Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Rev. osteoporos. metab. miner. (Internet) ; 15(1): 21-28, Ene-Mar. 2023. graf, ilus
Artigo em Espanhol | IBECS | ID: ibc-218431

RESUMO

El esqueleto es un órgano metabólicamente activo que se remodela continuamente a la largo de nuestra vida. Estaremodelación implica un equilibrio entre la formación de hueso llevada a cabo por los osteoblastos y la resorción porlos osteoclastos. Los osteocitos son los encargados de regular estos dos procesos y su estimulación mecánica, es esen-cial para mantener el buen funcionamiento óseo y prevenir enfermedades como la osteoporosis. La estimulación de lososteocitos provoca una alteración en la producción y secreción de moléculas de señalización que regulan la actividadde los osteoblastos y los osteoclastos. Las células madre mesenquimales han sido propuestas como posible terapiacelular para la regeneración de distintos tejidos, incluido el tejido óseo. Hipotetizamos en el presente estudio que elsecretoma de células osteocíticas de ratón estimuladas mecánicamente afecta a la capacidad proliferativa, adhesiva ya la expresión génica de células mesenquimales indiferenciadas y células mesenquimales preosteoblásticas. Para ello,se analizaron los procesos biológicos mencionados en líneas continuas celulares preosteoblásticas y células mesenqui-males de ratón en presencia del medio condicionado por células osteocíticas MLO-Y4, después de ser sometidas aestímulo mecánico por flujo de fluido. Se observó que la proliferación aumentó en ambas líneas celulares en presenciadel secretoma de osteocitos estimulados mecánicamente frente al control, mientras que osteocitos no mecanoestimu-lados provocaban su disminución. También se observó un aumento en la capacidad adhesiva de células C3H/10T1/2 trasla estimulación con el secretoma de osteocitos mecanoestimulados. En cuanto a la expresión de genes, solo el factoradipogénico PPARᵞ sufrió alteraciones en células MC3T3-E1 por el secretoma de osteocitos. Estos estudios indican quelos osteocitos pueden modificar el comportamiento biológico de células mesenquimales mediante la secreción de factores solubles.(AU)


Assuntos
Humanos , Osteócitos , Esqueleto , Células-Tronco Mesenquimais , Osteoblastos , Osteoclastos , Reabsorção Óssea , Osteoporose , Doenças Ósseas
2.
Cancers (Basel) ; 13(3)2021 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-33498862

RESUMO

Advanced prostate cancer preferential metastasis to bone is associated with osteomimicry. MINDIN is a secreted matrix protein upregulated in prostate tumors that overexpresses bone-related genes during prostate cancer progression. Na+/H+ exchanger regulatory factor (NHERF-1) is a scaffold protein that has been involved both in tumor regulation and osteogenesis. We hypothesize that NHERF-1 modulation is a mechanism used by MINDIN to promote prostate cancer progression. We analyzed the expression of NHERF-1 and MINDIN in human prostate samples and in a premetastatic prostate cancer mouse model, based on the implantation of prostate adenocarcinoma TRAMP-C1 (transgenic adenocarcinoma of the mouse prostate) cells in immunocompetent C57BL/6 mice. The relationship between NHERF-1 and MINDIN and their effects on cell proliferation, migration, survival and osteomimicry were evaluated. Upregulation of MINDIN and downregulation of NHERF-1 expression were observed both in human prostate cancer samples and in the TRAMP-C1 model. MINDIN silencing restored NHERF-1 expression to control levels in the mouse model. Stimulation with MINDIN reduced NHERF-1 expression and triggered its mobilization from the plasma membrane to the cytoplasm in TRAMP-C1 cells. MINDIN-dependent downregulation of NHERF-1 promoted tumor cell migration and proliferation without affecting osteomimicry and adhesion. We propose that MINDIN downregulates NHERF-1 expression leading to promotion of processes involved in prostate cancer progression.

3.
Endocr Relat Cancer ; 27(7): 441-456, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32357310

RESUMO

Bone metastases are common in advanced prostate cancer patients, but mechanisms by which specific pro-metastatic skeletal niches are formed before tumor cell homing are unclear. We aimed to analyze the effects of proteins secreted by primary prostate tumors on the bone microenvironment before the settlement and propagation of metastases. Here, using an in vivo pre-metastatic prostate cancer model based on the implantation of prostate adenocarcinoma TRAMP-C1 cells in immunocompetent C57BL/6 mice, we identify MINDIN as a prostate tumor secreted protein that induces bone microstructural and bone remodeling gene expression changes before tumor cell homing. Associated with these changes, increased tumor cell adhesion to the endosteum ex vivo and to osteoblasts in vitro was observed. Furthermore, MINDIN promoted osteoblast proliferation and mineralization and monocyte expression of osteoclast markers. ß-catenin signaling pathway revealed to mediate MINDIN actions on osteoblast gene expression but failed to affect MINDIN-induced adhesion to prostate tumor cells or monocyte differentiation to osteoclasts. Our study evidences that MINDIN secretion by primary prostate tumors creates a favorable bone environment for tumor cell homing before metastatic spread.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , beta Catenina/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Transfecção
4.
Cancers (Basel) ; 12(5)2020 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-32344908

RESUMO

Advanced prostate cancers that progress to tumor metastases are often considered incurable or difficult to treat. The etiology of prostate cancers is multi-factorial. Among other factors, de-regulation of calcium signals in prostate tumor cells mediates several pathological dysfunctions associated with tumor progression. Calcium plays a relevant role on tumor cell death, proliferation, motility-invasion and tumor metastasis. Calcium controls molecular factors and signaling pathways involved in the development of prostate cancer and its progression. Such factors and pathways include calcium channels and calcium-binding proteins. Nevertheless, the involvement of calcium signaling on prostate cancer predisposition for bone tropism has been relatively unexplored. In this regard, a diversity of mechanisms triggers transient accumulation of intracellular calcium in prostate cancer cells, potentially favoring bone metastases development. New therapies for the treatment of prostate cancer include compounds characterized by potent and specific actions that target calcium channels/transporters or pumps. These novel drugs for prostate cancer treatment encompass calcium-ATPase inhibitors, voltage-gated calcium channel inhibitors, transient receptor potential (TRP) channel regulators or Orai inhibitors. This review details the latest results that have evaluated the relationship between calcium signaling and progression of prostate cancer, as well as potential therapies aiming to modulate calcium signaling in prostate tumor progression.

5.
Carcinogenesis ; 40(7): 828-839, 2019 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-31168562

RESUMO

Advanced prostate cancer cells preferentially metastasize to bone by acquiring a bone phenotype that allows metastatic cells to thrive in the skeletal environment. Identification of factors that promote the expression of ectopic bone genes-process known as osteomimicry-leading to tumor progression is crucial to prevent and treat metastatic prostate cancer and prolong life expectancy for patients. Here, we identify the extracelular matrix protein mindin in the secretome of prostate adenocarcinoma cells and show that mindin overexpression in human and mouse TRAMP-C1-induced prostate tumors correlates with upregulated levels of bone-related genes in the tumorigenic prostate tissues. Moreover, mindin silencing decreased osteomimicry in adenocarcinoma cells and in the prostate tumor mice model, as well as reduced tumor cell proliferation, migration and adhesion to bone cells. Inhibition of the extracellular signal-regulated kinase 1/2 (ERK 1/2) phosphorylation decreased the proliferative, migratory and pro-adhesion actions of mindin on prostate tumor cells. In addition, conditioned media obtained by crosstalk stimulation of either osteocytes or osteoblasts with the secretome of TRAMP-C1 cells promoted osteomimicry in prostate tumor cells; an effect inhibited by mindin silencing of TRAMP-C1 cells. In vivo, tibiae of primary tumor-bearing mice overexpressed the pro-angiogenic and pro-metastattic factor vascular endothelial growth factor receptor 2 (VEGFR2) in a mindin-dependent manner. Our findings indicate that mindin is a novel regulator of osteomimicry in prostate tumors and potentially mediates tumor-bone cell crosstalk, suggesting its promising role as a target to inhibit bone metastases.


Assuntos
Adenocarcinoma/patologia , Neoplasias Ósseas/secundário , Osso e Ossos/patologia , Proteínas da Matriz Extracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/patologia , Animais , Neoplasias Ósseas/patologia , Osso e Ossos/citologia , Carcinogênese/patologia , Adesão Celular , Comunicação Celular , Linhagem Celular Tumoral/transplante , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Proteínas da Matriz Extracelular/genética , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Proteínas de Neoplasias/genética , Osteoblastos/patologia , Osteócitos/patologia , Fosforilação , Próstata/citologia , Próstata/patologia , Regulação para Cima , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...