Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 101(3): 797-855, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33356915

RESUMEN

Skeletal metastases are frequent complications of many cancers, causing bone complications (fractures, bone pain, disability) that negatively affect the patient's quality of life. Here, we first discuss the burden of skeletal complications in cancer bone metastasis. We then describe the pathophysiology of bone metastasis. Bone metastasis is a multistage process: long before the development of clinically detectable metastases, circulating tumor cells settle and enter a dormant state in normal vascular and endosteal niches present in the bone marrow, which provide immediate attachment and shelter, and only become active years later as they proliferate and alter the functions of bone-resorbing (osteoclasts) and bone-forming (osteoblasts) cells, promoting skeletal destruction. The molecular mechanisms involved in mediating each of these steps are described, and we also explain how tumor cells interact with a myriad of interconnected cell populations in the bone marrow, including a rich vascular network, immune cells, adipocytes, and nerves. We discuss metabolic programs that tumor cells could engage with to specifically grow in bone. We also describe the progress and future directions of existing bone-targeted agents and report emerging therapies that have arisen from recent advances in our understanding of the pathophysiology of bone metastases. Finally, we discuss the value of bone turnover biomarkers in detection and monitoring of progression and therapeutic effects in patients with bone metastasis.


Asunto(s)
Neoplasias Óseas/secundario , Huesos/patología , Animales , Biomarcadores/metabolismo , Conservadores de la Densidad Ósea/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Huesos/metabolismo , Denosumab/uso terapéutico , Humanos
2.
Int J Mol Sci ; 22(4)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671469

RESUMEN

Bone metastasis remains the most frequent and the deadliest complication of prostate cancer (PCa). Mechanisms leading to the homing of tumor cells to bone remain poorly characterized. Role of chemokines in providing navigational cues to migrating cancer cells bearing specific receptors is well established. Bone is an adipocyte-rich organ since 50 to 70% of the adult bone marrow (BM) volume comprise bone marrow adipocytes (BM-Ads), which are likely to produce chemokines within the bone microenvironment. Using in vitro migration assays, we demonstrated that soluble factors released by human primary BM-Ads are able to support the directed migration of PCa cells in a CCR3-dependent manner. In addition, we showed that CCL7, a chemokine previously involved in the CCR3-dependent migration of PCa cells outside of the prostate gland, is released by human BM-Ads. These effects are amplified by obesity and ageing, two clinical conditions known to promote aggressive and metastatic PCa. In human tumors, we found an enrichment of CCR3 in bone metastasis vs. primary tumors at mRNA levels using Oncomine microarray database. In addition, immunohistochemistry experiments demonstrated overexpression of CCR3 in bone versus visceral metastases. These results underline the potential importance of BM-Ads in the bone metastatic process and imply a CCR3/CCL7 axis whose pharmacological interest needs to be evaluated.


Asunto(s)
Adipocitos/metabolismo , Adipocitos/patología , Médula Ósea/patología , Huesos/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores CCR3/metabolismo , Envejecimiento/patología , Médula Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Línea Celular Tumoral , Quimiocina CCL7/metabolismo , Quimiotaxis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Humanos , Masculino , Metástasis de la Neoplasia , Obesidad/complicaciones , Neoplasias de la Próstata/complicaciones
3.
Calcif Tissue Int ; 103(5): 567-580, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29916127

RESUMEN

Periostin is an extracellular matrix protein that actively contributes to tumor progression and metastasis. Here, we hypothesized that it could be a marker of bone metastasis formation. To address this question, we used two polyclonal antibodies directed against the whole molecule or its C-terminal domain to explore the expression of intact and truncated forms of periostin in the serum and tissues (lung, heart, bone) of wild-type and periostin-deficient mice. In normal bones, periostin was expressed in the periosteum and specific periostin proteolytic fragments were found in bones, but not in soft tissues. In animals bearing osteolytic lesions caused by 4T1 cells, C-terminal intact periostin (iPTN) expression disappeared at the invasive front of skeletal tumors where bone-resorbing osteoclasts were present. In vitro, we found that periostin was a substrate for osteoclast-derived cathepsin K, generating proteolytic fragments that were not recognized by anti-periostin antibodies directed against iPTN. In vivo, using an in-house sandwich immunoassay aimed at detecting iPTN only, we observed a noticeable reduction of serum periostin levels (- 26%; P < 0.002) in animals bearing osteolytic lesions caused by 4T1 cells. On the contrary, this decrease was not observed in women with breast cancer and bone metastases when periostin was measured with a human assay detecting total periostin. Collectively, these data showed that mouse periostin was degraded at the bone metastatic sites, potentially by cathepsin K, and that the specific measurement of iPTN in serum should assist in detecting bone metastasis formation in breast cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/sangre , Osteólisis/diagnóstico , Adulto , Anciano , Animales , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Persona de Mediana Edad
4.
J Biol Chem ; 289(10): 6551-6564, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24429286

