Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 61(19-20): 2523-34, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15526159

ABSTRACT

Defining the key players in normal breast differentiation is instrumental to understanding how morphogenesis becomes defective during breast cancer progression. During the past 2 decades much effort has been devoted to the development of technologies for purification and expansion of primary human breast cells in culture and optimizing a relevant microenvironment, which may help to define the niche that regulates breast differentiation and morphogenesis. In contrast to the general property of cancer, normal human cells have a finite lifespan. After a defined number of population doublings, normal cells enter an irreversible proliferation-arrested state referred to as replicative senescence. To overcome this obstacle for continuous long-term studies, replicative senescence can be bypassed by treatment of cells with chemical agents such as benzopyrene, by radiation or by transfection with viral oncogenes or the gene for human telomerase (human telomerase reverse transcriptase, hTERT). A drawback of some of these protocols is a concurrent introduction of chromosomal changes, which sometimes leads to a transformed phenotype and selection of a subpopulation, which may not be representative of the tissue of origin. In recent years, we have sought to establish immortalized primary breast cells, which retain crucial characteristics of their original in situ tissue pattern. This review discusses various approaches to immortalization of breast-derived epithelial and stromal cells and the application of such cell lines for studies on human breast morphogenesis.


Subject(s)
Breast/cytology , Breast/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence , DNA-Binding Proteins , Humans , Neoplasms/metabolism , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Papillomavirus E7 Proteins , Repressor Proteins/metabolism , Telomerase/metabolism
2.
Breast Cancer Res ; 3(4): 213-7, 2001.
Article in English | MEDLINE | ID: mdl-11434871

ABSTRACT

The human breast comprises three lineages: the luminal epithelial lineage, the myoepithelial lineage, and the mesenchymal lineage. It has been widely accepted that human breast neoplasia pertains only to the luminal epithelial lineage. In recent years, however, evidence has accumulated that neoplastic breast epithelial cells may be substantially more plastic in their differentiation repertoire than previously anticipated. Thus, along with an increasing availability of markers for the myoepithelial lineage, at least a partial differentiation towards this lineage is being revealed frequently. It has also become clear that conversions towards the mesenchymal lineage actually occur, referred to as epithelial to mesenchymal transitions. Indeed, some of the so-called myofibroblasts surrounding the tumor may have an epithelial origin rather than a mesenchymal origin. Because myoepithelial cells, epithelial to mesenchymal transition-derived cells, genuine stromal cells and myofibroblasts share common markers, we now need to define a more ambitious set of markers to distinguish these cell types in the microenvironment of the tumors. This is necessary because the different microenvironments may confer different clinical outcomes. The aim of this commentary is to describe some of the inherent complexities in defining cellular phenotypes in the microenvironment of breast cancer and to expand wherever possible on the implications for tumor suppression and progression.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation , Cell Transformation, Neoplastic , Biomarkers, Tumor , Epithelial Cells/pathology , Female , Humans , Mesoderm/pathology , Phenotype
3.
Mol Biol Cell ; 11(4): 1357-67, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10749935

ABSTRACT

To identify genes misregulated in the final stages of breast carcinogenesis, we performed differential display to compare the gene expression patterns of the human tumorigenic mammary epithelial cells, HMT-3522-T4-2, with those of their immediate premalignant progenitors, HMT-3522-S2. We identified a novel gene, called anti-zuai-1 (AZU-1), that was abundantly expressed in non- and premalignant cells and tissues but was appreciably reduced in breast tumor cell types and in primary tumors. The AZU-1 gene encodes an acidic 571-amino-acid protein containing at least two structurally distinct domains with potential protein-binding functions: an N-terminal serine and proline-rich domain with a predicted immunoglobulin-like fold and a C-terminal coiled-coil domain. In HMT-3522 cells, the bulk of AZU-1 protein resided in a detergent-extractable cytoplasmic pool and was present at much lower levels in tumorigenic T4-2 cells than in their nonmalignant counterparts. Reversion of the tumorigenic phenotype of T4-2 cells, by means described previously, was accompanied by the up-regulation of AZU-1. In addition, reexpression of AZU-1 in T4-2 cells, using viral vectors, was sufficient to reduce their malignant phenotype substantially, both in culture and in vivo. These results indicate that AZU-1 is a candidate breast tumor suppressor that may exert its effects by promoting correct tissue morphogenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Genes, Tumor Suppressor/genetics , Tumor Suppressor Proteins , Amino Acid Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Blotting, Northern , Blotting, Western , Breast Neoplasms , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Epithelial Cells/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Molecular Sequence Data , Precancerous Conditions , Protein Structure, Tertiary , RNA, Neoplasm/analysis , Sequence Alignment , Tumor Cells, Cultured
4.
Cancer Res ; 59(7 Suppl): 1757-1763s; discussion 1763s-1764s, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10197593

ABSTRACT

Because every cell within the body has the same genetic information, a significant problem in biology is to understand how cells within a tissue express genes selectively. A sophisticated network of physical and biochemical signals converge in a highly orchestrated manner to bring about the exquisite regulation that governs gene expression in diverse tissues. Thus, the ultimate decision of a cell to proliferate, express tissue-specific genes, or apoptose must be a coordinated response to its adhesive, growth factor, and hormonal milieu. The unifying hypothesis examined in this overview is that the unit of function in higher organisms is neither the genome nor the cell alone but the complex, three-dimensional tissue. This is because there are bidirectional connections between the components of the cellular microenvironment (growth factors, hormones, and extracellular matrix) and the nucleus. These connections are made via membrane-bound receptors and transmitted to the nucleus, where the signals result in modifications to the nuclear matrix and chromatin structure and lead to selective gene expression. Thus, cells need to be studied "in context", i.e., within a proper tissue structure, if one is to understand the bidirectional pathways that connect the cellular microenvironment and the genome. In the last decades, we have used well-characterized human and mouse mammary cell lines in "designer microenvironments" to create an appropriate context to study tissue-specific gene expression. The use of a three-dimensional culture assay, developed with reconstituted basement membrane, has allowed us to distinguish normal and malignant human breast cells easily and rapidly. Whereas normal cells become growth arrested and form organized "acini," tumor cells continue to grow, pile up, and in general fail to respond to extracellular matrix and microenvironmental cues. By correcting the extracellular matrix-receptor (integrin) signaling and balance, we have been able to revert the malignant phenotype when a human breast tumor cell is cultured in, or on, a basement membrane. Most recently, we have shown that whereas beta1 integrin and epidermal growth factor receptor signal transduction pathways are integrated reciprocally in three-dimensional cultures, on tissue culture plastic (two-dimensional monolayers), these are not coordinated. Finally, we have demonstrated that, rather than passively reflecting changes in gene expression, nuclear organization itself can modulate cellular and tissue phenotype. We conclude that the structure of the tissue is dominant over the genome, and that we may need a new paradigm for how epithelial-specific genes are regulated in vivo. We also argue that unless the structure of the tissue is critically altered, malignancy will not progress, even in the presence of multiple chromosomal mutations.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , Gene Expression Regulation/physiology , Animals , Breast Neoplasms/pathology , Cell Nucleus/pathology , Cells, Cultured , Female , Humans , Mammary Neoplasms, Experimental/genetics , Phenotype , Reference Values
5.
Dev Biol ; 206(1): 88-99, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9918697

ABSTRACT

The origin of the epithelial and myoepithelial cells in the human breast has not been delineated. In this study we have addressed whether luminal epithelial cells and myoepithelial cells are vertically connected, i.e., whether one is the precursor for the other. We used a primary culture assay allowing preservation of basic phenotypic traits of luminal epithelial and myoepithelial cells in culture. The two cell types were then separated immunomagnetically using antibodies directed against lineage-specific cell surface antigens into at best 100% purity. The cellular identity was ascertained by cytochemistry, immunoblotting, and 2-D gel electrophoresis. Luminal epithelial cells were identified by strong expression of cytokeratins 18 and 19 while myoepithelial cells were recognized by expression of vimentin and alpha-smooth muscle actin. We used a previously devised culture medium (CDM4) that allows vigorous expansion of proliferative myoepithelial cells and also devised a medium (CDM6) that allowed sufficient expansion of differentiated luminal epithelial cells based on addition of hepatocyte growth factor/scatter factor. The two different culture media supported each lineage for at least five passages without signs of interconversion. We used parallel cultures where we switched culture media, thus testing the ability of each lineage to convert to the other. Whereas the myoepithelial lineage showed no signs of interconversion, a subset of luminal epithelial cells, gradually, but distinctly, converted to myoepithelial cells. We propose that in the mature human breast, it is the luminal epithelial cell compartment that gives rise to myoepithelial cells rather than the other way around.


Subject(s)
Breast/growth & development , Stem Cells/metabolism , Actins/metabolism , Breast/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Culture Media/chemistry , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunohistochemistry , Keratins/metabolism , Microscopy, Phase-Contrast , Vimentin/metabolism
6.
Proc Natl Acad Sci U S A ; 95(25): 14711-6, 1998 Dec 08.
Article in English | MEDLINE | ID: mdl-9843954

ABSTRACT

What determines the nuclear organization within a cell and whether this organization itself can impose cellular function within a tissue remains unknown. To explore the relationship between nuclear organization and tissue architecture and function, we used a model of human mammary epithelial cell acinar morphogenesis. When cultured within a reconstituted basement membrane (rBM), HMT-3522 cells form polarized and growth-arrested tissue-like acini with a central lumen and deposit an endogenous BM. We show that rBM-induced morphogenesis is accompanied by relocalization of the nuclear matrix proteins NuMA, splicing factor SRm160, and cell cycle regulator Rb. These proteins had distinct distribution patterns specific for proliferation, growth arrest, and acini formation, whereas the distribution of the nuclear lamina protein, lamin B, remained unchanged. NuMA relocalized to foci, which coalesced into larger assemblies as morphogenesis progressed. Perturbation of histone acetylation in the acini by trichostatin A treatment altered chromatin structure, disrupted NuMA foci, and induced cell proliferation. Moreover, treatment of transiently permeabilized acini with a NuMA antibody led to the disruption of NuMA foci, alteration of histone acetylation, activation of metalloproteases, and breakdown of the endogenous BM. These results experimentally demonstrate a dynamic interaction between the extracellular matrix, nuclear organization, and tissue phenotype. They further show that rather than passively reflecting changes in gene expression, nuclear organization itself can modulate the cellular and tissue phenotype.


Subject(s)
Cell Nucleus/ultrastructure , Extracellular Matrix/ultrastructure , Morphogenesis , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation , Humans , Nuclear Proteins/genetics , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 95(25): 14821-6, 1998 Dec 08.
Article in English | MEDLINE | ID: mdl-9843973

ABSTRACT

Anchorage and growth factor independence are cardinal features of the transformed phenotype. Although it is logical that the two pathways must be coregulated in normal tissues to maintain homeostasis, this has not been demonstrated directly. We showed previously that down-modulation of beta1-integrin signaling reverted the malignant behavior of a human breast tumor cell line (T4-2) derived from phenotypically normal cells (HMT-3522) and led to growth arrest in a three-dimensional (3D) basement membrane assay in which the cells formed tissue-like acini (14). Here, we show that there is a bidirectional cross-modulation of beta1-integrin and epidermal growth factor receptor (EGFR) signaling via the mitogen-activated protein kinase (MAPK) pathway. The reciprocal modulation does not occur in monolayer (2D) cultures. Antibody-mediated inhibition of either of these receptors in the tumor cells, or inhibition of MAPK kinase, induced a concomitant down-regulation of both receptors, followed by growth-arrest and restoration of normal breast tissue morphogenesis. Cross-modulation and tissue morphogenesis were associated with attenuation of EGF-induced transient MAPK activation. To specifically test EGFR and beta1-integrin interdependency, EGFR was overexpressed in nonmalignant cells, leading to disruption of morphogenesis and a compensatory up-regulation of beta1-integrin expression, again only in 3D. Our results indicate that when breast cells are spatially organized as a result of contact with basement membrane, the signaling pathways become coupled and bidirectional. They further explain why breast cells fail to differentiate in monolayer cultures in which these events are mostly uncoupled. Moreover, in a subset of tumor cells in which these pathways are misregulated but functional, the cells could be "normalized" by manipulating either pathway.


Subject(s)
Basement Membrane/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Integrin beta1/metabolism , Basement Membrane/ultrastructure , Breast , Cell Line , Epithelial Cells/cytology , ErbB Receptors/ultrastructure , Female , Humans , Integrin beta1/ultrastructure , Protein Binding , Protein Conformation
8.
Adv Cancer Res ; 75: 135-61, 1998.
Article in English | MEDLINE | ID: mdl-9709809

ABSTRACT

In this brief review, the development of breast cancer is discussed from the vantage of phenotypic differentiation, similar to what has been considered over the years for leukemias and melanomas, both of which express easily visible differentiation markers (Hart and Easty, 1991; Clarke et al., 1995; Lynch, 1995; Sachs, 1996; Sledge, 1996). The review is divided into a theoretical background for human breast differentiation and a discussion of recent experimental results in our laboratories with differentiation of breast epithelial cells. In the theoretical background, in situ markers of differentiation of normal breast and carcinomas are discussed with emphasis on their possible implications for tumor therapy. So far, most of the emphasis regarding differentiation therapy of tumors has been focused on the possible action of soluble factors, such as colony-stimulating factors in leukemias and retinoic acids in solid tumors (Lotan, 1996; Sachs, 1996). However, an emerging and promising new avenue in this area appears to point to additional factors, such as the cellular form and extracellular matrix (ECM) (Bissel et al., 1982; Bissel and Barcellos-Hoff, 1987; Ingber, 1992). The recent interest in these parameters has evolved along with an increasing understanding of the molecular composition of the ECM, and of the molecular basis of the classical findings that normal cell--in contrast to tumor cells--are anchorage dependent for survival and growth (Folkman and Moscona, 1978; Hannigan et al., 1996). We now know that this is the case for epithelial as well as fibroblastic cells, and that interaction with ECM is crucial for such regulation. Indeed, ECM and integrins are emerging as the central regulators of differentiation, apoptosis, and cancer (Boudreau et al., 1995; Boudreau and Bissel, 1996; Werb et al., 1996; Bissell, 1997; Weaver, et al., 1997). In the experimental part, we elaborate on our own recent experiments with functional culture models of the human breast, with particular emphasis on how "normal" and cancer cells could be defined within a reconstituted ECM. Special attention is given to integrins, the prominent ECM receptors. We further discuss a number of recent experimental results, all of which point to the same conclusion: namely that phenotypic reversion toward a more normal state for epithelial tumors is no longer an elusive goal. Thus "therapy by differentiation" could be broadened to include not only blood-borne tumors, but also solid tumors of epithelial origin.


Subject(s)
Breast Neoplasms/pathology , Basement Membrane/metabolism , Biomarkers, Tumor , Cell Differentiation/physiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Integrins/metabolism , Receptors, Estrogen/metabolism , Tumor Cells, Cultured
9.
J Cell Biol ; 137(1): 231-45, 1997 Apr 07.
Article in English | MEDLINE | ID: mdl-9105051

ABSTRACT

In a recently developed human breast cancer model, treatment of tumor cells in a 3-dimensional culture with inhibitory beta1-integrin antibody or its Fab fragments led to a striking morphological and functional reversion to a normal phenotype. A stimulatory beta1-integrin antibody proved to be ineffective. The newly formed reverted acini re-assembled a basement membrane and re-established E-cadherin-catenin complexes, and re-organized their cytoskeletons. At the same time they downregulated cyclin D1, upregulated p21(cip,wat-1), and stopped growing. Tumor cells treated with the same antibody and injected into nude mice had significantly reduced number and size of tumors in nude mice. The tissue distribution of other integrins was also normalized, suggesting the existence of intimate interactions between the different integrin pathways as well as adherens junctions. On the other hand, nonmalignant cells when treated with either alpha6 or beta4 function altering antibodies continued to grow, and had disorganized colony morphologies resembling the untreated tumor colonies. This shows a significant role of the alpha6/beta4 heterodimer in directing polarity and tissue structure. The observed phenotypes were reversible when the cells were disassociated and the antibodies removed. Our results illustrate that the extracellular matrix and its receptors dictate the phenotype of mammary epithelial cells, and thus in this model system the tissue phenotype is dominant over the cellular genotype.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Breast Neoplasms/therapy , Integrin beta1/immunology , Animals , Antigens, CD/genetics , Basement Membrane/chemistry , Basement Membrane/cytology , Binding, Competitive/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Division/physiology , Extracellular Matrix/chemistry , Female , Fluorescent Antibody Technique , Genotype , Humans , Immunoglobulin Fab Fragments/pharmacology , Integrin beta1/genetics , Integrin beta4 , Mice , Phenotype , Rats , Signal Transduction/physiology , Transformation, Genetic/physiology , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology
10.
Electrophoresis ; 17(11): 1776-80, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8982611

ABSTRACT

Actins comprise six isoforms of which the nonmuscle isoforms beta-/gamma-actins are expressed by all eukaryotic cells. The expression pattern of one of the muscle actin isoforms, alpha-sm actin, previously believed to be restricted to smooth muscle, has been broadened to encompass activated fibroblasts (myofibroblasts) as well. The significance of this molecular conversion has remained largely unknown. We have recently shown that a reduction in filamentous alpha-sm actin by electroinjected specific antibodies or antisense oligodeoxynucleotides leads to increased motility in breast myofibroblasts (Rønnov-Jessen, L., Petersen, O. W. J. Cell Biol. 1996, 134, 67-80). In the present study we have expanded on the functional significance of actin isotypes in fibroblasts from the opposite point of view, namely filamentous nonmuscle actin. Nonmuscle actins in fibroblasts and myofibroblasts were ADP-ribosylated by Clostridium botulinum C2 toxin. The substrate for C2 toxin is globular actin, which upon ribosylation cannot incorporate into microfilaments. The pattern of actin ADP-ribosylation in (myo)fibroblasts in the presence of [32P]NAD was analyzed by isoelectric focusing, fluorography and immunoblotting. The influence of C2 toxin on microfilaments in intact cells was further assessed by immunofluorescence, and motility was measured in a mass migration assay and by computerized video time-lapse microscopy. We show here that C2 toxin specifically ribosylates beta- and gamma-actin in both fibroblasts and myofibroblasts. Whereas fibroblasts rapidly round up and stop migrating when filamentous beta-/gamma-actin is reduced by short-term ADP-ribosylation, myofibroblasts maintain their flattened morphology and a basic low motility.


Subject(s)
Actin Cytoskeleton/drug effects , Actins/metabolism , Botulinum Toxins/pharmacology , Fibroblasts/drug effects , Poly(ADP-ribose) Polymerases/pharmacology , Protein Processing, Post-Translational/drug effects , Actin Cytoskeleton/ultrastructure , Blotting, Western , Breast/cytology , Cell Movement/drug effects , Female , Glycosylation/drug effects , Humans , Isoelectric Focusing , Microscopy, Fluorescence , Muscles/cytology
11.
J Cell Biol ; 134(1): 67-80, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8698823

ABSTRACT

Actins are known to comprise six mammalian isoforms of which beta- and gamma-nonmuscle actins are present in all cells, whereas alpha-smooth muscle (alpha-sm) actin is normally restricted to cells of the smooth muscle lineages. alpha-Sm actin has been found also to be expressed transiently in certain nonmuscle cells, in particular fibroblasts, which are referred to as myofibroblasts. The functional significance of alpha-sm actin in fibroblasts is unknown. However, myofibroblasts appear to play a prominent role in stromal reaction in breast cancer, at the site of wound repair, and in fibrotic reactions. Here, we show that the presence of alpha-sm actin is a signal for retardation of migratory behavior in fibroblasts. Comparison in a migration assay of fibroblast cell strains with and without alpha-sm actin revealed migratory restraint in alpha-sm actin-positive fibroblasts. Electroporation of monoclonal antibody (mAb) 1A4, which recognizes specifically the NH2-terminal Ac-EEED sequence of alpha-sm actin, significantly increased the frequency of migrating cells over that obtained with an unrelated antibody or a mAb against beta-actin. Time-lapse video microscopy revealed migratory rates of 4.8 and 3.0 microns/h, respectively. To knock out the alpha-sm actin protein, several antisense phosphorothioate oligodeoxynucleotide (ODNs) were tested. One of these, 3'UTI, which is complementary to a highly evolutionary conserved 3' untranslated (3'UT) sequence of alpha-sm actin mRNA, was found to block alpha-sm actin synthesis completely without affecting the synthesis of any other proteins as analyzed by two-dimensional gel electrophoresis. Targeting by antisense 3'UTI significantly increased motility compared with the corresponding sense ODN. alpha-Sm actin inhibition also led to the formation of less prominent focal adhesions as revealed by immunofluorescence staining against vinculin, talin, and beta1-integrin. We propose that an important function of filamentous alpha-sm actin is to immobilize the cells.


Subject(s)
Actins/physiology , Cell Movement , Fibroblasts/physiology , Actin Cytoskeleton/ultrastructure , Base Sequence , Breast/cytology , DNA, Antisense/chemistry , Electrophoresis, Gel, Two-Dimensional , Extracellular Matrix/physiology , Female , Fluorescent Antibody Technique, Indirect , Humans , Molecular Sequence Data , Muscle, Smooth/chemistry , Video Recording
12.
Mol Cell Endocrinol ; 119(1): 47-59, 1996 May 17.
Article in English | MEDLINE | ID: mdl-8793853

ABSTRACT

Estrogens play an important role in breast cancer and the effect of estrogen on growth of breast cancer cells has been extensively studied. However, only little information is available about the response of normal breast epithelial cells to estrogen, mainly due to the difficulties in establishing estrogen receptor (ER)-positive human breast epithelial cells in culture. We have stably transfected the human estrogen receptor (hER) wt cDNA into the ER-negative, spontaneously immortalized human breast epithelial cell line, HMT-3522S1, in order to develop a model for studying the effect of estrogen on nonmalignant human breast epithelial cells. Characterization of the transfected clone F9 confirmed incorporation of the estrogen receptor gene in the genome, expression of hER mRNA and hER protein. However, proliferation of F9 cells was inhibited by both estradiol (E2) and tamoxifen, whereas the pure antiestrogen ICI 182,780 had no effect on cell proliferation. This seems paradoxical since E2 stimulated the expression of the endogenous genes, TGF-alpha, cathepsin D, and alpha1-antitrypsin. In breast cancer cell lines, high expression of these genes is correlated to estrogen-stimulated cell proliferation. The spontaneously immortalized HMT-3522S1 cells transfected with wt ER cDNA behave similarly to cell lines from nonmalignant breast tissue immortalized by carcinogens and transfected with mutated ER cDNA as described by others. The discrepancy between growth inhibition and induction of positive growth factors by E2 indicates that either ER-positive nonmalignant breast epithelial cells are growth-inhibited by E2 in contrast to malignant cells or that introduction of the ER into ER-negative cells is not sufficient for restoring "normal' estrogen responsiveness.


Subject(s)
Gene Expression Regulation , Receptors, Estrogen/genetics , Animals , Breast/cytology , Breast/metabolism , Cell Division , Cell Line , DNA, Complementary , Epithelial Cells , Epithelium/metabolism , Estradiol/metabolism , Female , Humans , Mice , Receptors, Estrogen/metabolism , Transfection , Tumor Cells, Cultured
13.
Cancer Res ; 56(9): 2039-44, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8616848

ABSTRACT

We have reported previously on the first spontaneously immortalized, nonmalignant human breast epithelial cell line, HMT-3522, which is entirely dependent on exogenous epidermal growth factor (EGF). In passage 118, cells were adapted to grow in medium without EGF and a new growth-transformed subline, HMT-3522/gt-1, was generated and propagated at high growth rate without exogenous EGF (Madsen et al., Cancer Res., 52: 1210-1217, 1992). Here we have used this subline and the continuum of the parent line, HMT-3522/wt, to pose the question whether a relevant change in a physiological parameter of the microenvironment will induce malignant transformation. The two cell lines were cultured under identical conditions with the only exception that EGF was omitted in the medium for gt-1. Initially, wt and gt-1 were identical in terms of karyotype as well as morphology, growth rate, and protein expression as revealed by two-dimensional gel electrophoresis. A highly dramatic shift to phenotype was observed in passage 238 when the gt-1 line became tumorigenic in nude mice. After two mouse-culture passages, the resulting malignantly transformed cell line (HMT-3522/mt-1) was refractory to the growth-modulating effect of EGF and presented an extra copy of a chromosome marker, 7q-, as the only cytogenetic difference from the gt-1. Our results suggest that microenvironmental cues are powerful factors in the induction of malignancy. A major role of EGF receptor in the malignant transformation is emphasized by loss of EGF sensitivity and acquisition of an extra chromosome 7p harboring the EGF receptor gene. We hypothesize that during premalignant hyperplasia, a population of EGF/transforming growth factor alpha autonomous epithelial cells in situ may develop as a consequence of local transforming growth factor alpha deprivation, a condition reflected in the culture model as autonomy after EGF withdrawal.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/drug effects , Chromosomes, Human, Pair 7 , Epidermal Growth Factor/pharmacology , Trisomy , Animals , Breast Neoplasms/pathology , Cell Division/drug effects , Culture Media , Humans , Immunoenzyme Techniques , Karyotyping , Mice , Tumor Cells, Cultured
14.
Physiol Rev ; 76(1): 69-125, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8592733

ABSTRACT

Reciprocal interactions between epithelium and mesenchyme mediate crucial aspects of embryonic development and direct the coordinated organogenesis, correct spatial orientation, and the timely expression of functional activity consistent with physiological demands. The mesenchymal equivalent in the adult organism is the stroma, i.e., the loose connective tissue that is separated from the epithelial compartment by an intact basement membrane. In carcinomas, the cellular organization is dramatically changed, and the stroma is extensively modified. The basement membrane is penetrated in a process of degradation and/or decreased synthesis, and direct contact between tumor cells and the surrounding stroma coincides with neovascularization, inflammatory cell influx, and extensive remodeling of extracellular matrix. In this review, we highlight our current knowledge of tumor cell stromal interactions in the mammary gland with particular emphasis on cellular origins and functional phenotypes. We focus both on normal mammary gland and breast tumors and on culture systems developed to dissect individual aspects of cell-cell and cell-extracellular matrix interactions.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Cell Transformation, Neoplastic , Stromal Cells/pathology , Animals , Breast/cytology , Breast Neoplasms/metabolism , Humans
15.
Oncogene ; 10(11): 2125-34, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7784057

ABSTRACT

To explore regulation and function of cyclin D2, a candidate cell cycle-regulatory proto-oncogene, we examined subcellular localisation, cell type- and cell cycle-dependent expression, and requirement of cyclin D2 protein for G1 progression, in a panel of 40 human normal and cancer cell types. Except for lymphoid cells and sarcoma cell lines, expression of cyclin D2 was considerably more restricted than that of cyclin D1, whereas both D-type cyclin proteins were low or undetectable in cells lacking functional retinoblastoma gene product. In G1 cells, the cyclin D2 protein was more resistant to extraction and localised predominantly to nuclei, whereas it became more soluble and distributed in both nuclei and cytoplasm from G1/S transition onwards. Centrifugal elutriation and multiparameter flow cytometry analyses of several cell types showed moderate cell cycle oscillation with maximum levels of the cyclin D2 protein reached in late G1. Microinjection and/or electroporation of antibodies to cyclin D2 during G1 arrested the cyclin D2-expressing lymphocytes, breast myoepithelium, and U-2-OS sarcoma cells in G1 phase, whereas cyclin D2-negative cell types were unaffected by such treatment. Consistent with the putative proto-oncogenic role of cyclin D2 in specific cell types, our data show that this G1 cyclin has properties closely resembling those of cyclin D1, including the essential positive role in regulation of G1.


Subject(s)
Cyclins/physiology , G1 Phase/physiology , Nucleoproteins/physiology , Animals , Antibodies, Monoclonal/immunology , Cell Line , Cyclin D1 , Cyclin D2 , Cyclins/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Nucleoproteins/immunology , Oncogene Proteins/physiology , Proto-Oncogene Mas , Subcellular Fractions/metabolism , Tumor Cells, Cultured
16.
Semin Cancer Biol ; 6(3): 175-84, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7495986

ABSTRACT

Normal mammary homeostasis, and by implication tumorigenesis, are dependent upon the dynamic interplay between epithelial cells, stromal components and the extracellular matrix. To study the evolution of human breast cancer, a functionally relevant cell culture model is required which recognizes the complexity of the mammary gland's microenvironment. The development of an appropriate breast epithelial cancer cell model will be dependent on the ability to recreate the 'normal' and 'neoplastic' tissue microenvironment in culture. Towards this goal, a 3-dimensional extracellular matrix (ECM) assay, employing a reconstituted basement membrane, has been developed which allows for the rapid and accurate discrimination of normal and neoplastic cells when cultured. To investigate stromal/epithelial cell interactions, we have developed a tumor environment assay which essentially mirrors the tumor microenvironment histologically. The use of a novel, near diploid, human breast epithelial cell line, HMT-3522, which has transformed spontaneously with passage in culture, together with these 3-dimensional culture assays is expected to provide meaningful markers of initiation and progression.


Subject(s)
Breast Neoplasms/pathology , Tumor Cells, Cultured/pathology , Breast Neoplasms/genetics , Disease Progression , Humans
17.
J Cell Sci ; 108 ( Pt 5): 1945-57, 1995 May.
Article in English | MEDLINE | ID: mdl-7544798

ABSTRACT

We previously established a rapid three-dimensional assay for discrimination of normal and malignant human breast epithelial cells using a laminin-rich reconstituted basement membrane. In this assay, normal epithelial cells differentiate into well-organized acinar structures whereas tumor cells fail to recapitulate this process and produce large, disordered colonies. The data suggest that breast acinar morphogenesis and differentiation is regulated by cell-extra-cellular matrix (ECM) interactions and that these interactions are altered in malignancy. Here, we investigated the role of ECM receptors (integrins) in these processes and report on the expression and function of potential laminin receptors in normal and tumorigenic breast epithelial cells. Immunocytochemical analysis showed that normal and carcinoma cells in a three-dimensional substratum express profiles of integrins similar to normal and malignant breast tissues in situ. Normal cells express alpha 1, alpha 2, alpha 3, alpha 6, beta 1 and beta 4 integrin subunits, whereas breast carcinoma cells show variable losses, disordered expression, or downregulation of these subunits. Function-blocking experiments using inhibitory anti-integrin subunit antibodies showed a > 5-fold inhibition of the formation of acinar structures by normal cells in the presence of either anti-beta 1 or anti-alpha 3 antibodies, whereas anti-alpha 2 or -alpha 6 had little or no effect. In experiments where collagen type I gels were used instead of basement membrane, acinar morphogenesis was blocked by anti-beta 1 and -alpha 2 antibodies but not by anti-alpha 3. These data suggest a specificity of integrin utilization dependent on the ECM ligands encountered by the cell. The interruption of normal acinar morphogenesis by anti-integrin antibodies was associated with an inhibition of cell growth and induction of apoptosis. Function-blocking antibodies had no inhibitory effect on the rate of tumor cell growth, survival or capacity to form colonies. Thus under our culture conditions breast acinar formation is at least a two-step process involving beta 1-integrin-dependent cellular growth followed by polarization of the cells into organized structures. The regulation of this pathway appears to be impaired or lost in the tumor cells, suggesting that tumor colony formation occurs by independent mechanisms and that loss of proper integrin-mediated cell-ECM interaction may be critical to breast tumor formation.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Carcinoma/pathology , Integrins/physiology , Neoplasm Proteins/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/physiology , Basement Membrane , Breast/metabolism , Breast Neoplasms/metabolism , Carcinoma/metabolism , Carcinoma, Ehrlich Tumor , Cell Differentiation , Cell Division , Cell Survival , Collagen , Epithelium/metabolism , Extracellular Matrix/physiology , Female , Humans , Integrin beta1 , Integrins/antagonists & inhibitors , Integrins/immunology , Laminin , Mice , Mice, Inbred C57BL , Morphogenesis , Receptors, Laminin/physiology
18.
J Clin Invest ; 95(2): 859-73, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7532191

ABSTRACT

The origin of myofibroblasts in stromal reaction has been a subject of controversy. To address this question definitively, we developed techniques for purification and characterization of major stromal cell types. We defined a panel of markers that could, in combination, unequivocally distinguish these cell types by immunocytochemistry, iso-electric focusing, immunoblotting, and two-dimensional gel electrophoresis. We then devised an assay to recapitulate in culture, within two weeks of incubation, critical aspects of the microenvironment in vivo including the typical tissue histology and stromal reaction. When confronted with tumor cells in this assay, fibroblasts readily converted into a graded pattern of myogenic differentiation, strongest in the immediate vicinity of tumor cells. Vascular smooth muscle cells (VSMC), in contrast, did not change appreciably and remained coordinately smooth muscle differentiated. Midcapillary pericytes showed only a slight propensity for myogenic differentiation. Analysis of ten primary tumors implicated converted fibroblasts (10/10), vascular smooth muscle cells (4/10), and pericytes (1/10) in the stromal reaction. Tumor cells were shown to specifically denude the venules both in culture and in vivo, explaining the VSMC phenotype in the stroma. The establishment of this assay and clarification of the origin of these cells pave the way for further analysis of the mechanisms of conversion, and of the consequence of such heterogeneity for diagnosis and treatment.


Subject(s)
Blood Vessels/pathology , Breast Neoplasms/pathology , Biomarkers/analysis , Breast/cytology , Breast/pathology , Breast Neoplasms/blood supply , Cell Separation/methods , Chondroitin Sulfates/analysis , Cytoskeletal Proteins/analysis , Epithelium/pathology , Factor VIII/analysis , Fibroblasts/pathology , HLA-DQ Antigens/analysis , Humans , Immunoblotting , Immunoenzyme Techniques , Immunohistochemistry , Integrins/analysis , Keratins/analysis , Organoids/pathology , Organoids/ultrastructure , Tumor Cells, Cultured
19.
J Natl Cancer Inst ; 86(24): 1838-44, 1994 Dec 21.
Article in English | MEDLINE | ID: mdl-7990158

ABSTRACT

BACKGROUND: We have developed a culture system using reconstituted basement membrane components in which normal human mammary epithelial cells exhibit several aspects of the development and differentiation process, including formation of acinar-like structures, production and basal deposition of basement membrane components, and production and apical secretion of sialomucins. Cell lines and cultures from human breast carcinomas failed to recapitulate this process. The data indicate the importance of cellular interactions with the basement membrane in the regulation of normal breast differentiation and, potentially, its loss in neoplasia. PURPOSE: Our purpose was to use this assay to investigate the role of the putative metastasis suppressor gene nm23-H1 in mammary development and differentiation. METHODS: The metastatic human breast carcinoma cell line MDA-MB-435, clones transfected with a control pCMVBamneo vector, and clones transfected with pCMVBamneo vector containing nm23-H1 complementary DNA (the latter of which exhibited a substantial reduction in spontaneous metastatic potential in vivo) were cultured within a reconstituted basement membrane. Clones were examined for formation of acinus-like spheres, deposition of basement membrane components, production of sialomucin, polarization, and growth arrest. RESULTS: In contrast to the parental cell line and control transfectants, MDA-MB-435 breast carcinoma cells overexpressing Nm23-H1 protein regained several aspects of the normal phenotype within reconstituted basement membrane. Nm23-H1 protein-positive cells formed organized acinus-like spheres, deposited the basement membrane components type IV collagen and, to some extent, laminin to the outside of the spheres, expressed sialomucin, and growth arrested. Growth arrest of Nm23-H1 protein-positive cells was preceded by and correlated with formation of a basement membrane, suggesting a causal relationship. CONCLUSION: The data indicate a previously unidentified cause-and-effect relationship between nm23-H1 gene expression and morphological-biosynthetic-growth aspects of breast differentiation in this model system. IMPLICATIONS: While the basement membrane microenvironment is capable of directing the differentiation of normal human breast cells, neoplastic transformation abrogates this relationship, suggesting that intrinsic cellular events are also critical to this process. The data identify nm23-H1 gene expression as one of these events, suggesting an important role in the modulation of cellular responsiveness to the microenvironment. The data also identify previously unknown growth inhibitory effects of nm23-H1 gene overexpression.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase , Transcription Factors/physiology , Up-Regulation/genetics , Animals , Basement Membrane/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , NM23 Nucleoside Diphosphate Kinases , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...