scispace - formally typeset
Search or ask a question

Showing papers in "Journal of Mammary Gland Biology and Neoplasia in 2001"


Journal ArticleDOI
Sandra J. Gendler1
TL;DR: Recent discoveries that suggest that MUC1 may be a multifunctional protein, located on the surfaces of cells as a sensor of the environment, poised to signal to the interior when things go awry are highlighted.
Abstract: MUC1 is a large, heavily glycosylated mucin expressed on the apical surfaces of most simple, secretory epithelia including the mammary gland, gastrointestinal, respiratory, urinary and reproductive tracts. Although MUC1 was thought to be an epithelial-specific protein, it is now known to be expressed on a variety of hematopoietic cells as well. Mucins function in protection and lubrication of epithelial surfaces. Transmembrane mucins, which contain cytoplasmic tail domains, appear to have additional functions through their abilities to interact with many proteins involved in signal transduction and cell adhesion. The goal of this review is to highlight recent discoveries that suggest that MUC1 may be a multifunctional protein, located on the surfaces of cells as a sensor of the environment, poised to signal to the interior when things go awry.

550 citations


Journal ArticleDOI
TL;DR: The loss, at least to some degree, of core 2 based glycans is a consistent feature of MUC1 mucin when it is expressed by mammary tumours as demonstrated by the unmasking of the SM3 epitope in greater than 90% of breast carcinomas.
Abstract: Glycosylation is a very important posttranslational modification of many biologically relevant molecules. A change in the structure of glycans added to glycoproteins and glycolipids is a common feature of the change to malignancy. With the cloning of many of the glycosyltransferases and the identification of specific target molecules, it is now possible to define these changes at the molecular level and to dissect the mechanisms involved. Within the mammary gland, mucin-type O-linked glycosylation has been studied most extensively. In normal resting, pregnant and lactating breast, mucin O-glycans are largely extended (core 2 type) structures. In contrast, mucin O-glycans found in breast carcinomas are often truncated (core 1 type). One mechanism that is responsible for this increase in core 1 structures is a change in the expression of glycosyltransferases, particularly an increase in the expression of the sialyltransferase, ST3Gal-I. The loss, at least to some degree, of core 2 based glycans is a consistent feature of MUC1 mucin when it is expressed by mammary tumours as demonstrated by the unmasking of the SM3 epitope in greater than 90% of breast carcinomas.

230 citations


Journal ArticleDOI
TL;DR: An overview of issues that need to be considered when analyzing studies of prognostic factors as well as a review of the currently recognized and the newer candidate prognostic Factors are provided.
Abstract: Decisions regarding the use of adjuvant therapy for breast cancer are strongly influenced by the risk of disease recurrence and death. These risks are now determined by examining the currently recognized breast cancer prognostic factors, including clinical stage, axillary nodal status, tumor size and grade, hormone receptor status, and presence of lymphovascular involvement. Newer factors are being evaluated in an attempt to more precisely define disease-related prognosis. This paper provides an overview of issues that need to be considered when analyzing studies of prognostic factors as well as a review of the currently recognized and the newer candidate prognostic factors.

219 citations


Journal ArticleDOI
TL;DR: There is reason for optimism that agents targeting c-erbB-2 signaling will have profound and selective effects in breast cancer, either as single agents or more likely in combination with other therapeutic agents, to enhance their potency.
Abstract: Gene amplification and/or overexpression of the c-erbB-2/HER2/neu tyrosine kinase are linked with poor prognosis in breast cancer. This is manifest in shorter disease-free intervals, increased risk of metastasis, and resistance to many types of therapy. The molecular mechanisms and signaling circuitry underlying these phenomena are now being elucidated. c-erbB-2, although having no known soluble ligand, is transactivated by heterodimerization with other family members (EGFR, c-erbB-3, c-erbB-4). Receptor activation potentiates tumor cell motility, protease secretion and invasion, and also modulates cell cycle checkpoint function, DNA repair, and apoptotic responses. Since it is expressed at low levels in normal adult tissues, c-erbB-2 is an ideal target for therapy. There is reason for optimism that agents targeting c-erbB-2 signaling will have profound and selective effects in breast cancer, either as single agents or more likely in combination with other therapeutic agents, to enhance their potency.

196 citations


Journal ArticleDOI
TL;DR: A number of angiogenic factors and matrix degrading enzymes (MMPs) are tightly complexed with CD44 isoforms, suggesting that they are involved in the onset of oncogenic signals required for breast tumor cell invasion and migration.
Abstract: CD44, a hyaluronan (HA) receptor, belongs to a family of transmembrane glycoproteins which exists as several isoforms. Cell surface expression of certain CD44 isoforms is closely correlated with the progression and prognosis of breast cancers. A number of angiogenic factors (e.g., VEGF and FGF-2) and matrix degrading enzymes (MMPs) are tightly complexed with CD44 isoforms, suggesting that they are involved in the onset of oncogenic signals required for breast tumor cell invasion and migration. Most importantly, interaction of extracellular matrix components (e.g., HA) with cells triggers the cytoplasmic domain of CD44 isoforms to bind its unique downstream effectors (e.g., the cytoskeletal protein ankyrin or various oncogenic signaling molecules-Tiam1, RhoA-activated ROK, c-Src kinase and p185HER2) and to coordinate intracellular signaling pathways (e.g., Rho/Ras signaling and receptor-linked/non-receptor-linked tyrosine kinase pathways), leading to a concomitant onset of multiple cellular functions (e.g., tumor cell growth, migration and invasion) and breast tumor progression.

158 citations


Journal ArticleDOI
TL;DR: The structure and function of the Notch receptor, as well as the components that comprise and modify the signaling pathway, are summarized and the potential role of Notch in mammary gland development and tumorigenesis is discussed.
Abstract: The Notch receptor protein and its signaling pathway have been well conserved throughout evolution and appear to be pivotal components in cell fate decisions during development. Recent studies suggest that, depending on the cellular and developmental context, Notch signaling may also affect cell proliferation and programmed cell death. Mammals have four related Notch genes. One of these, designated Notch-4, was found to be a common integration site for the mouse mammary tumor virus in mouse mammary tumors. One consequence of this type of viral integration event is the ectopic expression of the intracellular domain of Notch-4 that corresponds to a gain-of-function mutation. Expression of “activated” Notch-4 in mammary epithelium has profound effects on mammary gland development and tumorigenesis. In this review, we briefly summarize the structure and function of the Notch receptor, as well as the components that comprise and modify the signaling pathway. Finally we discuss the potential role of Notch in mammary gland development and tumorigenesis.

143 citations


Journal ArticleDOI
TL;DR: Clinical studies that show that OPN levels in tumors and blood are elevated in women with metastatic breast cancer and may offer promise as prognostic markers in breast cancer are summarized.
Abstract: Osteopontin (OPN) is a secreted, integrin-binding protein which has been implicated in cancer, as well as other pathologies and some aspects of normal development. Here we focus on the role of OPN in breast cancer. We describe studies that have shown that OPN plays a role in normal mammary gland development as well as in progression of breast cancer. We also summarize studies that have shown that OPN can play a functional role in malignancy of breast cancer. At least some of these effects are mediated by specific cell surface integrins (αvβ3 vs. αvβ1 and αvβ5) and lead to increased cell migration, activation of growth factor/receptor pathways (e.g. HGF and EGF), and increased proteolytic enzyme activity (e.g. uPA). We also summarize clinical studies that show that OPN levels in tumors and blood are elevated in women with metastatic breast cancer and may offer promise as prognostic markers in breast cancer.

137 citations


Journal ArticleDOI
TL;DR: It is established that normal development and homeostasis of the mammary gland is critically dependent on regulated ERBB signaling and the value of animal models in deciphering roles for the complex ERBB network in this dynamic tissue is illustrated.
Abstract: The four ERBB receptors and their multiple polypeptide ligands are differentially expressed during development of the mouse mammary gland. Profiles suggest that ERBB1/EGF receptor (EGFR)4 and ERBB2/Neu are required during ductal morphogenesis, whereas the Neuregulin (NRG) receptors, ERBB3 and ERBB4, are preferentially expressed through alveolar morphogenesis and lactation. Consistent with these profiles, recent gene knockouts established that EGFR and its ligand, Amphiregulin (AR), are essential for ductal morphogenesis in the adolescent mouse and likely provide the required epithelial-stromal signal. In contrast, the phenotypes of transgenic mice expressing dominant negative ERBB2 and ERBB4 proteins suggest that these receptors differentially act to promote or maintain alveolar differentiation. This view of ERBB action provides a conceptual framework for future testing using more sophisticated conditional knockout models. New or existing transgenic mice are also being used to better understand the contributions of ERBB receptors and ligands to mammary tumorigenesis, as well as to more closely mimic the human disease. Recent studies have focused on defining molecular events in neoplastic progression, and in the case of ERBB2/Neu, the requirement for ERBB heterodimerization partners as well as the relative importance of gene amplification versus gene mutation. Collectively, these recent studies establish that normal development and homeostasis of the mammary gland is critically dependent on regulated ERBB signaling. They also illustrate the value of animal models in deciphering roles for the complex ERBB network in this dynamic tissue.

118 citations


Journal ArticleDOI
TL;DR: The hypothesis is that the persistent disruption of the microenvironment in irradiated tissue compromises its ability to suppress carcinogenesis, and that tissue forces must be subjugated to establish a tumor.
Abstract: How do normal tissues limit the development of cancer? This review discusses the evidence that normal cells effectively restrict malignant behavior, and that such tissue forces must be subjugated to establish a tumor. The action of ionizing radiation will be specifically discussed regarding the disruption of the microenvironment that promotes the transition from preneoplastic to neoplastic growth. Unlike the highly unpredictable nature of genetic mutations, the response of normal cells to radiation damage follows an epigenetic program similar to wound healing and other damage responses. Our hypothesis is that the persistent disruption of the microenvironment in irradiated tissue compromises its ability to suppress carcinogenesis.

116 citations


Journal ArticleDOI
TL;DR: While the mechanisms of action for most of these genes are not fully elucidated, some clues are emerging and are presented.
Abstract: Once cancer cells have spread and formed secondary masses, breast cancers are largely incurable even with state-of-the-art medicine. To improve diagnosis and therapy, better markers are needed to distinguish cells which have a high probability for causing clinically relevant, macroscopic metastases. In this review, we summarize the several genes that regulate breast cancer metastasis. Two categories of genes are presented--metastasis activator (ras, MEK1, mta1, proteinases, adhesion molecules, chemoattractants/receptors, autotaxin, PKC, S100A4, RhoC, osteopontin) and metastasis suppressor (Nm23, E-cadherin, TIMPs, KiSS1, Kai1, Maspin, MKK4, BRMS1). While the mechanisms of action for most of these genes are not fully elucidated, some clues are emerging and are presented.

104 citations


Journal ArticleDOI
TL;DR: Findings suggest that epigenetic changes might play a crucial role in gene inactivation in breast cancer, and inhibition of DNA methylation and histone deacetylation might be a therapeutic strategy in breast cancers, especially for those cancers with ER and PR negative phenotypes.
Abstract: DNA methylation is an epigenetic modification that is associated with transcriptional silencing of gene expression in mammalian cells. Hypermethylation of the promoter CpG islands contributes to the loss of gene function of several tumor related genes, including estrogen receptor a (ER) and progesterone receptor (PR). Gene expression patterns are also heavily influenced by changes in chromatin structure during transcription. Indeed both the predominant mammalian DNA methyltransferase (DNMTI), and the histone deacetylases (HDACs) play crucial roles in maintaining transcriptionally repressive chromatin by forming suppressive complexes at replication foci. These new findings suggest that epigenetic changes might play a crucial role in gene inactivation in breast cancer. Further, inhibition of DNA methylation and histone deacetylation might be a therapeutic strategy in breast cancer, especially for those cancers with ER and PR negative phenotypes.

Journal ArticleDOI
TL;DR: Despite compelling evidence that TGF-β has tumor suppressor activity in the mammary gland, neither T GF-β receptors nor Smads are genetically inactivated in human breast cancer, though receptor expression is reduced.
Abstract: Ligands of the TGF-β superfamily are unique in that they signal through transmembrane receptor serine-threonine kinases, rather than tyrosine kinases. The receptor complex couples to a signal transduction pathway involving a novel family of proteins, the Smads. On phosphorylation, Smads translocate to the nucleus where they modulate transcriptional responses. However, TGF-βs can also activate the mitogen-activated protein kinase (MAPK)4 pathway, and the different biological responses to TGF-β depend to varying degrees on activation of either or both of these two pathways. The Smad pathway is a nexus for cross-talk with other signal transduction pathways and for modulation by many different interacting proteins. Despite compelling evidence that TGF-β has tumor suppressor activity in the mammary gland, neither TGF-β receptors nor Smads are genetically inactivated in human breast cancer, though receptor expression is reduced. Possible reasons are discussed in relation to the dual role of TGF-β as tumor suppressor and oncogene.

Journal ArticleDOI
T. W. Keenan1
TL;DR: A brief historical account of research on milk fat globules, their surrounding membrane, and on aspects of the intracellular origin, growth, and secretion of milk lipid globules is presented.
Abstract: Most of the lipids in milk are triacylglycerols that occur in globules surrounded by a membrane derived from cellular membranes. This membrane, the milk-fat or milk-lipid globule membrane (MLGM),2 surrounds globules during the process of their secretion from the cell. The nature and cellular origin of the milk lipid globule membrane has been the subject of a considerable amount of research. Milk lipid globules originate as very small lipid droplets formed on or in the endoplasmic reticulum followed by release into the cytosol. These droplets consist of a triacylglycerol-rich core coated with a layer of proteins and polar lipids. How these droplets are formed, how they can grow in volume, how they move through the cell, and how they are secreted are questions that have been the basis for a number of investigations. While the general outlines of droplet formation, growth, movement, and secretion are known, virtually no molecular details of any of these processes have been elucidated. In this article I have presented a brief historical account of research on milk fat globules, their surrounding membrane, and on aspects of the intracellular origin, growth, and secretion of milk lipid globules. I have also attempted to call attention to those areas where further research is needed to gain a better understanding of the processes involved.

Journal ArticleDOI
TL;DR: It is now important to understand the mechanism of constitutive Stat activity and to develop strategies which will abrogate aberrant Stat signaling in tumors in vivo.
Abstract: Two members of the Stat family of transcription factors play a vital role in mouse mammary gland development. Stat5a was originally described as a regulator of milk protein gene expression and was subsequently shown to be essential for mammary development and lactogenesis. In contrast, Stat3 is an essential mediator of apoptosis and post-lactational regression. Other members of the Stat family may have specific, but as yet undemonstrated, functions in mammary development. However, since Stat1 activity is regulated during mammary development in a pattern different from Stats 3 and 5, this factor too may have a functional role. Although both Stat4 and Stat6 are expressed in mammary tissue, it seems unlikely that they will have a significant function as each of these Stats is activated in response to a limited number of cytokines. Given the essential regulatory roles of Stat signaling molecules in mammary development, it was not surprising to discover that constitutively activated Stat factors are a feature of human breast cancers. Sustained Stat activity has also been described in a variety of tumors including leukemias. The cause of this sustained activation is not clear but probably involves mutation of one of the many Stat regulatory proteins or dysregulation of other signaling pathways which modulate Stat activity. It is now important to understand the mechanism of constitutive Stat activity and to develop strategies which will abrogate aberrant Stat signaling in tumors in vivo.

Journal ArticleDOI
TL;DR: Functional studies utilizing both in vitro and in vivo models demonstrate that Ras signaling can regulate a variety of endpoints relevant to breast cancer progression, including anchorage dependent and independent growth, tumorigenesis, steroid sensitivity and invasion.
Abstract: The Ras superfamily of GTPases act as important regulatory switches to co-ordinate extracellular stimuli with activation of intracellular signaling pathways and appropriate biological responses. The Ras branch of this superfamily includes H-, K- and N-Ras, which are commonly mutated in particular human cancers, but notably not in those of the breast. Instead, in breast cancer the signaling pathways involving these GTPases may be upregulated due to increased coupling to growth factor receptors or other tyrosine kinases commonly overexpressed in this disease, or increased expression of regulators, the Ras protein itself, or downstream effectors. Functional studies utilizing both in vitro and in vivo models demonstrate that Ras signaling can regulate a variety of endpoints relevant to breast cancer progression, including anchorage dependent and independent growth, tumorigenesis, steroid sensitivity and invasion. Finally, analysis of the processing and signaling mechanisms of the Ras superfamily has identified potential targets for therapeutic intervention.

Journal ArticleDOI
TL;DR: Functionally, MUC4 is proposed to provide a protective mechanism for vulnerable epithelia, such as those of the airway, eye, female reproductive tract and mammary gland, and two mechanisms are proposed to contribute to the M UC4 functions.
Abstract: MUC4 is a one of the membrane mucins of the mucin gene (MUC) family, characterized by mucin tandem repeat domains and a transmembrane domain which associates it with the cell plasma membrane. Although MUC4 is encoded by a single gene, it is produced by epithelial cells as a heterodimer through a proteolytic cleavage mechanism. This heterodimer is found in both membrane and soluble forms associated with epithelia. Functionally, MUC4 is proposed to provide a protective mechanism for vulnerable epithelia, such as those of the airway, eye, female reproductive tract and mammary gland. The protective mechanism(s) may be highjacked by some carcinomas, such as those of the breast, to increase tumor progression. Two mechanisms are proposed to contribute to the MUC4 functions. First, MUC4 acts as an anti-adhesive or anti-recognition barrier at epithelial or tumor cell surfaces. Second, MUC4 can bind the receptor tyrosine kinase ErbB2 and alter its cellular signaling. Expression of MUC4 in mammary gland is repressed by posttranscriptional mechanisms involving basement membrane and TGF-β, which are relieved during pregnancy to permit secretion of MUC4 into milk. These mechanisms are also abrogated in some breast cancers, providing a scenario for promotion of tumor progression. These observations imply important functions for MUC4 in both normal mammary function and in breast cancer.

Journal ArticleDOI
TL;DR: Examining the repertoire of cadherins and catenins expressed by tumors may provide useful prognostic information for breast cancer.
Abstract: Cadherins are the transmembrane component of adherens junctions found between interacting cells in tissues. The cadherins bind cells to one another in a specific manner and link to the actin cytoskeleton through intracellular catenins. In addition to promoting strong cell-cell adhesion, cadherins appear to initiate and modify intracellular signaling pathways. The loss of E-cadherin function in epithelial cells is thought to be an important step in tumorigenesis. Moreover, anomalous expression of inappropriate cadherins in epithelial cells alters their behavior and may contribute to the tumorigenic phenotype. For breast cancer the decreased expression of E-cadherin alone may have limited value as a prognostic indicator; however, examining the repertoire of cadherins and catenins expressed by tumors may provide useful prognostic information.

Journal ArticleDOI
TL;DR: This review explores the mechanistic basis of breast carcinoma progression by focusing on the contribution of integrins, which are receptors for the laminin family of basement membrane components.
Abstract: This review explores the mechanistic basis of breast carcinoma progression by focusing on the contribution of integrins. Integrins are essential for progression not only for their ability to mediate physical interactions with extracellular matrices but also for their ability to regulate signaling pathways that control actin dynamics and cell movement, as well as for growth and survival. Our comments center on the α6 integrins (α6β1 and α6β4), which are receptors for the laminin family of basement membrane components. Numerous studies have implicated these integrins in breast cancer progression and have provided a rationale for studying the mechanistic basis of their contribution to aggressive disease. Recent work by our group and others on mechanisms of breast carcinoma invasion and survival that are influenced by the α6 integrins are discussed.

Journal ArticleDOI
TL;DR: The current state of knowledge of Wnt signal transduction in a range of model systems is summarized and the role of WNTs and Wnt signaling in mammary development and cancer is addressed.
Abstract: Wnt expression patterns during mammary development support a role for Wnts in breast development and in mammary epithelial responses to systemic hormones. The deregulation of Wnt signaling also plays a role in breast cancer. Activation of the Wnt signaling pathway is a major feature of several human neoplasias and appears to lead to the cytosolic stabilization of a transcriptional co-factor, β-catenin. This co-activator can then regulate transcription from a number of target genes including the cellular oncogenes cyclin D1 and c-myc. This review will summarize the current state of knowledge of Wnt signal transduction in a range of model systems and will then address the role of Wnts and Wnt signaling in mammary development and cancer.

Journal ArticleDOI
TL;DR: Proteoglycans serve to regulate the signal output of growth factor receptor tyrosine kinase and hence cell fate as well as the storage and diffusion of extracellular protein effectors and are active participants in mediating cell-cell and cell-matrix interactions.
Abstract: Proteoglycans consist of a core protein and an associated glycosaminoglycan (GAG)4 chain of heparan sulfate, chondroitin sulfate, dermatan sulfate or keratan sulfate, which are attached to a serine residue. The core proteins of cell surface proteoglycans may be transmembrane, e.g., syndecan, or GPI-anchored, e.g., glypican. Many different cell surface and matrix proteoglycan core proteins are expressed in the mammary gland and in mammary cells in culture. The level of expression of these core proteins, the structure of their GAG chains, and their degradation are regulated by many of the effectors that control the development and function of the mammary gland. Regulatory proteins of the mammary gland that bind GAG include many growth factors and morphogens (fibroblast growth factors, hepatocyte growth factor/scatter factor, members of the midkine family, wnts), matrix proteins (collagen, fibronectin, and laminin), enzymes (lipoprotein lipase) and microbial surface proteins. Structural diversity within GAG chains ensures that each protein-GAG interaction is as specific as necessary and a number of sequences of saccharides that recognize individual proteins have been elucidated. The GAG-protein interactions serve to regulate the signal output of growth factor receptor tyrosine kinase and hence cell fate as well as the storage and diffusion of extracellular protein effectors. In addition, GAGs clearly coordinate stromal and epithelial development, and they are active participants in mediating cell-cell and cell-matrix interactions. Since a single proteoglycan, even if it carries a single GAG chain, can bind multiple proteins, proteoglycans are also likely to act as multireceptors which promote the integration of cellular signals.

Journal ArticleDOI
TL;DR: Data suggesting that the ER may also play a pivotal role in the metastatic behavior of breast cancer is presented, and an argument that the up-regulation of ER and/or the selection of specific ER mutations are early events important for facilitating tumor progression is presented.
Abstract: It has long been appreciated that the estrogen receptor (ER) plays an important role in the biology of breast cancer. It is an accepted factor predicting favorable disease outcome and treatment response, and as such is generally considered to represent a "good" prognostic marker in breast cancer. In this review we present data suggesting that the ER may also play a pivotal role in the metastatic behavior of breast cancer, and present an argument that the up-regulation of ER and/or the selection of specific ER mutations are early events important for facilitating tumor progression. Thus, ER could serve dual roles in breast cancer, acting as a "bad" prognostic marker later in the disease process.

Journal ArticleDOI
TL;DR: Treatment of cancer patients with demethylating agents followed by retinoic acid may offer a new therapeutic modality and both the time of commencement of chemoprevention and the choice of substances that are able either to prevent de novo methylation or to reverse methylation-caused gene silencing may be important considerations.
Abstract: A growing body of evidence supports the hypothesis that the retinoic acid receptor beta2 (RAR-beta2) gene is a tumor suppressor gene which induces apoptosis and that the chemopreventive and therapeutic effects of retinoids are due to induction of RAR-beta2. During breast cancer progression, RAR-beta2 is reduced or even lost. It is known from studies of other tumor-suppressor genes that methylation of the 5'-region is the cause of loss of expression. Several groups demonstrated that this is also true for the RAR-beta2 in breast cancer by treating breast cancer cell lines with a demethylating agent and examining expression of the RAR-beta2 gene in response to a challenge with retinoic acid. Studies using sodium bisulfite genomic sequencing as well as methylation specific PCR showed that a number of breast cancer cell lines as well as breast cancer tissue showed signs of methylation. The RAR-beta2 gene was unmethylated in non-neoplastic breast tissue as well as in other normal tissues. A combination of retinoic acid with demethylating agents as well as with histone deacetylase inhibitors acts synergistically to inhibit growth. This review presents data that suggest that treatment of cancer patients with demethylating agents followed by retinoic acid may offer a new therapeutic modality. Both the time of commencement of chemoprevention and the choice of substances that are able either to prevent de novo methylation or to reverse methylation-caused gene silencing may be important considerations.

Journal ArticleDOI
TL;DR: Mammary glands of transgenic mice overexpressing the heparanase enzyme exhibit precocious branching of ducts and alveolar development, suggesting that the enzyme promotes normal morphogenesis and possibly pre-malignant changes in the mammary gland.
Abstract: Tumor spread involves degradation of various components of the extracellular matrix and blood vessel wall. Among these is heparan sulfate proteoglycan, which plays a key role in the self-assembly, insolubility and barrier properties of basement membranes and extracellular matrices. Expression of an endoglycosidase (heparanase) which degrades heparan sulfate correlates with the metastatic potential of tumor cells, and treatment with heparanase inhibitors markedly reduces the incidence of metastasis in experimental animals. Heparin-binding angiogenic proteins are stored as a complex with heparan sulfate in the microenvironment of tumors. These proteins are released and can induce new capillary growth when heparan sulfate is degraded by heparanase. Here, we describe the molecular properties, expression and involvement in tumor progression of a human heparanase. The enzyme is synthesized as a latent ∼65 kDa protein that is processed at the N-terminus into a highly active ∼50 kDa form. The heparanase mRNA and protein are preferentially expressed in metastatic human cell lines and in tumor biopsy specimens, including breast carcinoma. Overexpression of the heparanase cDNA in low-metastatic tumor cells conferred a high metastatic potential in experimental animals, resulting in an increased rate of mortality. The heparanase enzyme also released ECM-resident bFGF in vitro, and its overexpression elicited an angiogenic response in vivo. Heparanase may thus facilitate both tumor cell invasion and neovascularization, two critical steps in tumor progression. Mammary glands of transgenic mice overexpressing the heparanase enzyme exhibit precocious branching of ducts and alveolar development, suggesting that the enzyme promotes normal morphogenesis and possibly pre-malignant changes in the mammary gland.

Journal ArticleDOI
TL;DR: This review examines Cxs' involvement in breast cancer metastasis on the basis of the observations about Cx loss, and indicates that Cx expression is inversely correlated to metastatic potential.
Abstract: Gap junctional intercellular communication (GJIC) is a form of cell-cell communication mediating the exchange of small molecules between neighboring cells. Gap junctions (GJs) are formed by connexins (Cxs), and are subject to tight and dynamic regulation. They are involved in the cell cycle, differentiation, and cell signaling. The loss of Cxs and GJs is a hallmark of carcinogenesis, while their induction in cancer cells leads to a reversal of the cancer phenotype, induction of differentiation, and regulation of cell growth. On the basis of the observations about Cx loss in breast cancer, this review examines Cxs' involvement in breast cancer metastasis. Previous work indicates that Cx expression is inversely correlated to metastatic potential. This is probably because of the loss of cooperation between neighboring cells, leading to cell heterogeneity and cell dissociation in the tumor. The possible involvement of Cx activity during metastasis will be discussed.

Journal ArticleDOI
TL;DR: The complex biochemistry of DNA methylation—the molecular basis of most epigenetic regulation in mammalian genomes—is described and data connecting it to targeted modification and remodeling of chromatin structure is reviewed.
Abstract: Epigenetic regulation involves the maintenance of a particular state of gene expression--most commonly, repression--in the face of repeated mitosis, and frequently meiosis. Remarkably, changes in such heritable expression states occur without an alteration of the primary DNA sequence. We present a brief history of research in epigenetics, beginning with pioneering work in the 1950s by B. McClintock and R. A. Brink on maize kernel color inheritance. We describe the complex biochemistry of DNA methylation--the molecular basis of most epigenetic regulation in mammalian genomes--and review data connecting it to targeted modification and remodeling of chromatin structure. Several prominent examples of epigenetically regulated loci--X chromosome inactivation, imprinting, repetitive DNA silencing, and aberrant methylation patterns in neoplasia--are reviewed along with a description of our current understanding of the underlying molecular mechanisms. A common theme that emerges is the complex integration of epigenetic regulatory pathways with the chromatin infrastructure over target DNA loci.

Journal ArticleDOI
TL;DR: The similarities among hedgehog mutation-induced ductal dysplasias and human breast pathologies suggest a role for altered hedgehog signaling in the development of mammary cancer.
Abstract: Genetic analyses of two hedgehog signal transduction network genes, Patched-1 and Gli2, has demonstrated a critical role for hedgehog signaling in mediating epithelial-stromal tissue interactions during ductal development. Disruption of either gene leads to similar, yet distinct, defects in ductal morphogenesis. Defects are mainly ductal dysplasias that closely resemble some hyperplasias of the human breast. Phenotypic analyses have been coupled with in situ hybridization, transplantation and tissue recombination analyses to formulate a model for tissue compartment-specific control of mouse mammary gland development by hedgehog signaling. In addition, the similarities among hedgehog mutation-induced ductal dysplasias and human breast pathologies suggest a role for altered hedgehog signaling in the development of mammary cancer.

Journal ArticleDOI
TL;DR: PEA3 is the founding member of a subfamily of closely related ets genes that includes ER81 and ERM, and it is differentially expressed during postnatal mammary gland development and oncogenesis.
Abstract: PEA3 is the founding member of a subfamily of closely related ets genes that includes ER81 and ERM. PEA3 is expressed in the epithelial cells of mammary buds at the time that these first appear during mouse embryogenesis, and it is differentially expressed during postnatal mammary gland development. PEA3 expression is highest at the onset of puberty and during early pregnancy, times of extensive epithelial outgrowth and branching. PEA3 is expressed in undifferentiated epithelial cap cells of terminal end buds, and in differentiated myoepithelial cells of ducts and alveoli. Loss-of-function mutations in the PEA3 gene compromise mammary ductal branching at the onset of puberty and early during pregnancy. PEA3 is overexpressed in the vast majority of human breast tumors and in nearly all of the HER2-positive subclass of such tumors. PEA3 is similarly overexpressed in transgenic mouse models of this malignancy. Expression of dominant-negative PEA3 in the mouse mammary gland of MMTV-HER2 transgenic mice dramatically delays the onset and reduces the incidence of mammary tumors. Hence PEA3 and/or its close relatives play key regulatory roles in both mammary gland development and oncogenesis.

Journal ArticleDOI
TL;DR: The epigenetic changes that disrupt normal chromatin architecture and modify the expression of key genes in breast cancer cells represent vulnerabilities in the cancer cell that can be exploited as novel targets for new prevention and therapeutic strategies.
Abstract: The production of heritable changes in gene expression is the driving force in the development and progression of breast cancer. Such changes can result from mutations or from epigenetic events such as hypermethylation of DNA and hypoacetylation of histones. Histone acetylation and DNA methylation are major determinants of chromatin structure, and chromatin structure is a primary regulator of gene transcription. Cancer cells frequently contain both mutated genes and genes with altered expression due to one or more epigenetic mechanisms. This review describes the epigenetic changes that disrupt normal chromatin architecture and modify the expression of key genes in breast cancer cells. The structural integrity of the latter genes is usually intact, but their expression has been substantially altered due to methylation in their promoter region or deacetylation of histones that interact with their promoter region or both mechanisms. Genes affected by epigenetic changes in breast cancers include HoxA5, p21WAF, gelsolin, BRCA1, BRCA2, E-cadherin, steroid hormone receptors, and retinoic acid receptor II. Because these epigenetic modifications are usually reversible by treatment with certain drugs, they represent vulnerabilities in the cancer cell that can be exploited as novel targets for new prevention and therapeutic strategies.

Journal ArticleDOI
TL;DR: A methylation event that occurs during the propagation of human mammary epithelial cells (HMEC) in culture is described and the accompanying genetic alterations that have been observed are described.
Abstract: The genomic changes that foster cancer can be either genetic or epigenetic in nature Early studies focused on genetic changes and how mutational events contribute to changes in gene expression These point mutations, deletions and amplifications are known to activate oncogenes and inactivate tumor suppressor genes More recently, multiple epigenetic changes that can have a profound effect on carcinogenesis have been identified These epigenetic events, such as the methylation of promoter sequences in genes, are under active investigation In this review we will describe a methylation event that occurs during the propagation of human mammary epithelial cells (HMEC) in culture and detail the accompanying genetic alterations that have been observed

Journal ArticleDOI
Nancy E. Hynes, Heidi Lane1
TL;DR: The role of Myc as a downstream effector of the ErbB2 receptor tyrosine kinase which is overexpressed and constitutively activate in many mammary tumors is discussed and data from the laboratory will be presented showing that deregulated Erb B2 activity strongly stimulates cytoplasmic signaling pathways which impinge on Myc at multiple levels causing its deregulated expression.
Abstract: The proto-oncogene c-myc encodes a transcription factor which plays a major role in the regulation of normal cellular proliferation and is aberrantly expressed in many breast tumors. In a normal cell Myc expression levels are tightly regulated being subject to many layers of control. Errantly expressed Myc collaborates with other oncogenes to promote transformation. In this review we will focus on the association between abnormal Myc expression and mammary cancer. In particular, we will discuss the role of Myc as a downstream effector of the ErbB2 receptor tyrosine kinase which is overexpressed and constitutively activate in many mammary tumors. The cooperation between Myc and ErbB2 in transformation will be discussed in relation to clinical studies on Myc in human cancer and with consideration of transgenic models of Myc-induced mammary cancer. Data from our laboratory will be presented showing that deregulated ErbB2 activity strongly stimulates cytoplasmic signaling pathways which in turn impinge on Myc at multiple levels causing its deregulated expression.