RESUMEN

Lysophosphatidic acid (LPA) is a natural bioactive lipid that acts through six different G protein-coupled receptors (LPA1-6) with pleiotropic activities on multiple cell types. We have previously demonstrated that LPA is necessary for successful in vitro osteoclastogenesis of bone marrow cells. Bone cells controlling bone remodeling (i.e. osteoblasts, osteoclasts, and osteocytes) express LPA1, but delineating the role of this receptor in bone remodeling is still pending. Despite Lpar1(-/-) mice displaying a low bone mass phenotype, we demonstrated that bone marrow cell-induced osteoclastogenesis was reduced in Lpar1(-/-) mice but not in Lpar2(-/-) and Lpar3(-/-) animals. Expression of LPA1 was up-regulated during osteoclastogenesis, and LPA1 antagonists (Ki16425, Debio0719, and VPC12249) inhibited osteoclast differentiation. Blocking LPA1 activity with Ki16425 inhibited expression of nuclear factor of activated T-cell cytoplasmic 1 (NFATc1) and dendritic cell-specific transmembrane protein and interfered with the fusion but not the proliferation of osteoclast precursors. Similar to wild type osteoclasts treated with Ki16425, mature Lpar1(-/-) osteoclasts had reduced podosome belt and sealing zone resulting in reduced mineralized matrix resorption. Additionally, LPA1 expression markedly increased in the bone of ovariectomized mice, which was blocked by bisphosphonate treatment. Conversely, systemic treatment with Debio0719 prevented ovariectomy-induced cancellous bone loss. Moreover, intravital multiphoton microscopy revealed that Debio0719 reduced the retention of CX3CR1-EGFP(+) osteoclast precursors in bone by increasing their mobility in the bone marrow cavity. Overall, our results demonstrate that LPA1 is essential for in vitro and in vivo osteoclast activities. Therefore, LPA1 emerges as a new target for the treatment of diseases associated with excess bone loss.


Asunto(s)
Resorción Ósea/patología , Proteínas de la Membrana/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Osteoclastos/patología , Receptores del Ácido Lisofosfatídico/fisiología , Animales , Células de la Médula Ósea/patología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/genética , Diferenciación Celular/efectos de los fármacos , Movimiento Celular , Femenino , Isoxazoles/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ácidos Oléicos/farmacología , Organofosfatos/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Propionatos/farmacología , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/genética
5.
Arthritis Rheum ; 63(8): 2374-84, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21506092

RESUMEN

OBJECTIVE: We reported previously that the orphan nuclear receptor, estrogen receptor-related receptor α (ERRα), is expressed in articular chondrocytes and is dysregulated in a mouse model of inflammatory arthritis. The aim of this study, therefore, was to determine whether ERRα is also dysregulated in patients with osteoarthritis (OA). METHODS: ERRα messenger RNA (mRNA) and protein were quantified in normal and OA cartilage samples and in OA chondrocytes in vitro, with and without short-term treatment with a variety of OA-associated factors and signaling pathway agonists and inhibitors. RESULTS: ERRα expression was lower in OA than in normal articular cartilage. Interleukin-1ß (IL-1ß) markedly up-regulated ERRα expression in OA chondrocytes in vitro, and agonist or inhibitor treatment indicated that the up-regulation was dependent on cyclooxygenase 2 (COX-2; NS398), prostaglandin E(2), cAMP (8-bromo-cAMP), and protein kinase A (PKA; KT5720). Treatment with the ERRα inverse agonist XCT790 decreased the expression of SOX9 and the up-regulation of ERRα by IL-1ß, suggesting autoregulation of ERRα in the IL-1ß pathway. Matrix metalloproteinase 13 (MMP-13) expression was also decreased by treatment with XCT790 plus IL-1ß versus IL-1ß alone, and the down-regulation of MMP-13 mRNA and protein observed with XCT790 alone suggests that the up-regulation of MMP-13 by IL-1ß is ERRα-dependent. CONCLUSION: We report the first evidence that ERRα expression is regulated by IL-1ß in COX-2-, cAMP-, and PKA-dependent pathways in OA chondrocytes. We confirmed that SOX9 is an ERRα target gene in human, as in rodent, chondrocytes and identified MMP-13 as a potential new target gene, which suggests that ERRα may both respond to the healing signal and contribute to extracellular degradation in OA cartilage.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , AMP Cíclico/metabolismo , Interleucina-1beta/metabolismo , Osteoartritis/metabolismo , Prostaglandinas E/metabolismo , Receptores de Estrógenos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , AMP Cíclico/genética , Femenino , Humanos , Interleucina-1beta/genética , Masculino , Persona de Mediana Edad , Osteoartritis/genética , Prostaglandinas E/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/genética , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/fisiología , Receptor Relacionado con Estrógeno ERRalfa
6.
Front Immunol ; 13: 899104, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677054

RESUMEN

Bone loss associated with estrogen deficiency indicates a fundamental role of these hormones in skeletal growth and bone remodeling. In the last decades, growing recent evidence demonstrated that estrogens can also affect the immune compartment of the bone. In this review, we summarize the impacts of estrogens on bone immune cells and their consequences on bone homeostasis, metastasis settlement into the bone and tumor progression. We also addressed the role of an orphan nuclear receptor ERRalpha ("Estrogen-receptor Related Receptor alpha") on macrophages and T lymphocytes, and as an immunomodulator in bone metastases. Hence, this review links estrogens to bone immune cells in osteo-oncology.


Asunto(s)
Neoplasias Óseas , Estrógenos , Remodelación Ósea , Huesos , Humanos , Factores Inmunológicos
7.
Cancers (Basel) ; 13(17)2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34503299

RESUMEN

This Special Issue of Cancers covers different aspects of bone physiopathology in oncology that combine the microenvironment and the factors involved in bone metastasis dormancy and progression [...].

8.
JBMR Plus ; 5(6): e10496, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34189385

RESUMEN

The current paradigm of osteoblast fate is that the majority undergo apoptosis, while some further differentiate into osteocytes and others flatten and cover bone surfaces as bone lining cells. Osteoblasts have been described to exhibit heterogeneous expression of a variety of osteoblast markers at both transcriptional and protein levels. To explore further this heterogeneity and its biological significance, Venus-positive (Venus+) cells expressing the fluorescent protein Venus under the control of the 2.3-kb Col1a1 promoter were isolated from newborn mouse calvariae and subjected to single-cell RNA sequencing. Functional annotation of the genes expressed in 272 Venus+ single cells indicated that Venus+ cells are osteoblasts that can be categorized into four clusters. Of these, three clusters (clusters 1 to 3) exhibited similarities in their expression of osteoblast markers, while one (cluster 4) was distinctly different. We identified a total of 1920 cluster-specific genes and pseudotime ordering analyses based on established concepts and known markers showed that clusters 1 to 3 captured osteoblasts at different maturational stages. Analysis of gene co-expression networks showed that genes involved in protein synthesis and protein trafficking between endoplasmic reticulum (ER) and Golgi are active in these clusters. However, the cells in these clusters were also defined by extensive heterogeneity of gene expression, independently of maturational stage. Cells of cluster 4 expressed Cd34 and Cxcl12 with relatively lower levels of osteoblast markers, suggesting that this cell type differs from actively bone-forming osteoblasts and retain or reacquire progenitor properties. Based on expression and machine learning analyses of the transcriptomes of individual osteoblasts, we also identified genes that may be useful as new markers of osteoblast maturational stages. Taken together, our data show much more extensive heterogeneity of osteoblasts than previously documented, with gene profiles supporting diversity of osteoblast functional activities and developmental fates. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

9.
Oncogene ; 40(7): 1284-1299, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33420367

RESUMEN

Bone metastasis remains a major cause of mortality and morbidity in breast cancer. Therefore, there is an urgent need to better select high-risk patients in order to adapt patient's treatment and prevent bone recurrence. Here, we found that integrin alpha5 (ITGA5) was highly expressed in bone metastases, compared to lung, liver, or brain metastases. High ITGA5 expression in primary tumors correlated with the presence of disseminated tumor cells in bone marrow aspirates from early stage breast cancer patients (n = 268; p = 0.039). ITGA5 was also predictive of poor bone metastasis-free survival in two separate clinical data sets (n = 855, HR = 1.36, p = 0.018 and n = 427, HR = 1.62, p = 0.024). This prognostic value remained significant in multivariate analysis (p = 0.028). Experimentally, ITGA5 silencing impaired tumor cell adhesion to fibronectin, migration, and survival. ITGA5 silencing also reduced tumor cell colonization of the bone marrow and formation of osteolytic lesions in vivo. Conversely, ITGA5 overexpression promoted bone metastasis. Pharmacological inhibition of ITGA5 with humanized monoclonal antibody M200 (volociximab) recapitulated inhibitory effects of ITGA5 silencing on tumor cell functions in vitro and tumor cell colonization of the bone marrow in vivo. M200 also markedly reduced tumor outgrowth in experimental models of bone metastasis or tumorigenesis, and blunted cancer-associated bone destruction. ITGA5 was not only expressed by tumor cells but also osteoclasts. In this respect, M200 decreased human osteoclast-mediated bone resorption in vitro. Overall, this study identifies ITGA5 as a mediator of breast-to-bone metastasis and raises the possibility that volociximab/M200 could be repurposed for the treatment of ITGA5-positive breast cancer patients with bone metastases.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Integrinas/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Anciano , Anticuerpos Monoclonales/administración & dosificación , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Osteólisis/genética , Supervivencia sin Progresión
10.
Cancer Res ; 80(13): 2914-2926, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32366476

RESUMEN

Bone is the most common metastatic site for breast cancer. Although the estrogen-related receptor alpha (ERRα) has been implicated in breast cancer cell dissemination to the bone from the primary tumor, its role after tumor cell anchorage in the bone microenvironment remains elusive. Here, we reveal that ERRα inhibits the progression of bone metastases of breast cancer cells by increasing the immune activity of the bone microenvironment. Overexpression of ERRα in breast cancer bone metastases induced expression of chemokines CCL17 and CCL20 and repressed production of TGFß3. Subsequently, CD8+ T lymphocytes recruited to bone metastases escaped TGFß signaling control and were endowed with exacerbated cytotoxic features, resulting in significant reduction in metastases. The clinical relevance of our findings in mice was confirmed in over 240 patients with breast cancer. Thus, this study reveals that ERRα regulates immune properties in the bone microenvironment that contributes to decreasing metastatic growth. SIGNIFICANCE: This study places ERRα at the interplay between the immune response and bone metastases of breast cancer, highlighting a potential target for intervention in advanced disease.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/prevención & control , Neoplasias de la Mama/prevención & control , Receptores de Estrógenos/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Óseas/inmunología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Quimiocina CCL17/genética , Quimiocina CCL17/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Receptores de Estrógenos/genética , Transducción de Señal , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Receptor Relacionado con Estrógeno ERRalfa
11.
Commun Biol ; 3(1): 30, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949279

RESUMEN

Communication between osteoblasts and osteoclasts plays a key role in bone metabolism. We describe here an unexpected role for matrix vesicles (MVs), which bud from bone-forming osteoblasts and have a well-established role in initiation of bone mineralization, in osteoclastogenesis. We show that the MV cargo miR-125b accumulates in the bone matrix, with increased accumulation in transgenic (Tg) mice overexpressing miR-125b in osteoblasts. Bone formation and osteoblasts in Tg mice are normal, but the number of bone-resorbing osteoclasts is reduced, leading to higher trabecular bone mass. miR-125b in the bone matrix targets and degrades Prdm1, a transcriptional repressor of anti-osteoclastogenic factors, in osteoclast precursors. Overexpressing miR-125b in osteoblasts abrogates bone loss in different mouse models. Our results show that the MV cargo miR-125b is a regulatory element of osteoblast-osteoclast communication, and that bone matrix provides extracellular storage of miR-125b that is functionally active in bone resorption.


Asunto(s)
Matriz Ósea/metabolismo , Resorción Ósea/genética , Resorción Ósea/metabolismo , Vesículas Extracelulares/metabolismo , MicroARNs/genética , Animales , Transporte Biológico , Biomarcadores , Resorción Ósea/patología , Comunicación Celular , Regulación de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Transgénicos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Interferencia de ARN , Transducción de Señal
12.
Cancer Res ; 67(20): 9894-902, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17942921

RESUMEN

Osteoclasts mediate bone destruction in breast cancer skeletal metastases. Cathepsin K is a proteinase that is secreted by osteoclasts and degrades bone. Here, immunohistochemistry revealed that cathepsin K was expressed not only by osteoclasts but also by breast cancer cells that metastasize to bone. Following intratibial injection with cathepsin K-expressing human BT474 breast cancer cells, tumor-bearing mice treated with a clinical dosing regimen of cathepsin K inhibitor (CKI; 50 mg/kg, twice daily) had osteolytic lesions that were 79% smaller than those of tumor-bearing mice treated with the vehicle. The effect of CKI was also studied in a mouse model in which the i.v. inoculation of human B02 breast cancer cells expressing cathepsin K leads to bone metastasis formation. Drug administration was started before (preventive protocol) or after (treatment protocol) the occurrence of osteolytic lesions. In treatment protocols, CKI (50 mg/kg, twice daily) or a single clinical dose of 100 microg/kg zoledronic acid (osteoclast inhibitor) reduced the progression of osteolytic lesions by 59% to 66%. CKI therapy also reduced skeletal tumor burden by 62% compared with vehicle, whereas zoledronic acid did not decrease the tumor burden. The efficacy of CKI at inhibiting skeletal tumor burden was similar in the treatment and preventive protocols. By contrast, CKI did not block the growth of s.c. B02 tumor xenografts in animals. Thus, CKI may render the bone a less favorable microenvironment for tumor growth by inhibiting bone resorption. These findings raise the possibility that cathepsin K could be a therapeutic target for the treatment of bone metastases.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Catepsinas/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Animales , Neoplasias Óseas/enzimología , Neoplasias Óseas/prevención & control , Neoplasias de la Mama/patología , Catepsina K , Catepsinas/biosíntesis , Línea Celular Tumoral , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Osteólisis/tratamiento farmacológico , Osteólisis/enzimología , Osteólisis/prevención & control , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Eur J Cell Biol ; 87(8-9): 459-68, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18294724

RESUMEN

Osteoclasts are large monocyte-derived multinucleated cells whose function is to resorb bone, i.e. a mineralised extracellular matrix. They exhibit two different actin cytoskeleton organisations according to their substratum. On non-mineralised substrates they form canonical podosomes, but on mineralised extracellular matrices they form a sealing zone. Podosomes consist of two functionally different actin subdomains: a podosome core, probably made of branched actin organised through a CD44 transmembrane receptor, and an actin cloud of actin cables organised around alphavbeta3 integrin. During osteoclast differentiation, podosome patterning is highly dynamic, and we propose that it ends up in a sealing zone in mature bone-resorbing osteoclasts after a complete reorganisation of the two subdomains. In addition to matrix degradation, osteoclasts share with tumour cells the ability to transmigrate through cell layers and-for that purpose-can arrange their cytoskeleton in long protrusions reminiscent of invadopodia. In this review, we discuss the relationships between podosomes and sealing zone, comparing their structures, their molecular composition and their abilities to degrade extracellular matrices. The dynamic actin remodelling in osteoclasts appears then as a major factor to understand their unusual abilities reminiscent of metastatic tumour cells.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Matriz Extracelular/metabolismo , Osteoclastos/ultraestructura , Citoesqueleto de Actina/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Humanos , Modelos Biológicos , Osteoclastos/metabolismo
14.
Bone ; 42(1): 129-38, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17945546

RESUMEN

Strontium ranelate is a newly developed drug that has been shown to significantly reduce the risk of vertebral and non-vertebral fractures, including those of the hip, in postmenopausal women with osteoporosis. In contrast to other available treatments for osteoporosis, strontium ranelate increases bone formation and decreases resorption. In this study, the dual mode of action of strontium ranelate in bone was tested in vitro, on primary murine osteoblasts and osteoclasts derived from calvaria and spleen cells, respectively. We show that strontium ranelate treatment, either continuously or during proliferation or differentiation phases of mouse calvaria cells, stimulates osteoblast formation. Indeed after 22 days of continuous treatment with strontium ranelate, the expression of the osteoblast markers ALP, BSP and OCN was increased, and was combined with an increase in bone nodule numbers. On the other hand, the number of mature osteoclasts strongly decreased after strontium ranelate treatment. Similarly to previous studies, we confirm that osteoclasts resorbing activity was also reduced but we found that strontium ranelate treatment was associated with a disruption of the osteoclast actin-containing sealing zone. Therefore, our in vitro assays performed on primary murine bone cells confirmed the dual action of strontium ranelate in vivo as an anabolic agent on bone remodeling. It stimulates bone formation through its positive action on osteoblast differentiation and function, and decreases osteoclast differentiation as well as function by disrupting actin cytoskeleton organization.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Compuestos Organometálicos/farmacología , Compuestos Organometálicos/uso terapéutico , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Tiofenos/farmacología , Tiofenos/uso terapéutico , Animales , Biomarcadores , Resorción Ósea/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Ratones , Osteoclastos/metabolismo
15.
Cancer Lett ; 438: 32-43, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201302

RESUMEN

Prostate cancers have a strong propensity to metastasize to bone and promote osteoblastic lesions. TMPRSS2:ERG is the most frequent gene rearrangement identified in prostate cancer, but whether it is involved in prostate cancer bone metastases is largely unknown. We exploited an intratibial metastasis model to address this issue and we found that ectopic expression of the TMPRSS2:ERG fusion enhances the ability of prostate cancer cell lines to induce osteoblastic lesions by stimulating bone formation and inhibiting the osteolytic response. In line with these in vivo results, we demonstrate that the TMPRSS2:ERG fusion protein increases the expression of osteoblastic markers, including Collagen Type I Alpha 1 Chain and Alkaline Phosphatase, as well as Endothelin-1, a protein with a documented role in osteoblastic bone lesion formation. Moreover, we determined that the TMPRSS2:ERG fusion protein is bound to the regulatory regions of these genes in prostate cancer cell lines, and we report that the expression levels of these osteoblastic markers are correlated with the expression of the TMPRSS2:ERG fusion in patient metastasis samples. Taken together, our results reveal that the TMPRSS2:ERG gene fusion is involved in osteoblastic lesion formation induced by prostate cancer cells.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/genética , Osteoblastos/metabolismo , Neoplasias de la Próstata/genética , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Línea Celular Tumoral , Cadena alfa 1 del Colágeno Tipo I , Endotelina-1/genética , Endotelina-1/metabolismo , Humanos , Masculino , Ratones SCID , Proteínas de Fusión Oncogénica/metabolismo , Osteoblastos/patología , Células PC-3 , Fenotipo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Trasplante Heterólogo , Carga Tumoral/genética
16.
Bone ; 106: 187-193, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29051055

RESUMEN

INTRODUCTION: Pierson syndrome is caused by a mutation of LAMB2, encoding for laminin ß2. Clinical phenotype is variable but usually associates congenital nephrotic syndrome (CNS) and ocular abnormalities. Neuromuscular impairment has also been described. METHODS: We report on a 15-year old girl, suffering from Pierson Syndrome, who developed severe bone deformations during puberty. This patient initially displayed CNS and microcoria, leading to the clinical diagnosis of Pierson syndrome. Genetic analysis revealed a truncating mutation and a splice site mutation of LAMB2. The patient received a renal transplantation (R-Tx) at the age of 3. After R-Tx, renal evolution was simple, the patient receiving low-dose corticosteroids, tacrolimus and mycophenolate mofetil. At the age of 12, bone deformations progressively appeared. At the time of bone impairment, renal function was subnormal (glomerular filtration rate using iohexol clearance 50mL/min per 1.73m2), and parameters of calcium/phosphate metabolism were normal (calcium 2.45mmol/L, phosphorus 1.30mmol/L, PTH 81ng/L, ALP 334U/L, 25OH-D 73nmol/L). Radiographs showed major deformations such as scoliosis, genu varum and diffuse epiphyseal abnormalities. A high resolution scanner (HR-pQCT) was performed, demonstrating a bone of "normal low" quantity and quality; major radial and cubital deformations were observed. Stainings of laminin ß2 were performed on bone and renal samples from the patient and healthy controls: as expected, laminin ß2 was expressed in the control kidney but not in the patient's renal tissue, and a similar pattern was observed in bone. CONCLUSION: This is the first case of skeletal impairment ever described in Pierson syndrome. Integrin α3ß1, receptor for laminin ß2, are found in podocytes and osteoblasts, and the observation of both the presence of laminin ß2 staining in healthy bone and its absence in the patient's bone raises the question of a potential role of laminin ß2 in bone physiology.


Asunto(s)
Anomalías Múltiples/metabolismo , Anomalías Múltiples/fisiopatología , Anomalías del Ojo/metabolismo , Anomalías del Ojo/fisiopatología , Laminina/metabolismo , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/fisiopatología , Trastornos de la Pupila/metabolismo , Trastornos de la Pupila/fisiopatología , Anomalías Múltiples/genética , Adolescente , Anomalías del Ojo/genética , Femenino , Humanos , Laminina/genética , Mutación , Síndromes Miasténicos Congénitos , Síndrome Nefrótico/genética , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/fisiopatología , Trastornos de la Pupila/genética
17.
Cancer Res ; 78(18): 5259-5273, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30042152

RESUMEN

miRNAs are master regulators of gene expression that play key roles in cancer metastasis. During bone metastasis, metastatic tumor cells must rewire their biology and express genes that are normally expressed by bone cells (a process called osteomimicry), which endow tumor cells with full competence for outgrowth in the bone marrow. Here, we establish miR-30 family members miR-30a, miR-30b, miR-30c, miR-30d, and miR-30e as suppressors of breast cancer bone metastasis that regulate multiple pathways, including osteomimicry. Low expression of miR-30 in primary tumors from patients with breast cancer were associated with poor relapse-free survival. In addition, estrogen receptor (ER)-negative/progesterone receptor (PR)-negative breast cancer cells expressed lower miR-30 levels than their ER/PR-positive counterparts. Overexpression of miR-30 in ER/PR-negative breast cancer cells resulted in the reduction of bone metastasis burden in vivoIn vitro, miR-30 did not affect tumor cell proliferation, but did inhibit tumor cell invasion. Furthermore, overexpression of miR-30 restored bone homeostasis by reversing the effects of tumor cell-conditioned medium on osteoclastogenesis and osteoblastogenesis. A number of genes associated with osteoclastogenesis stimulation (IL8, IL11), osteoblastogenesis inhibition (DKK-1), tumor cell osteomimicry (RUNX2, CDH11), and invasiveness (CTGF, ITGA5, ITGB3) were identified as targets for repression by miR-30. Among these genes, silencing CDH11 or ITGA5 in ER-/PR-negative breast cancer cells recapitulated inhibitory effects of miR-30 on skeletal tumor burden in vivo Overall, our findings provide evidence that miR-30 family members employ multiple mechanisms to impede breast cancer bone metastasis and may represent attractive targets for therapeutic intervention.Significance: These findings suggest miR-30 family members may serve as an effective means to therapeutically attenuate metastasis in triple-negative breast cancer. Cancer Res; 78(18); 5259-73. ©2018 AACR.


Asunto(s)
Neoplasias Óseas/metabolismo , Huesos/patología , Neoplasias de la Mama/metabolismo , MicroARNs/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Células 3T3 , Animales , Médula Ósea/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Integrina beta3/metabolismo , Integrinas/metabolismo , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Osteoblastos/metabolismo , Neoplasias de la Mama Triple Negativas/patología
18.
Cancer Res ; 77(2): 268-278, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27742687

RESUMEN

Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase whose primary function is to drive collagen crosslinking and extracellular matrix stiffness. LOX in colorectal cancer synergizes with hypoxia-inducible factor-1 (HIF-1) to promote tumor progression. Here we investigated whether LOX/HIF1 endows colorectal cancer cells with full competence for aggressive colonization in bone. We show that a high LOX expression in primary tumors from patients with colorectal cancer was associated with poor clinical outcome, irrespective of HIF-1 In addition, LOX was expressed by tumor cells in the bone marrow from colorectal cancer patients with bone metastases. In vivo experimental studies show that LOX overexpression in colorectal cancer cells or systemic delivery of the conditioned medium from LOX-overexpressing colorectal cancer cells promoted tumor cell dissemination in the bone marrow and enhanced osteolytic lesion formation, irrespective of HIF-1 Conversely, silencing or pharmacologic inhibition of LOX activity blocked dissemination of colorectal cancer cells in the bone marrow and tumor-driven osteolytic lesion formation. In vitro, tumor-secreted LOX supported the attachment and survival of colorectal cancer cells to and in the bone matrix, and inhibited osteoblast differentiation. LOX overexpression in colorectal cancer cells also induced a robust production of IL6. In turn, both LOX and IL6 were acting in concert to promote RANKL-dependent osteoclast differentiation, thereby creating an imbalance between bone resorption and bone formation. Collectively, our findings show that LOX supports colorectal cancer cell dissemination in the bone marrow and they reveal a novel mechanism through which LOX-driven IL6 production by colorectal cancer cells impairs bone homeostasis. Cancer Res; 77(2); 268-78. ©2016 AACR.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/secundario , Invasividad Neoplásica/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Animales , Western Blotting , Huesos/metabolismo , Huesos/patología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
J Bone Miner Res ; 21(12): 1913-23, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17002556

RESUMEN

UNLABELLED: Even though it is assumed that multinucleated osteoclasts are migrating cells on the bone surface to be resorbed, we show that they can also selectively transmigrate through layers of cells usually found in the bone microenvironment. This activity is associated with c-src and MMPs and can be stimulated by bone metastatic breast cancer cells, a process blocked by bisphosphonate treatment. INTRODUCTION: Osteoclasts have an hematopoietic origin and are bone-resorbing cells. Monocytic precursors migrate to the bone surface where they fuse to form multinucleated osteoclasts able to migrate over the bone surface. We studied whether multinucleated osteoclasts were also able to transmigrate through tissues. MATERIALS AND METHODS: Murine spleen-derived and green fluorescent protein (GFP)-Raw derived osteoclasts were seeded on osteoblasts and several other cell types. The cells were fixed for 20 minutes, 4 or 12 h after osteoclast seeding, and stained with phalloidin to visualize actin using confocal microscopy. Drugs such as PP2 and GM6001, inhibitors of c-src and matrix metalloproteinases (MMPs), respectively, and risedronate were used to determine osteoclast transmigration regulating factors. RESULTS: We observed by confocal microscopy that multinucleated osteoclasts specifically transmigrate through confluent layers of various cell types present in the bone microenvironment in vitro. This is an efficient process associated with c-src and MMPs but is independent of podosomes. Moreover, conditioned medium from bone metastatic breast cancer cells stimulates osteoclast transmigration in vitro, a process inhibited by bisphosphonate treatment. CONCLUSIONS: Our data describe a new property of mature multinucleated osteoclasts to transmigrate through various cell types. The ability to control this highly regulated osteoclast transmigration process may offer new therapeutic strategies for bone diseases associated with an imbalance in bone remodeling caused by excessive osteoclast resorption.


Asunto(s)
Remodelación Ósea/fisiología , Movimiento Celular/fisiología , Células Gigantes/enzimología , Metaloproteinasas de la Matriz/metabolismo , Osteoclastos/enzimología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Animales , Remodelación Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Dipéptidos/farmacología , Femenino , Células Gigantes/citología , Masculino , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Osteoclastos/citología , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Pirimidinas/farmacología
20.
Oncotarget ; 7(47): 77071-77086, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27776343

RESUMEN

Bone metastases are one of the main complications of prostate cancer and they are incurable. We investigated whether and how estrogen receptor-related receptor alpha (ERRα) is involved in bone tumor progression associated with advanced prostate cancer. By meta-analysis, we first found that ERRα expression is correlated with castration-resistant prostate cancer (CRPC), the hallmark of progressive disease. We then analyzed tumor cell progression and the associated signaling pathways in gain-of-function/loss-of-function CRPC models in vivo and in vitro. Increased levels of ERRα in tumor cells led to rapid tumor progression, with both bone destruction and formation, and direct impacts on osteoclasts and osteoblasts. VEGF-A, WNT5A and TGFß1 were upregulated by ERRα in tumor cells and all of these factors also significantly and positively correlated withERRα expression in CRPC patient specimens. Finally, high levels of ERRα in tumor cells stimulated the pro-metastatic factor periostin expression in the stroma, suggesting that ERRα regulates the tumor stromal cell microenvironment to enhance tumor progression. Taken together, our data demonstrate that ERRα is a regulator of CRPC cell progression in bone. Therefore, inhibiting ERRα may constitute a new therapeutic strategy for prostate cancer skeletal-related events.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Neoplasias Óseas/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores de Estrógenos/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Wnt-5a/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA