Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Chem Sci ; 9(3): 743-753, 2018 Jan 21.
Article in English | MEDLINE | ID: mdl-29629144

ABSTRACT

Breast neoplasms frequently colonize bone and induce development of osteolytic bone lesions by disrupting the homeostasis of the bone microenvironment. This degenerative process can lead to bone pain and pathological bone fracture, a major cause of cancer morbidity and diminished quality of life, which is exacerbated by our limited ability to monitor early metastatic disease in bone and assess fracture risk. Spurred by its label-free, real-time nature and its exquisite molecular specificity, we employed spontaneous Raman spectroscopy to assess and quantify early metastasis driven biochemical alterations to bone composition. As early as two weeks after intracardiac inoculations of MDA-MB-435 breast cancer cells in NOD-SCID mice, Raman spectroscopic measurements in the femur and spine revealed consistent changes in carbonate substitution, overall mineralization as well as crystallinity increase in tumor-bearing bones when compared with their normal counterparts. Our observations reveal the possibility of early stage detection of biochemical changes in the tumor-bearing bones - significantly before morphological variations are captured through radiographic diagnosis. This study paves the way for a better molecular understanding of altered bone remodeling in such metastatic niches, and for further clinical studies with the goal of establishing a non-invasive tool for early metastasis detection and prediction of pathological fracture risk in breast cancer.

2.
Oncol Lett ; 15(1): 871-878, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29399152

ABSTRACT

The transcriptional co-regulator Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain-2 (CITED2) may promote breast tumor growth; however, the mechanisms by which its effects are mediated remain to be fully elucidated. Tumor-associated macrophages serve an important function in tumor development and progression and are recruited by chemotactic factors produced by cells within the tumor microenvironment. The present study assessed the effects of CITED2 silencing on macrophage recruitment in two xenograft mouse models of human breast cancer, one in which tumor growth was sensitive to CITED2 silencing (MDA-MB-231) and one in which it was insensitive (MDA-MB-468). The present study identified that silencing CITED2 significantly attenuated macrophage infiltration in MDA-MB-231 but not MDA-MB-468 orthotopic tumors, concordant with its effect on tumor growth. Correspondingly, conditioned media obtained from CITED2-silenced MDA-MB-231 cells exhibited a significantly decreased ability to induce macrophage recruitment by Transwell migration assay, whereas the chemotactic effect of MDA-MB-468 conditioned media was unaffected. Examining the expression of macrophage chemoattractants within orthotopic tumors and tumor cell-conditioned media revealed a significant decrease in C-C motif chemokine ligand (CCL)20 mRNA and protein expression following CITED2-silencing in MDA-MB-231 cells, compared with that in cells transfected with scramble shRNA. However, mRNA and protein expression was unaffected by CITED2-silencing in MDA-MB-468 cells. Furthermore, chromatin immunoprecipitation analysis revealed that CITED2 was localized to the CCL20 promoter in MDA-MB-231 cells, suggesting that it serves a direct function in its regulation, which is consistent with the effect of CITED2 silencing on CCL20 expression. Lastly, neutralizing CCL20 in the conditioned media of MDA-MB-231 cells significantly inhibited macrophage recruitment. Collectively, these results suggest that CITED2 is involved in modulating macrophage recruitment, representing a novel mechanism through which it may influence tumor growth. This may be partly mediated by regulating tumor cell production of the chemokine CCL20.

3.
Oncotarget ; 8(4): 6169-6178, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28008154

ABSTRACT

While we previously demonstrated that CITED2 expression in primary breast tumor tissues is elevated relative to normal mammary epithelium and inversely correlated with patient survival, its functional impact on primary tumor development and progression remained unknown. To address this issue, we examined the effect of CITED2 silencing on the growth of human breast cancer cell lines MDA-MB-231 and MDA-MB-468 following orthotopic administration in vivo. Here, we show that CITED2 silencing significantly attenuated MDA-MB-231 primary tumor growth concordant with reduced tumor vascularization, while MDA-MB-468 primary tumor growth and tumor vascularization remained unaffected. Correspondingly, expression of VEGFA was significantly reduced in shCITED2-expressing MDA-MB-231, but not MDA-MB-468 tumors. Consistent with the observed pattern of vascularization and VEGFA expression, we found that TGF-ß stimulation induced expression of VEGFA and enhanced CITED2 recruitment to the VEGFA promoter in MDA-MA-231 cells, while failing to induce VEGFA expression in MDA-MB-468 cells. Further supporting its involvement in TGF-ß-induced expression of VEGFA, CITED2 silencing prevented TGF-ß induction of VEGFA expression in MDA-MB-231 cells. Collectively, these data indicate that CITED2 regulates primary breast tumor growth, likely by influencing tumor vasculature via TGF-ß-dependent regulation of VEGFA.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Neovascularization, Pathologic , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Promoter Regions, Genetic , RNA Interference , Repressor Proteins/genetics , Signal Transduction/drug effects , Time Factors , Trans-Activators/genetics , Transfection , Tumor Burden , Vascular Endothelial Growth Factor A/genetics
4.
Mol Cancer Res ; 14(8): 730-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27216153

ABSTRACT

UNLABELLED: Previously, we identified the transcriptional coactivator CITED2 as a potential facilitator of bone metastasis using a murine mammary cancer model. Extending these studies to human breast cancer, it was observed that CITED2 mRNA expression was significantly elevated in patient specimens of metastatic breast cancer relative to primary tumors, with highest levels in metastasis to bone relative to non-bone sites. To further evaluate CITED2 functions in breast cancer metastasis, CITED2 expression was stably reduced in the human breast cancer cell lines MDA-MB-231 and MDA-MB-468, which are metastatic in animal models. While CITED2 knockdown had no effect on cell proliferation, cell migration and invasion were significantly reduced, as was the establishment of metastasis following intracardiac administration in athymic nude mice. To explore the mechanism behind these effects, gene expression following CITED2 knockdown in MDA-MB-231 cells by cDNA microarray was performed. As confirmed at the mRNA and protein levels in both MDA-MB-231 and MDA-MB-468 cells, expression of the NF-κB regulator IKKα was significantly reduced, along with several NF-κB targets with known roles in metastasis (OPN, MMP9, uPA, SPARC, IL11, and IL1ß). Furthermore, ChIP assay revealed recruitment of CITED2 to the promoter of IKKα, indicating a direct role in regulating its expression. Consistent with reduced IKKα expression, CITED2 knockdown inhibited both canonical and noncanonical NF-κB signaling. Finally, restoration of IKKα expression following CITED2 knockdown in MDA-MB-231 and MDA-MB-468 cells rescued their invasive ability. Collectively, these data demonstrate that CITED2 modulates metastatic ability in human breast cancer cells, at least in part, through the regulation of IKKα. IMPLICATIONS: The current study highlights the role of CITED2 in facilitating breast cancer metastasis, partly via regulation of IKKα. Mol Cancer Res; 14(8); 730-9. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , I-kappa B Kinase/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/pathology , Down-Regulation , Female , Humans , I-kappa B Kinase/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Repressor Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Transfection
5.
J Bone Miner Res ; 31(7): 1381-90, 2016 07.
Article in English | MEDLINE | ID: mdl-26890063

ABSTRACT

CCL20 is a member of the macrophage inflammatory protein family and is reported to signal monogamously through the receptor CCR6. Although studies have identified the genomic locations of both Ccl20 and Ccr6 as regions important for bone quality, the role of CCL20/CCR6 signaling in regulating bone mass is unknown. By micro-computed tomography (µCT) and histomorphometric analysis, we show that global loss of Ccr6 in mice significantly decreases trabecular bone mass coincident with reduced osteoblast numbers. Notably, CCL20 and CCR6 were co-expressed in osteoblast progenitors and levels increased during osteoblast differentiation, indicating the potential of CCL20/CCR6 signaling to influence osteoblasts through both autocrine and paracrine actions. With respect to autocrine effects, CCR6 was found to act as a functional G protein-coupled receptor in osteoblasts and although its loss did not appear to affect the number or proliferation rate of osteoblast progenitors, differentiation was significantly inhibited as evidenced by delays in osteoblast marker gene expression, alkaline phosphatase activity, and mineralization. In addition, CCL20 promoted osteoblast survival concordant with activation of the PI3K-AKT pathway. Beyond these potential autocrine effects, osteoblast-derived CCL20 stimulated the recruitment of macrophages and T cells, known facilitators of osteoblast differentiation and survival. Finally, we generated mice harboring a global deletion of Ccl20 and found that Ccl20(-/-) mice exhibit a reduction in bone mass similar to that observed in Ccr6(-/-) mice, confirming that this phenomenon is regulated by CCL20 rather than alternate CCR6 ligands. Collectively, these data indicate that CCL20/CCR6 signaling may play an important role in regulating bone mass accrual, potentially by modulating osteoblast maturation, survival, and the recruitment of osteoblast-supporting cells. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Cancellous Bone/metabolism , Chemokine CCL20/metabolism , Osteoblasts/metabolism , Receptors, CCR6/metabolism , Signal Transduction/physiology , Animals , Cancellous Bone/cytology , Cell Survival , Chemokine CCL20/genetics , Mice , Mice, Knockout , Organ Size/physiology , Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CCR6/genetics
6.
Cancer Res ; 74(22): 6509-18, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25217524

ABSTRACT

Acquired resistance to TGFß is a key step in the early stages of tumorigenesis. Mutations in TGFß signaling components are rare, and little is known about the development of resistance in breast cancer. On the other hand, an activated Notch pathway is known to play a substantial role in promoting breast cancer development. Here, we present evidence of crosstalk between these two pathways through HEYL. HEYL, a basic helix-loop-helix transcription factor and a direct target of Notch signaling, is specifically overexpressed in breast cancer. HEYL represses TGFß activity by binding to TGFß-activated Smads. HeyL(-/-) mice have defective mammary gland development with fewer terminal end buds. On the other hand, HeyL transgenic mice show accelerated mammary gland epithelial proliferation and 24% of multiparous mice develop mammary gland cancer. Therefore, repression of TGFß signaling by Notch acting through HEYL may promote initiation of breast cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Breast Neoplasms/pathology , Receptors, Notch/physiology , Repressor Proteins/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , Animals , Cells, Cultured , Female , Humans , Mice , Mice, Transgenic , Signal Transduction/drug effects , Smad3 Protein/physiology
7.
PLoS One ; 8(7): e66752, 2013.
Article in English | MEDLINE | ID: mdl-23861746

ABSTRACT

Bone is the most common site of breast cancer metastasis and once established, it is frequently incurable. Critical to our ability to prevent and treat bone metastasis is the identification of the key factors mediating its establishment and understanding their biological function. To address this issue we previously carried out an in vivo selection process to isolate murine mammary tumor sublines possessing an enhanced ability to colonize the bone. A comparison of gene expression between parental cells and sublines by genome-wide cDNA microarray analysis revealed several potential mediators of bone metastasis, including the pyrophosphate-generating ectoenzyme Enpp1. By qRT-PCR and Western analysis we found that expression of Enpp1 was elevated in human breast cancer cell lines known to produce bone metastasis in animal models compared to non-metastatic and normal mammary epithelial cell lines. Further, in clinical specimens, levels of Enpp1 were significantly elevated in human primary breast tumors relative to normal mammary epithelium, with highest levels observed in breast-bone metastasis as determined by qRT-PCR and immunohistochemical analysis. To examine the potential role of Enpp1 in the development of bone metastasis, Enpp1 expression was stably increased in the breast cancer cell line MDA-MB-231 and the ability to colonize the bone following intracardiac and direct intratibial injection of athymic nude mice was determined. By both routes of administration, increased expression of Enpp1 enhanced the ability of MDA-MB-231 cells to form tumors in the bone relative to cells expressing vector alone, as determined by digital radiography and histological analysis. Taken together, these data suggest a potential role for Enpp1 in the development of breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Animals , Bone Neoplasms/diagnostic imaging , Cell Line, Tumor , Enzyme Activation , Female , Gene Expression , Humans , Mammary Neoplasms, Experimental , Mice , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/genetics , Radiography
8.
Biochem Biophys Res Commun ; 437(2): 261-6, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23811274

ABSTRACT

Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (CITED2) is a member of the CITED family of non-DNA binding transcriptional co-activators of the p300/CBP-mediated transcription complex. Previously, we identified CITED2 as being overexpressed in human breast tumors relative to normal mammary epithelium. Upon further investigation within the estrogen receptor (ER)-positive subset of these breast tumor samples, we found that CITED2 mRNA expression was elevated in those associated with poor survival. In light of this observation, we investigated the effect of elevated CITED2 levels on ER function. While ectopic overexpression of CITED2 in three ER-positive breast cancer cell lines (MCF-7, T47D, and CAMA-1) did not alter cell proliferation in complete media, growth was markedly enhanced in the absence of exogenous estrogen. Correspondingly, cells overexpressing CITED2 demonstrated reduced sensitivity to the growth inhibitory effects of the selective estrogen receptor modulator, 4-hydroxytamoxifen. Subsequent studies revealed that basal ER transcriptional activity was elevated in CITED2-overexpressing cells and was further increased upon the addition of estrogen. Similarly, basal and estrogen-induced expression of the ER-regulated genes trefoil factor 1 (TFF1) and progesterone receptor (PGR) was higher in cells overexpressing CITED2. Concordant with this observation, ChIP analysis revealed higher basal levels of CITED2 localized to the TFF-1 and PGR promoters in cells with ectopic overexpression of CITED2, and these levels were elevated further in response to estrogen stimulation. Taken together, these data indicate that CITED2 functions as a transcriptional co-activator of ER in breast cancer cells and that its increased expression in tumors may result in estrogen-independent ER activation, thereby reducing estrogen dependence and response to anti-estrogen therapy.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Estrogen/metabolism , Repressor Proteins/physiology , Trans-Activators/physiology , Transcription, Genetic/physiology , Base Sequence , Breast Neoplasms/pathology , Cell Division , Cell Line, Tumor , DNA Primers , Female , Humans , Polymerase Chain Reaction
9.
PLoS One ; 7(7): e40799, 2012.
Article in English | MEDLINE | ID: mdl-22792407

ABSTRACT

Pathological bone resorption is a source of significant morbidity in diseases affecting the skeleton such as rheumatoid arthritis, periodontitis, and cancer metastasis to bone. Evidence indicates that elevated levels of inflammatory mediators such as IL-1, IL-6, and TNF-α play a role in this process by promoting the formation of bone-resorbing osteoclasts. Additionally, current studies have identified inflammatory chemokines of the macrophage inflammatory protein (MIP) family as potential mediators of pathological bone resorption, where both MIP-1α and -3α have been shown to enhance osteoclast (OCL) development. In this study we provide evidence that MIP-1δ, whose expression is associated with renal cell carcinoma bone metastasis and rheumatoid arthritis, enhances OCL formation in vitro via a direct effect on OCL precursors. Consistent with this ability, exposure of OCL precursors to MIP-1δ resulted in the activation of PLCγ2 and NF-κB, two signaling pathways known to regulate OCL differentiation. Moreover, MIP-1δ induced expression and nuclear translocation of NFATc1, a master regulator of osteoclastogenesis, which was dependent on activation of both the PLCγ2 and NFκB signaling pathways. Lastly, consistent with in vitro studies, in vivo administration of MIP-1δ significantly increased OCL number and resorption area as determined using a murine calvarial bone resorption model. Taken together, these data highlight the potential of MIP-1δ as a mediator of pathological bone resorption and provide insight into the molecular mechanism through which MIP-1δ enhances osteoclastogenesis.


Subject(s)
Macrophage Inflammatory Proteins/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/physiology , Osteolysis/metabolism , Phospholipase C gamma/metabolism , Signal Transduction , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Enzyme Activation , Female , Mice , NFATC Transcription Factors/genetics , Osteoclasts/cytology , Osteolysis/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism
10.
Sci Transl Med ; 3(106): 106ra108, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22030751

ABSTRACT

Most breast cancers originate in the epithelial cells lining the breast ducts. Intraductal administration of cancer therapeutics would lead to high drug exposure to ductal cells and eliminate preinvasive neoplasms while limiting systemic exposure. We performed preclinical studies in N-methyl-N'-nitrosourea-treated rats to compare the effects of 5-fluorouracil, carboplatin, nanoparticle albumin-bound paclitaxel, and methotrexate to the previously reported efficacy of pegylated liposomal doxorubicin (PLD) on treatment of early and established mammary tumors. Protection from tumor growth was observed with all five agents, with extensive epithelial destruction present only in PLD-treated rats. Concurrently, we initiated a clinical trial to establish the feasibility, safety, and maximum tolerated dose of intraductal PLD. In each eligible woman awaiting mastectomy, we visualized one ductal system and administered dextrose or PLD using a dose-escalation schema (2 to 10 mg). Intraductal administration was successful in 15 of 17 women with no serious adverse events. Our preclinical studies suggest that several agents are candidates for intraductal therapy. Our clinical trial supports the feasibility of intraductal administration of agents in the outpatient setting. If successful, administration of agents directly into the ductal system may allow for "breast-sparing mastectomy" in select women.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Adult , Animals , Carboplatin/administration & dosage , Carboplatin/therapeutic use , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Drug Administration Routes , Female , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Humans , Mammary Glands, Animal , Mammary Neoplasms, Animal/drug therapy , Methylnitrosourea/therapeutic use , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/therapeutic use , Rats , Rats, Sprague-Dawley , Young Adult
11.
Int J Cancer ; 126(4): 876-84, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19642106

ABSTRACT

Breast cancer metastases develop in the bone more frequently than any other site and are a common cause of morbidity in the form of bone pain, pathological fractures, nerve compression and life-threatening hypercalcemia. Despite ongoing research efforts, the molecular and cellular mechanisms that regulate breast cancer cell homing to and colonization of the bone as well as resultant pathological bone alteration remain poorly understood. To identify key mediators promoting breast cancer metastasis to bone, we utilized an immunocompetent, syngeneic murine model of breast cancer metastasis employing the mammary tumor cell line NT2.5. Following intracardiac injection of NT2.5 cells in neu-N mice, metastases developed in the bone, liver and lung, closely mimicking the anatomical distribution of metastases in patients with breast cancer. Using an in vivo selection process, we established NT2.5 sublines demonstrating an enhanced ability to colonize the bone and liver. Genome-wide cDNA microarray analysis comparing gene expression between parental NT2.5 cells and established sublines revealed both known and novel mediators of bone metastasis and osteolysis, including the transcriptional co-activator CITED2. In further studies, we found that expression of CITED2 was elevated in human primary breast tumors and bone metastasis compared to normal mammary epithelium and was highest in breast cancer cell lines that cause osteolytic bone metastasis in animal models. In addition, reducing CITED2 expression in NT2.5 cells inhibited the establishment of bone metastasis and osteolysis in vivo, suggesting a potential role for CITED2 in promoting breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/pathology , Breast Neoplasms/pathology , Repressor Proteins/genetics , Trans-Activators/genetics , Animals , Bone Neoplasms/secondary , Cell Adhesion/physiology , Cell Division/physiology , Cell Line, Tumor , DNA, Neoplasm/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mice , Neoplasm Metastasis/pathology , Oligonucleotide Array Sequence Analysis , Osteolysis/pathology , RNA, Messenger/genetics , Repressor Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Trans-Activators/physiology , Transcription, Genetic
12.
Clin Exp Metastasis ; 25(8): 865-70, 2008.
Article in English | MEDLINE | ID: mdl-18709334

ABSTRACT

Bone metastasis occurs frequently in renal cell carcinoma (RCC) patients causing significant morbidity by stimulating excessive osteolysis, yet the mechanisms responsible have been little studied. Matrix metalloproteinases (MMPs) are over-expressed in many cancer types and are believed to play a role in bone metastasis, however, the expression of MMPs in RCC bone metastasis (RBM) has not been investigated. Due to their ability to degrade the main component of organic bone matrix, type I collagen, we investigated the expression of MMP-1, -2, -8, -9, and -13 in RBM. By quantitative (q)RT-PCR, expression of MMP-13 was significantly increased in RBM tissues relative to that in RCC and adjacent normal kidney while no differences in the expression of MMP-1, -2, -8, or -9 mRNA were observed. Correspondingly, increased expression of MMP-13 protein was also observed in RBM relative to RCC by immunohistochemical analysis. Intriguingly, the expression of MMP-13 in the human RBM cell line RBM1-IT4 was stimulated by TGF-beta1, a growth factor abundant in the bone microenvironment and known to promote RBM-induced osteolysis in animals. Exposure of RBM1-IT4 cells to TGF-beta1 increased MMP-13 mRNA levels as well as the latent and active forms of MMP-13 protein. Further, stable expression of a dominant-negative TGF-beta type II receptor in RBM1-IT4 cells inhibited MMP-13 expression following TGF-beta1 exposure. These data suggest that MMP-13 expression is elevated in RBM relative to primary RCC and adjacent normal kidney, and is regulated at the cellular level by TGF-beta1.


Subject(s)
Bone Neoplasms/enzymology , Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , Matrix Metalloproteinase 13/metabolism , Transforming Growth Factor beta1/metabolism , Blotting, Western , Bone Neoplasms/secondary , Carcinoma, Renal Cell/secondary , Enzyme Induction , Humans , Immunoenzyme Techniques , Kidney Neoplasms/pathology , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Osteolysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured
13.
Cancer Res ; 68(5): 1261-6, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316587

ABSTRACT

Approximately 30% of patients with renal cell carcinoma (RCC) develop bone metastasis, which is characterized by extensive osteolysis leading to severe bone pain and pathologic fracture. Although the mechanism of RCC-induced osteolysis is unknown, studies of bone metastasis have shown that tumor-induced changes in bone remodeling are likely mediated by alterations in the bone microenvironment. Here, we report the discovery of a novel osteoclast stimulatory factor secreted by RCC bone metastasis (RBM). Through microarray analysis, we found expression of the chemokine, macrophage inflammatory protein-1 delta (MIP-1 delta), to be increased in RBM versus patient-matched primary RCC tissues and confirmed this finding by quantitative reverse transcription-PCR (qRT-PCR) and ELISA (P < 0.05). Furthermore, MIP-1 delta expression in RBM tissues was significantly (P < 0.001) higher than in human bone marrow, suggesting a potential alteration of the bone microenvironment. The receptors for MIP-1 delta, CCR1 and CCR3, were expressed in both osteoclast precursors and mature, bone-resorbing osteoclasts as shown by qRT-PCR and Western analysis. In functional studies, MIP-1 delta stimulated chemotaxis of two osteoclast precursor cell types: murine bone marrow mononuclear cells (BM-MNC) and RAW 264.7 cells. Furthermore, MIP-1 delta treatment of murine calvaria caused increased bone resorption as determined by measurement of released calcium. Correspondingly, MIP-1 delta significantly enhanced osteoclast formation and activity in response to RANKL in both BM-MNC and RAW 264.7 cells. Taken together, these data suggest that MIP-1 delta expression is increased in RBM relative to RCC and bone marrow, and may promote RBM-induced osteolysis by stimulating the recruitment and differentiation of osteoclast precursors into mature osteoclasts.


Subject(s)
Bone and Bones/pathology , Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Macrophage Inflammatory Proteins/physiology , Osteoclasts/metabolism , Animals , Bone and Bones/metabolism , Carcinoma, Renal Cell/pathology , Cell Differentiation , Cell Movement , Humans , Kidney Neoplasms/metabolism , Leukocytes, Mononuclear/metabolism , Macrophage Inflammatory Proteins/chemistry , Mice , Neoplasm Metastasis , RANK Ligand/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Cancer Res ; 67(17): 7977-82, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804705

ABSTRACT

Brain metastasis is the most commonly occurring intracranial tumor whose incidence seems to be increasing. With standard therapy, the average survival time of patients is approximately 8 months, and treatment often leads to neurologic dysfunction in long-term survivors, emphasizing the need for novel therapeutics. Clostridium perfringens enterotoxin (CPE) has recently been shown to rapidly and specifically destroy cancer cells expressing CPE receptors claudin-3 and claudin-4. Unfortunately, the utility of CPE is precluded by systemic toxicity because its receptors are expressed in numerous organs. Here, we provide the first preclinical evidence that CPE may be uniquely suited to the local treatment of brain metastasis. By immunohistochemical analysis, claudin-3 and claudin-4 were expressed frequently in metastases from breast (15 of 18), lung (15 of 20), and colon (12 of 14) carcinoma, and infrequently in metastases from renal cell carcinoma (2 of 16) and melanoma (2 of 16). In contrast, expression of claudin-3 and claudin-4 was absent in adjacent normal brain tissue. Further examination of the central nervous system (CNS) revealed low or undetectable levels of claudin-3 and claudin-4 in all regions tested by Western and immunohistochemical analysis. Treatment of breast cancer cell lines (MCF-7, MDA-MB-468, NT2.5-luc) and normal human astrocytes with CPE in vitro resulted in rapid and dose-dependent cytolysis exclusively in breast cancer cells, correlating with claudin-3 and claudin-4 expression. Moreover, intracranial CPE treatment significantly inhibited tumor growth and increased survival in two murine models of breast cancer brain metastasis, without any apparent local or systemic toxicity. These data suggest that CPE therapy may have efficacy against a wide variety of brain metastases without CNS toxicity.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/pathology , Enterotoxins/therapeutic use , Animals , Apoptosis/drug effects , Astrocytes/metabolism , Breast Neoplasms/mortality , Carcinoma/mortality , Claudin-3 , Claudin-4 , Drug Evaluation, Preclinical , Female , Humans , Membrane Proteins/metabolism , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Cancer Metastasis Rev ; 26(3-4): 691-704, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17768599

ABSTRACT

The development of bone metastasis from renal cell carcinoma (RCC) signals a transition to a terminal state for the patient with previously isolated disease. These patients may suffer the morbidity of severe, persistent pain, pathologic fractures, and spinal compression from vertebral metastasis before they succumb to their cancer. Although recent advancements have been made in the understanding of breast and prostate bone metastasis, there has been less knowledge in the area of metastatic RCC to the skeleton. This particular cancer in bone remains relatively resistant to standard forms of treatment such as radiation and chemotherapy. A better understanding of the biology of RCC bone metastasis is critically needed in order to improve treatment. Bone-derived cell lines and an experimental animal model have been developed in order to explore the relevant mechanisms of how RCC cells survive within and destroy the bone. This review will focus on the growth factor signaling pathways most important for the RCC-stimulated osteoclast-mediated bone destruction, namely the epidermal growth factor receptor (EGF-R) and transforming growth factor-beta receptor (TGF-betaR) pathways. By inhibiting these receptors, growth of RCC within the bone is decreased which, directly or indirectly, decreases bone destruction.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Animals , Diphosphonates/therapeutic use , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Humans , Interleukin-6/physiology , Mice , Signal Transduction , Transforming Growth Factor beta/physiology
16.
J Bone Miner Res ; 22(1): 37-44, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17032147

ABSTRACT

UNLABELLED: Bone metastases develop in approximately 30% of patients with RCC, and the mechanisms responsible for this phenomenon are unknown. We found that TGF-beta1 stimulation of RCC bone metastasis cells promotes tumor growth and bone destruction possibly by stimulating paracrine interactions between tumor cells and the bone. INTRODUCTION: Bone metastasis is a frequent complication and causes marked morbidity in patients with renal cell carcinoma (RCC). Surprisingly, the specific mechanisms of RCC interaction with bone have been scarcely studied despite the inability to prevent or effectively treat bone metastasis. Bone is a reservoir for various growth factors including the pleiotropic cytokine TGF-beta1. TGF-beta1 has been shown to have tumor-supportive effects on advanced cancers and evidence suggests its involvement in promoting the development of breast cancer bone metastasis. Here, we studied the potential role of TGF-beta1 in the growth of RCC bone metastasis (RBM). MATERIALS AND METHODS: To inhibit TGF-beta1 signaling, RBM cells stably expressing a dominant-negative (DN) TGF-betaRII cDNA were generated. The in vivo effect of TGF-beta1 on RBM tumor growth and osteolysis was determined by histological and radiographic analysis, respectively, of athymic nude mice after intratibial injection of parental, empty vector, or DN RBM cells. The in vitro effect of TGF-beta1 on RBM cell growth was determined after TGF-beta1 treatment by MTT assay. RESULTS: TGF-beta1 and the TGF-beta receptors I and II (TGF-betaRI/II) were consistently expressed in both RBM tissues and cell lines. Inhibition of TGF-beta1 signaling in RBM cells significantly reduced tumor establishment and osteolysis observed in vivo after injection into the murine tibia, although no effect on tumor establishment was observed after injection of RBM cells subcutaneously or into the renal subcapsule. Treatment of five RBM cell lines with TGF-beta1 in vitro either had no effect (2/5) or resulted in a significant inhibition (3/5) of cell growth, suggesting that TGF-beta1 may promote RBM tumor growth indirectly in vivo. CONCLUSIONS: TGF-beta1 stimulation of RBM cells plays a role in promoting tumor growth and subsequent osteolysis in vivo, likely through the initiation of tumor-promoting paracrine interactions between tumor cells and the bone microenvironment. These data suggest that inhibition of TGF-beta1 signaling may be useful in the treatment of RBM.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Animals , Cell Division , Cell Line, Tumor , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mice , Mice, Nude , Osteolysis , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
17.
Expert Rev Mol Med ; 8(18): 1-11, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16887048

ABSTRACT

The claudin (CLDN) family of transmembrane proteins plays a critical role in the maintenance of epithelial and endothelial tight junctions. In addition to their function in preserving the structure of tight junctions, CLDNs might also play a role in the maintenance of the cytoskeleton and in cell signalling. Interestingly, several studies have recently reported specific CLDN family members to be overexpressed in a wide variety of cancer types. Although their functional role in cancer progression remains unclear, the differential expression of these proteins between tumour and normal cells, in addition to their membrane localisation, makes them prime candidates for cancer therapy. Preclinical studies have shown that tumour cells overexpressing CLDNs can be successfully targeted via several approaches, including the use of anti-CLDN antibodies as well as the cytolytic enterotoxin from Clostridium perfringens. Further studies are needed to determine the potential systemic toxicity of this approach considering the ubiquitous expression of CLDNs in the body, but CLDN-targeted therapeutics appear to have promise in the treatment of cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/physiology , Neoplasms/genetics , Neoplasms/therapy , Amino Acid Motifs , Claudin-1 , Clostridium perfringens/metabolism , Endothelium/metabolism , Epithelium/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Protein Conformation , Tight Junctions/metabolism , Tissue Distribution
19.
Cancer Res ; 66(2): 638-45, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16423990

ABSTRACT

In cancer patients and in those at high risk, systemic exposure to agents for therapy or prevention is accompanied by undesirable side effects. We hypothesized that it is possible to prevent and treat breast cancer by introducing anticancer agents into the mammary ductal network. Here, we show the efficacy of intraductally administered anticancer agents 4-hydroxytamoxifen and pegylated liposomal doxorubicin (PLD) in the prevention and treatment of breast cancer using the rat N-methyl-N'-nitrosourea-induced and spontaneous HER-2/neu transgenic mouse (neu-N) models of breast cancer. Intraductal administration of PLD to neu-N mice caused regression of established tumors and prevented tumor development more effectively than i.v. injection (P < 0.0001). Intraductal administration resulted in lower circulating levels of PLD compared with i.v. administration, with no evidence of systemic toxicity or long-term histopathologic changes in the mammary gland. Compared with systemic administration, intraductal injection provides direct access to breast lesions with higher local and lower systemic drug exposure. These studies suggest that this approach has potential for application to prevention and neoadjuvant therapy of early breast cancer.


Subject(s)
Doxorubicin/analogs & derivatives , Estrogen Antagonists/pharmacology , Mammary Neoplasms, Animal/prevention & control , Polyethylene Glycols/pharmacology , Tamoxifen/analogs & derivatives , Animals , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Administration Routes , Estrogen Antagonists/administration & dosage , Female , Humans , Mammary Glands, Animal , Mice , Mice, Transgenic , Neoadjuvant Therapy , Polyethylene Glycols/administration & dosage , Rats , Tamoxifen/administration & dosage , Tamoxifen/pharmacology
20.
Cancer Res ; 65(23): 10801-9, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322226

ABSTRACT

Aggressive cancer phenotypes are a manifestation of many different genetic alterations that promote rapid proliferation and metastasis. In this study, we show that stable overexpression of Twist in a breast cancer cell line, MCF-7, altered its morphology to a fibroblastic-like phenotype, which exhibited protein markers representative of a mesenchymal transformation. In addition, it was observed that MCF-7/Twist cells had increased vascular endothelial growth factor (VEGF) synthesis when compared with empty vector control cells. The functional changes induced by VEGF in vivo were analyzed by functional magnetic resonance imaging (MRI) of MCF-7/Twist-xenografted tumors. MRI showed that MCF-7/Twist tumors exhibited higher vascular volume and vascular permeability in vivo than the MCF-7/vector control xenografts. Moreover, elevated expression of Twist in breast tumor samples obtained from patients correlated strongly with high-grade invasive carcinomas and with chromosome instability, particularly gains of chromosomes 1 and 7. Taken together, these results show that Twist overexpression in breast cancer cells can induce angiogenesis, correlates with chromosomal instability, and promotes an epithelial-mesenchymal-like transition that is pivotal for the transformation into an aggressive breast cancer phenotype.


Subject(s)
Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Chromosomal Instability , Nuclear Proteins/biosynthesis , Twist-Related Protein 1/biosynthesis , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Capillary Permeability/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Claudins , Female , Humans , Membrane Proteins/genetics , Mesoderm/metabolism , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Transcriptional Activation , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL