PCa-induced bone response with X-ray and micro-CT analyses. Histologic changes in bone were studied by performing bone histomorphometric evaluations. PCa cells and PMOs expressed TGF-ß receptor I. TGF-ß1 induced pathway activation (as assessed by induced expression of p-Smad2) and inhibited cell growth in PC-3 cells and PMOs but not in MDA PCa 2b cells. LY2109761 had no effect on PCa cells but induced PMO proliferation in vitro. As expected, LY2109761 reversed the TGF-ß1-induced pathway activation and growth inhibition in PC-3 cells and PMOs. In vivo, LY2109761 treatment for 6weeks resulted in increased volume in normal bone and increased osteoblast and osteoclast parameters. In addition, LY2109761 treatment significantly inhibited the growth of MDA PCa 2b and PC-3 in the bone of SCID mice (p<0.05); moreover, it resulted in significantly less bone loss and change in osteoclast-associated parameters in the PC-3 tumor-bearing bones than in the untreated mice. In summary, we report for the first time that targeting TGF-ß receptors with LY2109761 can control PCa bone growth while increasing the mass of normal bone. This increased bone mass in nontumorous bone may be a desirable side effect of LY2109761 treatment for men with osteopenia or osteoporosis secondary to androgen-ablation therapy, reinforcing the benefit of effectively controlling PCa growth in bone. Thus, targeting TGF-ß receptor I is a valuable intervention in men with advanced PCa.


Asunto(s)
Antineoplásicos/farmacología , Huesos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirroles/farmacología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Neoplasias Óseas/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fémur/efectos de los fármacos , Fémur/metabolismo , Fémur/patología , Humanos , Masculino , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Cancer Res ; 18(3): 726-36, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22298898

RESUMEN

PURPOSE: To study Wnt/ß-catenin in castrate-resistant prostate cancer (CRPC) and understand its function independently of the ß-catenin-androgen receptor (AR) interaction. EXPERIMENTAL DESIGN: We carried out ß-catenin immunocytochemical analysis, evaluated TOP-flash reporter activity (a reporter of ß-catenin-mediated transcription), and sequenced the ß-catenin gene in MDA prostate cancer 118a, MDA prostate cancer 118b, MDA prostate cancer 2b, and PC-3 prostate cancer cells. We knocked down ß-catenin in AR-negative MDA prostate cancer 118b cells and carried out comparative gene-array analysis. We also immunohistochemically analyzed ß-catenin and AR in 27 bone metastases of human CRPCs. RESULTS: ß-Catenin nuclear accumulation and TOP-flash reporter activity were high in MDA prostate cancer 118b but not in MDA prostate cancer 2b or PC-3 cells. MDA prostate cancer 118a and MDA prostate cancer 118b cells carry a mutated ß-catenin at codon 32 (D32G). Ten genes were expressed differently (false discovery rate, 0.05) in MDA prostate cancer 118b cells with downregulated ß-catenin. One such gene, hyaluronan synthase 2 (HAS2), synthesizes hyaluronan, a core component of the extracellular matrix. We confirmed HAS2 upregulation in PC-3 cells transfected with D32G-mutant ß-catenin. Finally, we found nuclear localization of ß-catenin in 10 of 27 human tissue specimens; this localization was inversely associated with AR expression (P = 0.056, Fisher's exact test), suggesting that reduced AR expression enables Wnt/ß-catenin signaling. CONCLUSION: We identified a previously unknown downstream target of ß-catenin, HAS2, in prostate cancer, and found that high ß-catenin nuclear localization and low or no AR expression may define a subpopulation of men with bone metastatic prostate cancer. These findings may guide physicians in managing these patients.


Asunto(s)
Glucuronosiltransferasa/genética , Neoplasias de la Próstata/genética , Transducción de Señal/fisiología , beta Catenina/genética , Animales , Western Blotting , Neoplasias Óseas/secundario , Perfilación de la Expresión Génica , Glucuronosiltransferasa/metabolismo , Humanos , Hialuronano Sintasas , Inmunohistoquímica , Masculino , Ratones , Ratones SCID , Mutación , Neoplasias de la Próstata/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , beta Catenina/metabolismo
15.
Clin Cancer Res ; 17(8): 2328-38, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21343372

RESUMEN

PURPOSE: A hallmark of prostate cancer (PCa) progression is the development of osteoblastic bone metastases, which respond poorly to available therapies. We previously reported that VEGF(121)/rGel targets osteoclast precursors and tumor neovasculature. Here we tested the hypothesis that targeting nontumor cells expressing these receptors can inhibit tumor progression in a clinically relevant model of osteoblastic PCa. EXPERIMENTAL DESIGN: Cells from MDA PCa 118b, a PCa xenograft obtained from a bone metastasis in a patient with castrate-resistant PCa, were injected into the femurs of mice. Osteoblastic progression was monitored following systemic administration of VEGF(121)/rGel. RESULTS: VEGF(121)/rGel was cytotoxic in vitro to osteoblast precursor cells. This cytotoxicity was specific as VEGF(121)/rGel internalization into osteoblasts was VEGF(121) receptor driven. Furthermore, VEGF(121)/rGel significantly inhibited PCa-induced bone formation in a mouse calvaria culture assay. In vivo, VEGF(121)/rGel significantly inhibited the osteoblastic progression of PCa cells in the femurs of nude mice. Microcomputed tomographic analysis revealed that VEGF(121)/rGel restored the bone volume fraction of tumor-bearing femurs to values similar to those of the contralateral (non-tumor-bearing) femurs. VEGF(121)/rGel significantly reduced the number of tumor-associated osteoclasts but did not change the numbers of peritumoral osteoblasts. Importantly, VEGF(121)/rGel-treated mice had significantly less tumor burden than control mice. Our results thus indicate that VEGF(121)/rGel inhibits osteoblastic tumor progression by targeting angiogenesis, osteoclastogenesis, and bone formation. CONCLUSIONS: Targeting VEGF receptor (VEGFR)-1- or VEGFR-2-expressing cells is effective in controlling the osteoblastic progression of PCa in bone. These findings provide the basis for an effective multitargeted approach for metastatic PCa.


Asunto(s)
Neovascularización Patológica/prevención & control , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Línea Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Técnicas de Cultivo de Órganos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Biol Ther ; 8(22): 2153-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19855158

RESUMEN

PURPOSE: Therapies to target prostate cancer bone metastases have only limited effects. New treatments are focused on the interaction between cancer cells, bone marrow cells and the bone matrix. Osteoclasts play an important role in the development of bone tumors caused by prostate cancer. Since Src kinase has been shown to be necessary for osteoclast function, we hypothesized that dasatinib, a Src family kinase inhibitor, would reduce osteoclast activity and prostate cancer (PC-3) cell-induced osteoclast formation. RESULTS: Dasatinib inhibited RANKL-induced osteoclast differentiation of bone marrow-derived monocytes with an EC(50) of 7.5 nM. PC-3 cells, a human prostate cancer cell line, were able to differentiate RAW 264.7 cells, a murine monocytic cell line, into osteoclasts, and dasatinib inhibited this differentiation. In addition, conditioned medium from PC-3 cell cultures was able to differentiate RAW 264.7 cells into osteoclasts and this too, was inhibited by dasatinib. Even the lowest concentration of dasatinib, 1.25 nmol, inhibited osteoclast differentiation by 29%. Moreover, dasatinib inhibited osteoclast activity by 58% as measured by collagen 1 release. EXPERIMENTAL DESIGN: We performed in vitro experiments utilizing the Src family kinase inhibitor dasatinib to target osteoclast activation as a means of inhibiting prostate cancer bone metastases. CONCLUSION: Dasatinib inhibits osteoclast differentiation of mouse primary bone marrow-derived monocytes and PC-3 cell-induced osteoclast differentiation. Dasatinib also inhibits osteoclast degradation activity. Inhibiting osteoclast differentiation and activity may be an effective targeted therapy in patients with prostate cancer bone metastases.


Asunto(s)
Adenocarcinoma/secundario , Neoplasias Óseas/secundario , Proteínas de Neoplasias/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Neoplasias de la Próstata/patología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral/citología , Línea Celular Tumoral/efectos de los fármacos , Colágeno/metabolismo , Dasatinib , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/metabolismo , Ligando RANK/antagonistas & inhibidores
17.
Dev Dyn ; 224(2): 245-51, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12112477

RESUMEN

Cell- and time-specific gene inactivation should enhance our knowledge of bone biology. Implementation of this technique requires construction of transgenic mouse lines expressing Cre recombinase in osteoblasts, the bone forming cell. We tested several promoter fragments for their ability to drive efficient Cre expression in osteoblasts. In the first mouse transgenic line, the Cre gene was placed under the control of the 2.3-kb proximal fragment of the alpha1(I)-collagen promoter, which is expressed at high levels in osteoblasts throughout their differentiation. Transgenic mice expressing this transgene in bone were bred with the ROSA26 reporter (R26R) strain in which the ROSA26 locus is targeted with a conditional LacZ reporter cassette. In R26R mice, Cre expression and subsequent Cre-mediated recombination lead to expression of the LacZ reporter gene, an event that can be monitored by LacZ staining. LacZ staining was detected in virtually all osteoblasts of alpha1(I)-Cre;R26R mice indicating that homologous recombination occurred in these cells. No other cell type stained blue. In the second line studied, the 1.3-kb fragment of osteocalcin gene 2 (OG2) promoter, which is active in differentiated osteoblasts, was used to drive Cre expression. OG2-Cre mice expressed Cre specifically in bone. However, cross of OG2-Cre mice with R26R mice did not lead to any detectable LacZ staining in osteoblasts. Lastly, we tested a more active artificial promoter derived from the OG2 promoter. The artificial OG2-Cre transgene was expressed by reverse transcriptase-polymerase chain reaction in cartilage and bone samples. After cross of the artificial OG2-Cre mice with R26R mice, we detected a LacZ staining in articular chondrocytes but not in osteoblasts. Our data suggest that the only promoter able to drive Cre expression at a level sufficient to induce recombination in osteoblasts is the alpha1(I)-collagen promoter.


Asunto(s)
Colágeno Tipo I/genética , Técnicas de Transferencia de Gen , Integrasas/genética , Regiones Promotoras Genéticas , Proteínas Virales/genética , Animales , Células Cultivadas , Genes Reporteros , Integrasas/metabolismo , Operón Lac , Ratones , Ratones Transgénicos , Modelos Genéticos , Osteoblastos/metabolismo , Plásmidos/metabolismo , Recombinación Genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular , Transfección , Proteínas Virales/metabolismo
18.
Development ; 129(12): 2977-86, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12050144

RESUMEN

In developing murine growth plates, chondrocytes near the articular surface (periarticular chondrocytes) proliferate, differentiate into flat column-forming proliferating cells (columnar chondrocytes), stop dividing and finally differentiate into hypertrophic cells. Indian hedgehog (Ihh), which is predominantly expressed in prehypertrophic cells, stimulates expression of parathyroid hormone (PTH)-related peptide (PTHrP) which negatively regulates terminal chondrocyte differentiation through the PTH/PTHrP receptor (PPR). However, the roles of PTHrP and Ihh in regulating earlier steps in chondrocyte differentiation are unclear. We present novel mouse models with PPR abnormalities that help clarify these roles. In mice with chondrocyte-specific PPR ablation and mice with reduced PPR expression, chondrocyte differentiation was accelerated not only at the terminal step but also at an earlier step: periarticular to columnar differentiation. In these models, upregulation of Ihh action in the periarticular region was also observed. In the third model in which the PPR was disrupted in about 30% of columnar chondrocytes, Ihh action in the periarticular chondrocytes was upregulated because of ectopically differentiated hypertrophic chondrocytes that had lost PPR. Acceleration of periarticular to columnar differentiation was also noted in this mouse, while most of periarticular chondrocytes retained PPR signaling. These data suggest that Ihh positively controls differentiation of periarticular chondrocytes independently of PTHrP. Thus, chondrocyte differentiation is controlled at multiple steps by PTHrP and Ihh through the mutual regulation of their activities.


Asunto(s)
Diferenciación Celular/genética , Condrocitos/fisiología , Placa de Crecimiento/embriología , Proteínas/metabolismo , Transactivadores/metabolismo , Animales , Inducción Embrionaria/genética , Regulación del Desarrollo de la Expresión Génica , Ingeniería Genética/métodos , Placa de Crecimiento/citología , Proteínas Hedgehog , Ratones , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea , Proteínas/genética , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Transducción de Señal , Transactivadores/genética , Regulación hacia Arriba
19.
J Biol Chem ; 279(26): 27560-6, 2004 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-15090551

RESUMEN

Bone morphogenetic proteins (BMPs) function during various aspects of embryonic development including skeletogenesis. However, their biological functions after birth are less understood. To investigate the role of BMPs during bone remodeling, we generated a postnatal osteoblast-specific disruption of Bmpr1a that encodes the type IA receptor for BMPs in mice. Mutant mice were smaller than controls up to 6 months after birth. Irregular calcification and low bone mass were observed, but there were normal numbers of osteoblasts. The ability of the mutant osteoblasts to form mineralized nodules in culture was severely reduced. Interestingly, bone mass was increased in aged mutant mice due to reduced bone resorption evidenced by reduced bone turnover. The mutant mice lost more bone after ovariectomy likely resulting from decreased osteoblast function which could not overcome ovariectomy-induced bone resorption. In organ culture of bones from aged mice, ablation of the Bmpr1a gene by adenoviral Cre recombinase abolished the stimulatory effects of BMP4 on the expression of lysosomal enzymes essential for osteoclastic bone resorption. These results demonstrate essential and age-dependent roles for BMP signaling mediated by BMPRIA (a type IA receptor for BMP) in osteoblasts for bone remodeling.


Asunto(s)
Remodelación Ósea/fisiología , Osteoblastos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Peso Corporal , Desarrollo Óseo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Resorción Ósea/metabolismo , Calcificación Fisiológica , Diferenciación Celular , Femenino , Integrasas/genética , Integrasas/metabolismo , Lisosomas/enzimología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoblastos/ultraestructura , Osteoclastos/metabolismo , Ovariectomía , Fenotipo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento/deficiencia , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA