Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
J Cell Biochem ; 118(9): 2693-2699, 2017 09.
Article in English | MEDLINE | ID: mdl-28112417

ABSTRACT

Breast cancer is a complex and multifactorial disease. Tumors have a heterogeneous microenvironment, which have multiple interactions with other cell types, greatly influencing the behavior of tumor cells and response to therapy. The 3D culture mimics the microenvironment better found in vivo and is more appropriated than the traditional 2D culture made from plastic to test the cellular response to drugs. To investigate the effects of [10]-gingerol on breast tumor cells, we used physiologically relevant three-dimensional (3D) cultures of malignant and non-malignant human breast cells grown in laminin-rich extracellular matrix gels (lr-ECM). Our results showed selective cytotoxicity of [10]-gingerol against the malignant T4-2 breast cancer cell line compared to non-malignant S1 cells. The compound reverted the malignant phenotype of the cancer cells, downregulating the expression of epidermal growth factor receptor (EGFR) and ß1-integrin. Moreover, [10]-gingerol induced apoptosis in this cell line. These results suggest that [10]-gingerol may be an effective compound to use as adjuvant therapy in breast cancer treatment. J. Cell. Biochem. 118: 2693-2699, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Fatty Alcohols/pharmacology , Guaiacol/analogs & derivatives , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation/drug effects , ErbB Receptors/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Guaiacol/pharmacology , Humans , Integrin beta1/biosynthesis , Mice, Nude , Neoplasm Proteins/biosynthesis
2.
BMC Cancer ; 15: 660, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26449734

ABSTRACT

BACKGROUND: The REversion-inducing Cysteine-rich protein with Kazal motif (RECK) is a well-known inhibitor of matrix metalloproteinases (MMPs) and cellular invasion. Although high expression levels of RECK have already been correlated with a better clinical outcome for several tumor types, its main function, as well as its potential prognostic value for breast cancer patients, remain unclear. METHODS: The RECK expression profile was investigated in a panel of human breast cell lines with distinct aggressiveness potential. RECK functional analysis was undertaken using RNA interference methodology. RECK protein levels were also analyzed in 1040 cases of breast cancer using immunohistochemistry and tissue microarrays (TMAs). The association between RECK expression and different clinico-pathological parameters, as well as the overall (OS) and disease-free (DFS) survival rates, were evaluated. RESULTS: Higher RECK protein expression levels were detected in more aggressive breast cancer cell lines (T4-2, MDA-MB-231 and Hs578T) than in non-invasive (MCF-7 and T47D) and non-tumorigenic (S1) cell lines. Indeed, silencing RECK in MDA-MB-231 cells resulted in elevated levels of pro-MMP-9 and increased invasion compared with scrambled (control) cells, without any effect on cell proliferation. Surprisingly, by RECK immunoreactivity analysis on TMAs, we found no association between RECK positivity and survival (OS and DFS) in breast cancer patients. Even considering the different tumor subtypes (luminal A, luminal B, Her2 type and basal-like) or lymph node status, RECK remained ineffective for predicting the disease outcome. Moreover, by multivariate Cox regression analysis, we found that RECK has no prognostic impact for OS and DFS, relative to standard clinical variables. CONCLUSIONS: Although it continues to serve as an invasion and MMP inhibitor in breast cancer, RECK expression analysis is not useful for prognosis of these patients.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , GPI-Linked Proteins/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , GPI-Linked Proteins/genetics , Gene Expression , Humans , Kaplan-Meier Estimate , Matrix Metalloproteinase 9/metabolism , Middle Aged , Neoplasm Grading , Prognosis , Proportional Hazards Models , Risk Factors , Tumor Burden
3.
Sci Rep ; 5: 8919, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25747469

ABSTRACT

The interplay between host genetics, tumor microenvironment and environmental exposure in cancer susceptibility remains poorly understood. Here we assessed the genetic control of stromal mediation of mammary tumor susceptibility to low dose ionizing radiation (LDIR) using backcrossed F1 into BALB/c (F1Bx) between cancer susceptible (BALB/c) and resistant (SPRET/EiJ) mouse strains. Tumor formation was evaluated after transplantation of non-irradiated Trp53-/- BALB/c mammary gland fragments into cleared fat pads of F1Bx hosts. Genome-wide linkage analysis revealed 2 genetic loci that constitute the baseline susceptibility via host microenvironment. However, once challenged with LDIR, we discovered 13 additional loci that were enriched for genes involved in cytokines, including TGFß1 signaling. Surprisingly, LDIR-treated F1Bx cohort significantly reduced incidence of mammary tumors from Trp53-/- fragments as well as prolonged tumor latency, compared to sham-treated controls. We demonstrated further that plasma levels of specific cytokines were significantly correlated with tumor latency. Using an ex vivo 3-D assay, we confirmed TGFß1 as a strong candidate for reduced mammary invasion in SPRET/EiJ, which could explain resistance of this strain to mammary cancer risk following LDIR. Our results open possible new avenues to understand mechanisms of genes operating via the stroma that affect cancer risk from external environmental exposures.


Subject(s)
Breast Neoplasms/genetics , Cytokines/genetics , Genetic Predisposition to Disease/genetics , Neoplasms, Radiation-Induced/genetics , Quantitative Trait Loci/genetics , Tumor Microenvironment/genetics , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Risk Factors , Transforming Growth Factor beta1/genetics
4.
Development ; 142(6): 1028-42, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25758218

ABSTRACT

The development of the mammary gland is unique: the final stages of development occur postnatally at puberty under the influence of hormonal cues. Furthermore, during the life of the female, the mammary gland can undergo many rounds of expansion and proliferation. The mammary gland thus provides an excellent model for studying the 'stem/progenitor' cells that allow this repeated expansion and renewal. In this Review, we provide an overview of the different cell types that constitute the mammary gland, and discuss how these cell types arise and differentiate. As cellular differentiation cannot occur without proper signals, we also describe how the tissue microenvironment influences mammary gland development.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Mammary Glands, Human/cytology , Mammary Glands, Human/growth & development , Signal Transduction/physiology , Stem Cell Niche/physiology , Stem Cells/physiology , Adipocytes/physiology , Animals , Epithelial Cells/physiology , Female , Fibroblasts/physiology , Humans , Mice , Puberty/physiology
5.
J Cell Biochem ; 116(8): 1668-79, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25735873

ABSTRACT

The development of the mammary gland involves formation of a branched arboreal structure resulting from the penetration and proliferation of epithelial cells into the fat pad. The mammary cells invade by remodeling their surrounding extracellular matrix (ECM), which are rich in proteins, and glycans such as heparan sulfate proteoglycans (HSPGs). There is increasing literature on how the interaction between signaling by ECM and matrix metalloproteinases (MMPs) is relevant to morphogenetic and physiological contexts. Here we sought to understand how heparanase, the sole mammalian heparan sulfate-degrading endoglycosidase may regulate mammary gland development. We found a robust localization of heparanase within growing end buds during branching in vivo. Using three-dimensional (3D) organotypic cultures, we showed that heparanase expression and activity are required for mammary epithelial invasion/branching within dense collagen I gels. Morphometric analysis of glands from both heparanase-overexpressing and knockout mice showed a direct correlation between degree of branching and the heparanase levels, confirming our 3D organotypic culture observations. Finally, we uncovered a reciprocal association between levels of heparanase and MMP14, a membrane-bound MMP, shedding further light on how branching occurs within developing mammary glands.


Subject(s)
Glucuronidase/metabolism , Mammary Glands, Animal/growth & development , Matrix Metalloproteinase 14/metabolism , Morphogenesis , Animals , Cell Movement , Epithelial Cells/physiology , Female , Gene Expression Regulation, Developmental , Glucuronidase/genetics , Mammary Glands, Animal/enzymology , Mice , Organ Culture Techniques , Signal Transduction
6.
Dev Biol ; 399(1): 15-26, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25500615

ABSTRACT

Intercellular communication is essential for glandular functions and tissue homeostasis. Gap junctions couple cells homotypically and heterotypically and co-ordinate reciprocal responses between the different cell types. Connexins (Cxs) are the main mammalian gap junction proteins, and the distribution of some Cx subtypes in the heterotypic gap junctions is not symmetrical; in the murine mammary gland, Cx26, Cx30 and Cx32 are expressed only in the luminal epithelial cells and Cx43 is expressed only in myoepithelial cells. Expression of all four Cxs peaks during late pregnancy and throughout lactation suggesting essential roles for these proteins in the functional secretory activity of the gland. Transgenic (Tg) mice over-expressing Cx26 driven by keratin 5 promoter had an unexpected mammary phenotype: the mothers were unable to feed their pups to weaning age leading to litter starvation and demise in early to mid-lactation. The mammary gland of K5-Cx26 female mice developed normally and produced normal levels of milk protein, suggesting a defect in delivery rather than milk production. Because the mammary gland of K5-Cx26 mothers contained excessive milk, we hypothesized that the defect may be in an inability to eject the milk. Using ex vivo three-dimensional mammary organoid cultures, we showed that tissues isolated from wild-type FVB females contracted upon treatment with oxytocin, whereas, organoids from Tg mice failed to do so. Unexpectedly, we found that ectopic expression of Cx26 in myoepithelial cells altered the expression of endogenous Cx43 resulting in impaired gap junction communication, demonstrated by defective dye coupling in mammary epithelial cells of Tg mice. Inhibition of gap junction communication or knock-down of Cx43 in organoids from wild-type mice impaired contraction in response to oxytocin, recapitulating the observations from the mammary glands of Tg mice. We conclude that Cx26 acts as a trans-dominant negative for Cx43 function in myoepithelial cells, highlighting the importance of cell type-specific expression of Cxs for optimal contractile function of the mammary myoepithelium.


Subject(s)
Connexin 43/metabolism , Connexins/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cells, Cultured , Connexin 43/genetics , Connexins/genetics , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Gap Junctions/metabolism , Gap Junctions/physiology , Gene Expression , Lactation/genetics , Lactation/physiology , Male , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice, Transgenic , Microscopy, Confocal , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Contraction/physiology , Organ Culture Techniques , Oxytocin/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Videotape Recording
7.
Development ; 140(2): 343-52, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23250208

ABSTRACT

Epithelial cell invasion through the extracellular matrix (ECM) is a crucial step in branching morphogenesis. The mechanisms by which the mammary epithelium integrates cues from the ECM with intracellular signaling in order to coordinate invasion through the stroma to make the mammary tree are poorly understood. Because the cell membrane-bound matrix metalloproteinase Mmp14 is known to play a key role in cancer cell invasion, we hypothesized that it could also be centrally involved in integrating signals for mammary epithelial cells (MECs) to navigate the collagen 1 (CL-1)-rich stroma of the mammary gland. Expression studies in nulliparous mice that carry a NLS-lacZ transgene downstream of the Mmp14 promoter revealed that Mmp14 is expressed in MECs at the tips of the branches. Using both mammary organoids and 3D organotypic cultures, we show that MMP activity is necessary for invasion through dense CL-1 (3 mg/ml) gels, but dispensable for MEC branching in sparse CL-1 (1 mg/ml) gels. Surprisingly, however, Mmp14 without its catalytic activity was still necessary for branching. Silencing Mmp14 prevented cell invasion through CL-1 and disrupted branching altogether; it also reduced integrin ß1 (Itgb1) levels and attenuated MAPK signaling, disrupting Itgb1-dependent invasion/branching within CL-1 gels. FRET imaging revealed that Mmp14 associates directly with Itgb1. We identified a domain of Mmp14 that is required for modulating the levels of Itgb1, MEC signaling and the rate of invasion within CL-1. These results shed light on hitherto undescribed non-proteolytic activities of Mmp14 that are necessary for the Itgb1-dependent biochemical and mechanical signals that regulate branching in the mammary epithelium.


Subject(s)
Cell Membrane/metabolism , Integrin beta1/metabolism , MAP Kinase Signaling System , Matrix Metalloproteinase 14/metabolism , Animals , Catalytic Domain , Collagen/metabolism , Fluorescence Resonance Energy Transfer/methods , Gene Silencing , Lentivirus/metabolism , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Models, Biological , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Transgenes
8.
J Mammary Gland Biol Neoplasia ; 17(2): 103-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22573197

ABSTRACT

Tissue organogenesis is directed by both intercellular interactions and communication with the surrounding microenvironment. When cells are cultured on two-dimensional plastic substrata (2D), important signals controlling programs of cell proliferation, metabolism, differentiation and death responsible for the formation of correct tissue-specific architecture and function are lost. Designing three-dimensional (3D), physiologically relevant culture models, we can recapitulate some crucial aspects of the dynamic and reciprocal signaling necessary for establishing and maintaining tissue specific morphogenic programs. Here we briefly describe the details of robust methods for culturing mouse primary mammary organoids in 3D gels of different extracellular matrices and describe techniques for analyzing the resulting structures. These designer microenvironments are useful for both understanding branching morphogenesis and signaling integrations, but also for analysis of individual susceptibilities and drug testing.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Models, Anatomic , Morphogenesis , Organoids/growth & development , Tissue Culture Techniques , Animals , Biocompatible Materials/metabolism , Collagen/metabolism , Collagen Type I/metabolism , Drug Combinations , Extracellular Matrix/metabolism , Female , Imaging, Three-Dimensional , Inguinal Canal , Laminin/metabolism , Mice , Mice, Inbred BALB C , Organoids/cytology , Proteoglycans/metabolism , Staining and Labeling/methods
9.
Genes Dev ; 24(24): 2800-11, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159820

ABSTRACT

Organization into polarized three-dimensional structures defines whether epithelial cells are normal or malignant. In a model of morphogenesis, we show that inhibiting key signaling pathways in human breast cancer cells leads to "phenotypic reversion" of the malignant cells. Using architecture as an endpoint, we report that, in all cases, signaling through Raf/MEK/ERK disrupted tissue polarity via matrix metalloproteinase9 (MMP9) activity. Induction of Raf or activation of an engineered, functionally inducible MMP9 in nonmalignant cells led to loss of tissue polarity, and reinitiated proliferation. Conversely, inhibition of Raf or MMP9 with small molecule inhibitors or shRNAs restored the ability of cancer cells to form polarized quiescent structures. Silencing MMP9 expression also reduced tumor growth dramatically in a murine xenograft model. LC-MS/MS analysis comparing conditioned medium from nonmalignant cells with or without active MMP9 revealed laminin 111 (LM1) as an important target of MMP9. LM1 has been implicated in acinar morphogenesis; thus, its degradation by MMP9 provides a mechanism for loss of tissue polarity and reinitiation of growth associated with MMP9 activity. These findings underscore the importance of the dynamic reciprocity between the extracellular matrix integrity, tissue polarity, and Raf/MEK/ERK and MMP9 activities, providing an axis for either tissue homeostasis or malignant progression.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Matrix Metalloproteinase 9/metabolism , Morphogenesis , raf Kinases/physiology , Animals , Cell Culture Techniques , Cell Polarity , Cell Proliferation , Humans , Laminin/metabolism , Mice , Neoplasms, Experimental , Signal Transduction , Transplantation, Heterologous
10.
Mol Biosyst ; 4(5): 417-25, 2008 May.
Article in English | MEDLINE | ID: mdl-18414739

ABSTRACT

The backbone mobility of the C-terminal domain of procollagen C-proteinase enhancer (NTR PCOLCE1), part of a connective tissue glycoprotein, was determined using 15N NMR spectroscopy. NTR PCOLCE1 has been shown to be a netrin-like domain and adopts an OB-fold such as that found in the N-terminal domain of tissue inhibitors of metalloproteinases-1 (N-TIMP-1), N-TIMP-2, the laminin-binding domain of agrin and the C-terminal domain of complement protein C5. NMR relaxation dynamics of NTR PCOLCE1 highlight conformational flexibility in the N-terminus, strand A and the proximal CD loop. This region in N-TIMP is known to be essential for inhibitory activity against the matrix metalloproteinases and suggests that this region is of equal importance for NTR PCOLCE1, although the specific functional activity of the NTR PCOLCE1 domain is still unknown. Dynamics observed within the structural core of NTR PCOLCE1 that are not observed in N-TIMP molecules suggest that although the two domains have a similar architecture, the NTR PCOLCE1 domain will show different thermodynamic properties on binding and hence the target molecule could be somewhat different from that observed for the TIMPs. ModelFree order parameters show that NTR PCOLCE1 has more flexibility than both N-TIMP-1 and N-TIMP-2.


Subject(s)
Extracellular Matrix Proteins/chemistry , Glycoproteins/chemistry , Receptors, Cell Surface/chemistry , Tissue Inhibitor of Metalloproteinases/chemistry , Amino Acid Sequence , Cloning, Molecular , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Netrin Receptors , Protein Folding , Protein Precursors/chemistry , Protein Structure, Tertiary , Sequence Alignment
11.
Cancer Res ; 68(5): 1378-87, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316601

ABSTRACT

A crucial step in human breast cancer progression is the acquisition of invasiveness. There is a distinct lack of human cell culture models to study the transition from preinvasive to invasive phenotype as it may occur "spontaneously" in vivo. To delineate molecular alterations important for this transition, we isolated human breast epithelial cell lines that showed partial loss of tissue polarity in three-dimensional reconstituted basement membrane cultures. These cells remained noninvasive; however, unlike their nonmalignant counterparts, they exhibited a high propensity to acquire invasiveness through basement membrane in culture. The genomic aberrations and gene expression profiles of the cells in this model showed a high degree of similarity to primary breast tumor profiles. The xenograft tumors formed by the cell lines in three different microenvironments in nude mice displayed metaplastic phenotypes, including squamous and basal characteristics, with invasive cells exhibiting features of higher-grade tumors. To find functionally significant changes in transition from preinvasive to invasive phenotype, we performed attribute profile clustering analysis on the list of genes differentially expressed between preinvasive and invasive cells. We found integral membrane proteins, transcription factors, kinases, transport molecules, and chemokines to be highly represented. In addition, expression of matrix metalloproteinases MMP9, MMP13, MMP15, and MMP17 was up-regulated in the invasive cells. Using small interfering RNA-based approaches, we found these MMPs to be required for the invasive phenotype. This model provides a new tool for dissection of mechanisms by which preinvasive breast cells could acquire invasiveness in a metaplastic context.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Animals , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Metaplasia/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis
12.
Matrix Biol ; 27(3): 211-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18164932

ABSTRACT

Dialysis related amyloidosis (DRA) is a progressive and serious complication in patients under long-term hemodialysis and mainly leads to osteo-articular diseases. Although beta(2)-microglobulin (beta2-m) is the major structural component of beta2-m amyloid fibrils, the initiation of amyloid formation is not clearly understood. Here, we have identified procollagen C-proteinase enhancer-1 (PCPE-1) as a new interacting protein with beta2-m by screening a human synovium cDNA library. The interaction of beta2-m with full-length PCPE-1 was confirmed by immunoprecipitation, solid-phase binding and pull-down assays. By yeast two-hybrid analysis and pull-down assay, beta2-m appeared to interact with PCPE-1 via the NTR (netrin-like) domain and not via the CUB (C1r/C1s, Uegf and BMP-1) domain region. In synovial tissues derived from hemodialysis patients with DRA, beta2-m co-localized and formed a complex with PCPE-1. beta2-m did not alter the basal activity of bone morphogenetic protein-1/procollagen C-proteinase (BMP-1/PCP) nor BMP-1/PCP activity enhanced by PCPE-1. PCPE-1 did not stimulate beta2-m amyloid fibril formation from monomeric beta2-m in vitro under acidic and neutral conditions as revealed by thioflavin T fluorescence spectroscopy and electron microscopy. Since PCPE-1 is abundantly expressed in connective tissues rich in type I collagen, it may be involved in the initial accumulation of beta2-m in selected tissues such as tendon, synovium and bone. Furthermore, since such preferential deposition of beta2-m may be linked to subsequent beta2-m amyloid fibril formation, the disruption of the interaction between beta2-m and PCPE-1 may prevent beta2-m amyloid fibril formation and therefore PCPE-1 could be a new target for the treatment of DRA.


Subject(s)
Amyloid/chemistry , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , beta 2-Microglobulin/chemistry , Amino Acid Sequence , Bone Morphogenetic Protein 1 , Bone Morphogenetic Proteins/chemistry , Dose-Response Relationship, Drug , Enhancer Elements, Genetic , Gene Library , Humans , Metalloendopeptidases/chemistry , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Two-Hybrid System Techniques
13.
Cancer Res ; 67(23): 11106-10, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056432

ABSTRACT

Polo-like kinase 1 (PLK1) has important functions in maintaining genome stability via its role in mitosis. Because PLK1 is up-regulated in many invasive carcinomas, we asked whether it may also play a role in acquisition of invasiveness, a crucial step in transition to malignancy. In a model of metaplastic basal-like breast carcinoma progression, we found that PLK1 expression is necessary but not sufficient to induce invasiveness through laminin-rich extracellular matrix. PLK1 mediates invasion via vimentin and beta1 integrin, both of which are necessary. We observed that PLK1 phosphorylates vimentin on Ser82, which in turn regulates cell surface levels of beta1 integrin. We found PLK1 to be also highly expressed in preinvasive in situ carcinomas of the breast. These results support a role for the involvement of PLK1 in the invasion process and point to this pathway as a potential therapeutic target for preinvasive and invasive breast carcinoma treatment.


Subject(s)
Breast Neoplasms/enzymology , Cell Cycle Proteins/metabolism , Extracellular Matrix/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Movement , Collagen , Drug Combinations , Female , Humans , In Situ Nick-End Labeling , Integrin beta1/metabolism , Laminin/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proteoglycans , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Vimentin/metabolism , Polo-Like Kinase 1
14.
Cancer Res ; 67(18): 8662-70, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875706

ABSTRACT

Transforming growth factor beta1 (TGFbeta) is a tumor suppressor during the initial stage of tumorigenesis, but it can switch to a tumor promoter during neoplastic progression. Ionizing radiation (IR), both a carcinogen and a therapeutic agent, induces TGFbeta activation in vivo. We now show that IR sensitizes human mammary epithelial cells (HMEC) to undergo TGFbeta-mediated epithelial to mesenchymal transition (EMT). Nonmalignant HMEC (MCF10A, HMT3522 S1, and 184v) were irradiated with 2 Gy shortly after attachment in monolayer culture or treated with a low concentration of TGFbeta (0.4 ng/mL) or double treated. All double-treated (IR + TGFbeta) HMEC underwent a morphologic shift from cuboidal to spindle shaped. This phenotype was accompanied by a decreased expression of epithelial markers E-cadherin, beta-catenin, and ZO-1, remodeling of the actin cytoskeleton, and increased expression of mesenchymal markers N-cadherin, fibronectin, and vimentin. Furthermore, double treatment increased cell motility, promoted invasion, and disrupted acinar morphogenesis of cells subsequently plated in Matrigel. Neither radiation nor TGFbeta alone elicited EMT, although IR increased chronic TGFbeta signaling and activity. Gene expression profiling revealed that double-treated cells exhibit a specific 10-gene signature associated with Erk/mitogen-activated protein kinase (MAPK) signaling. We hypothesized that IR-induced MAPK activation primes nonmalignant HMEC to undergo TGFbeta-mediated EMT. Consistent with this, Erk phosphorylation was transiently induced by irradiation and persisted in irradiated cells treated with TGFbeta, and treatment with U0126, a MAP/Erk kinase (MEK) inhibitor, blocked the EMT phenotype. Together, these data show that the interactions between radiation-induced signaling pathways elicit heritable phenotypes that could contribute to neoplastic progression.


Subject(s)
Breast/drug effects , Breast/radiation effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Transforming Growth Factor beta/pharmacology , Breast/metabolism , Breast/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mesoderm/drug effects , Mesoderm/pathology , Mesoderm/radiation effects , Mitogen-Activated Protein Kinase Kinases/metabolism
15.
Radiat Res ; 166(6): 839-48, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17149983

ABSTRACT

The three mammalian transforming growth factor beta (TGF-beta) isoforms are each secreted in a latent complex in which TGF-beta homodimers are non-covalently associated with homodimers of their respective pro-peptide called the latency-associated peptide (LAP). Release of TGF-beta from its LAP, called activation, is required for binding of TGF-beta to cellular receptors, making extracellular activation a critical regulatory point for TGF-beta bioavailability. Our previous work demonstrated that latent TGF-beta1 (LTGF-beta1) is efficiently activated by ionizing radiation in vivo and by reactive oxygen species (ROS) generated by Fenton chemistry in vitro. In the current study, we determined the specific ROS and protein target that render LTGF-beta1 redox sensitive. First, we compared LTGF-beta1, LTGF-beta2 and LTGF-beta3 to determine the generality of this mechanism of activation and found that redox-mediated activation is restricted to the LTGF-beta1 isoform. Next, we used scavengers to determine that ROS activation was a function of OH(.) availability, confirming oxidation as the primary mechanism. To identify which partner of the LTGF-beta1 complex was functionally modified, each was exposed to ROS and tested for the ability to form a latent complex. Exposure of TGF-beta1 did not alter its ability to associate with LAP, but exposing LAP-beta1 to ROS prohibited this phenomenon, while treatment of ROS-exposed LAP-beta1 with a mild reducing agent restored its ability to neutralize TGF-beta1 activity. Taken together, these results suggest that ROS-induced oxidation in LAP-beta1 triggers a conformational change that releases TGF-beta1. Using site-specific mutation, we identified a methionine residue at amino acid position 253 unique to LAP-beta1 as critical to ROS-mediated activation. We propose that LTGF-beta1 contains a redox switch centered at methionine 253, which allows LTGF-beta1 to act uniquely as an extracellular sensor of oxidative stress in tissues.


Subject(s)
Latent TGF-beta Binding Proteins/chemistry , Reactive Oxygen Species/chemistry , Dose-Response Relationship, Drug , Protein Isoforms/chemistry
16.
Curr Opin Cell Biol ; 16(5): 558-64, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15363807

ABSTRACT

Matrix metalloproteinases (MMPs) are endopeptidases that contribute to growth, development and wound healing as well as to pathologies such as arthritis and cancer. Until recently, it has been thought that MMPs participate in these processes simply by degrading extracellular matrix (ECM) molecules. However, it is now clear that MMP activity is much more directed and causes the release of cryptic information from the ECM. By precisely cleaving large insoluble ECM components and ECM-associated molecules, MMPs liberate bioactive fragments and growth factors and change ECM architecture, all of which influence cellular behavior. Thus, MMPs have become a focal point for understanding matrix biology.


Subject(s)
Extracellular Matrix/physiology , Matrix Metalloproteinases/metabolism , Peptide Fragments/metabolism , Basement Membrane/physiology , Growth Substances/metabolism , Matrix Metalloproteinases/physiology , Models, Biological
17.
J Cell Biol ; 162(6): 1123-33, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12975354

ABSTRACT

During puberty, mouse mammary epithelial ducts invade the stromal mammary fat pad in a wave of branching morphogenesis to form a complex ductal tree. Using pharmacologic and genetic approaches, we find that mammary gland branching morphogenesis requires transient matrix metalloproteinase (MMP) activity for invasion and branch point selection. MMP-2, but not MMP-9, facilitates terminal end bud invasion by inhibiting epithelial cell apoptosis at the start of puberty. Unexpectedly, MMP-2 also represses precocious lateral branching during mid-puberty. In contrast, MMP-3 induces secondary and tertiary lateral branching of ducts during mid-puberty and early pregnancy. Nevertheless, the mammary gland is able to develop lactational competence in MMP mutant mice. Thus, specific MMPs refine the mammary branching pattern by distinct mechanisms during mammary gland branching morphogenesis.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells/enzymology , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/growth & development , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Dipeptides/pharmacology , Embryonic Induction/drug effects , Embryonic Induction/genetics , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Immunohistochemistry , Lactation/drug effects , Lactation/genetics , Mammary Glands, Animal/cytology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Mice, Transgenic , Morphogenesis/drug effects , Morphogenesis/genetics , Mutation/drug effects , Mutation/genetics , Organ Culture Techniques , Pregnancy , Protease Inhibitors/pharmacology , Sexual Maturation/drug effects , Sexual Maturation/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
18.
Breast Cancer Res ; 5(1): 53-6, 2003.
Article in English | MEDLINE | ID: mdl-12559047

ABSTRACT

The Era of Hope meeting addressed with a multidisciplinary approach the most critical issues in breast carcinogenesis. The issues that we summarize here include: a) the use of rodent models for the study of mammary gland development and breast tumorigenesis; b) the effects of stroma on mammary epithelial differentiation and malignant transformation; c) a further characterization of the interactions between steroid and growth factor receptors; d) the improvement of technologies for early detection of breast tumors and the establishment of their progression; and e) the development of vaccines as potential new therapies against specific tumor markers.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Breast/pathology , Breast Neoplasms/genetics , Cancer Vaccines/therapeutic use , DNA Repair , Female , Humans , Neovascularization, Pathologic/pathology
19.
Curr Biol ; 12(6): R209-11, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11909548

ABSTRACT

The unique membrane-associated inhibitor of matrix metalloproteinases, RECK, is required for vascular maturation during embryogenesis. The phenotype of a loss of function mutation of RECK shows the importance of pericellular proteolysis in development.


Subject(s)
Endothelium, Vascular/growth & development , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Amino Acid Sequence , Animals , Blood Vessels/embryology , Blood Vessels/metabolism , GPI-Linked Proteins , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Humans , Matrix Metalloproteinases/metabolism , Molecular Sequence Data , Mutation , Sequence Homology, Amino Acid , Tissue Inhibitor of Metalloproteinase-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL