Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Bosn J Basic Med Sci ; 16(1): 8-13, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26773172

ABSTRACT

Despite substantial similarities in embryological, cellular and molecular biology features, human and mouse prostates differ in organ morphology and tissue architecture. Thus, a clear understanding of the anatomy and histology of the mouse prostate is essential for the identification of urogenital phenotypes in genetically engineered mice, as well as for the study of the etiology, development, and treatment of human prostatic diseases for which mouse models are used. The purpose of this manuscript is to provide a brief guide for the dissection of the mouse prostate and the identification of its different lobes and histology, to both basic researchers and medical pathologists who are unfamiliar with mouse tissues.


Subject(s)
Prostate/anatomy & histology , Prostate/pathology , Animals , Disease Models, Animal , Dogs , Eosine Yellowish-(YS)/chemistry , Hematoxylin/chemistry , Histology , Humans , Male , Mice , Models, Animal , Phenotype , Prostate/surgery , Rats , Species Specificity
2.
Int J Cancer ; 136(1): 11-20, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-24798488

ABSTRACT

Loss of BRCA2 function stimulates prostate cancer (PCa) cell invasion and is associated with more aggressive and metastatic tumors in PCa patients. Concurrently, the receptor tyrosine kinase c-kit is highly expressed in skeletal metastases of PCa patients and induced in PCa cells placed into the bone microenvironment in experimental models. However, the precise requirement of c-kit for intraosseous growth of PCa and its relation to BRCA2 expression remain unexplored. Here, we show that c-kit expression promotes migration and invasion of PCa cells. Alongside, we found that c-kit expression in PCa cells parallels BRCA2 downregulation. Gene rescue experiments with human BRCA2 transgene in c-kit-transfected PCa cells resulted in reduction of c-kit protein expression and migration and invasion, suggesting a functional significance of BRCA2 downregulation by c-kit. The inverse association between c-kit and BRCA2 gene expressions in PCa cells was confirmed using laser capture microdissection in experimental intraosseous tumors and bone metastases of PCa patients. Inhibition of bone-induced c-kit expression in PCa cells transduced with lentiviral short hairpin RNA reduced intraosseous tumor incidence and growth. Overall, our results provide evidence of a novel pathway that links bone-induced c-kit expression in PCa cells to BRCA2 downregulation and supports bone metastasis.


Subject(s)
Bone Neoplasms/enzymology , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-kit/genetics , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism
3.
Neoplasia ; 10(5): 439-49, 2008 May.
Article in English | MEDLINE | ID: mdl-18472961

ABSTRACT

A variety of proteases have been implicated in prostate cancer (PC) bone metastasis, but the individual contributions of these enzymes remain unclear. Urokinase-type plasminogen activator (uPA), a serine protease, can activate plasminogen and stimulate signaling events on binding its receptor uPAR. In the present study, we investigated the functional role of PC cell-associated uPA in intraosseous tumor growth and bone matrix degradation. Using a severe combined immunodeficient-human mouse model, we found that PC3 cells were the major source of uPA in the experimental bone tumor. Injection of uPA-silenced PC3 cells in bone xenografts resulted in significant reduction of bone tumor burdens and protection of trabecular bones from destruction. The suppressed tumor growth was associated with the level of uPA expression but not with its activity. An increase in the expression of PAI-1, the endogenous uPA inhibitor, was found during in vitro tumor-stromal interactions. Up-regulation of PAI-1 in bone stromal cells and preosteoclasts/osteoblasts was due to soluble factor(s) released by PC cells, and the enhanced PAI-1 expression in turn stimulated PC cell migration. Our results indicate that both tumor-derived uPA and tumor-stroma-induced PAI-1 play important roles in intraosseous metastatic PC growth through regulation of a uPA-uPAR-PAI-1 axis by autocrine/paracrine mechanisms.


Subject(s)
Bone Neoplasms/metabolism , Bone Remodeling , Prostatic Neoplasms/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Coculture Techniques , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunoenzyme Techniques , Male , Mice , Mice, SCID , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Stromal Cells/pathology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...