1
|
Golias C, Batistatou A, Bablekos G, Charalabopoulos A, Peschos D, Mitsopoulos P, Charalabopoulos K. Physiology and pathophysiology of selectins, integrins, and IgSF cell adhesion molecules focusing on inflammation. A paradigm model on infectious endocarditis. ACTA ACUST UNITED AC 2011; 18:19-32. [PMID: 21892874 DOI: 10.3109/15419061.2011.606381] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The development of adhesion bonds, either among cells or among cells and components of the extracellular matrix, is a crucial process. These interactions are mediated by some molecules collectively known as adhesion molecules (CAMs). CAMs are ubiquitously expressed proteins playing a central role in controlling cell migration, proliferation, survival, and apoptosis. Besides their key function in physiological maintenance of tissue integrity, CAMs play an eminent role in various pathological processes such as cardiovascular disorders, atherogenesis, atherosclerotic plaque progression and regulation of the inflammatory response. CAMs such as selectins, integrins, and immunoglobulin superfamily take part in interactions between leukocyte and vascular endothelium (leukocyte rolling, arrest, firm adhesion, migration). Experimental data and pathologic observations support the assumption that pathogenic microorganisms attach to vascular endothelial cells or sites of vascular injury initiating intravascular infections. In this review a paradigm focusing on cell adhesion molecules pathophysiology and infective endocarditis development is given.
Collapse
Affiliation(s)
- Christos Golias
- Department of Physiology, Medical Faculty, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | | | | | | | | |
Collapse
|
2
|
Küppers M, Ittrich C, Faust D, Dietrich C. The transcriptional programme of contact-inhibition. J Cell Biochem 2010; 110:1234-43. [PMID: 20564218 DOI: 10.1002/jcb.22638] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Proliferation of non-transformed cells is regulated by cell-cell contacts, which are referred to as contact-inhibition. Vice versa, transformed cells are characterised by a loss of contact-inhibition. Despite its generally accepted importance for cell-cycle control, little is known about the intracellular signalling pathways involved in contact-inhibition. Unravelling the molecular mechanisms of contact-inhibition and its loss during tumourigenesis will be an important step towards the identification of novel target genes in tumour diagnosis and treatment. To better understand the underlying molecular mechanisms we identified the transcriptional programme of contact-inhibition in NIH3T3 fibroblast using high-density microarrays. Setting the cut off: >or=1.5-fold, P <or= 0.05, 853 genes and 73 cDNA sequences were differentially expressed in confluent compared to exponentially growing cultures. Importing these data into GenMAPP software revealed a comprehensive list of cell-cycle regulatory genes mediating G0/G1 arrest, which was confirmed by RT-PCR and Western blot. In a narrow analysis (cut off: >or=2-fold, P <or= 0.002), we found 110 transcripts to be differentially expressed representing 107 genes and 3 cDNA sequences involved, for example, in proliferation, signal transduction, transcriptional regulation, cell adhesion and communication. Interestingly, the majority of genes was upregulated indicating that contact-inhibition is not a passive state, but actively induced. Furthermore, we confirmed differential expression of eight genes by semi-quantitative RT-PCR and identified the potential tumour suppressor transforming growth factor-beta (TGF-beta)-1-induced clone 22 (TSC-22; tgfb1i4) as a novel protein to be induced in contact-inhibited cells.
Collapse
Affiliation(s)
- Monika Küppers
- Institute of Toxicology, Medical Center of the Johannes Gutenberg-University, Obere Zahlbacherstr 67, 55131 Mainz, Germany
| | | | | | | |
Collapse
|
3
|
Dietrich C, Kaina B. The aryl hydrocarbon receptor (AhR) in the regulation of cell-cell contact and tumor growth. Carcinogenesis 2010; 31:1319-28. [PMID: 20106901 PMCID: PMC6276890 DOI: 10.1093/carcin/bgq028] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 01/21/2010] [Accepted: 01/24/2010] [Indexed: 01/26/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor, which is activated by a large group of environmental pollutants including polycyclic aromatic hydrocarbons, dioxins and planar polychlorinated biphenyls. Ligand binding leads to dimerization of the AhR with aryl hydrocarbon receptor nuclear translocator and transcriptional activation of several xenobiotic phase I and phase II metabolizing enzymes, such as cytochrome P4501A1 and glutathione-S-transferase, respectively. Since phase I enzymes convert inert carcinogens to active genotoxins, the AhR plays a key role in tumor initiation. Besides this classical route, the AhR mediates tumor promotion and recent evidence suggests that the AhR also plays a role in tumor progression. To date, no mechanistic link could be established between the canonical pathway involving xenobiotic metabolism and AhR-dependent tumor promotion and progression. A hallmark of tumor promotion is unbalanced proliferation, whereas tumor progression is characterized by dedifferentiation, increased motility and metastasis of tumor cells. Tumor progression and presumably also tumor promotion are triggered by loss of cell-cell contact. Cell-cell contact is known to be a critical regulator of proliferation, differentiation and cell motility in vitro and in vivo. Increasing evidence suggests that activation of the AhR may lead to deregulation of cell-cell contact, thereby inducing unbalanced proliferation, dedifferentiation and enhanced motility. In line with this is the finding of increased AhR expression and malignancy in some animal and human cancers. Here, we summarize our current knowledge on non-canonical AhR-driven pathways being involved in deregulation of cell-cell contact and discuss the data with respect to tumor initiation, promotion and progression.
Collapse
Affiliation(s)
- Cornelia Dietrich
- Institute of Toxicology, Medical Center of the Johannes Gutenberg-University, Mainz, Germany.
| | | |
Collapse
|
4
|
A 3D hybrid model for tissue growth: the interplay between cell population and mass transport dynamics. Biophys J 2009; 97:401-14. [PMID: 19619455 DOI: 10.1016/j.bpj.2009.03.067] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 03/08/2009] [Accepted: 03/27/2009] [Indexed: 11/21/2022] Open
Abstract
To provide theoretical guidance for the design and in vitro cultivation of bioartificial tissues, we have developed a multiscale computational model that can describe the complex interplay between cell population and mass transport dynamics that governs the growth of tissues in three-dimensional scaffolds. The model has three components: a transient partial differential equation for the simultaneous diffusion and consumption of a limiting nutrient; a cellular automaton describing cell migration, proliferation, and collision; and equations that quantify how the varying nutrient concentration modulates cell division and migration. The hybrid discrete-continuous model was parallelized and solved on a distributed-memory multicomputer to study how transport limitations affect tissue regeneration rates under conditions encountered in typical bioreactors. Simulation results show that the severity of transport limitations can be estimated by the magnitude of two dimensionless groups: the Thiele modulus and the Biot number. Key parameters including the initial seeding mode, cell migration speed, and the hydrodynamic conditions in the bioreactor are shown to affect not only the overall rate, but also the pattern of tissue growth. This study lays the groundwork for more comprehensive models that can handle mixed cell cultures, multiple nutrients and growth factors, and other cellular processes, such as cell death.
Collapse
|
5
|
Okada M, Wang Y, Jang SW, Tang X, Neri LM, Ye K. Akt phosphorylation of merlin enhances its binding to phosphatidylinositols and inhibits the tumor-suppressive activities of merlin. Cancer Res 2009; 69:4043-51. [PMID: 19351837 PMCID: PMC2810124 DOI: 10.1158/0008-5472.can-08-3931] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The NF2 tumor suppressor gene encodes an intracellular membrane-associated protein, called merlin, which belongs to the band 4.1 family of cytoskeleton-associated proteins that link cell surface glycoproteins to the actin cytoskeleton. Merlin suppresses phosphatidylinositol 3-kinase (PI3K)/Akt signaling by directly binding and inhibiting the stimulatory activity of PIKE-L on PI3K. Akt feeds back and phosphorylates merlin and provokes its polyubiquitination and degradation. Here, we show that Akt phosphorylation and PI(3,4,5)P(3) binding mediate the tumor-suppressive activity of merlin. The extreme NH(2) terminus of merlin directly interacts with phosphatidylinositols, for which the unfolded conformation is required. Moreover, Akt phosphorylation enhances merlin binding affinity to phosphatidylinositols and inhibits its proapoptotic actions. Furthermore, Akt phosphorylation and phosphatidylinositols increase merlin binding to CD44. Epidermal growth factor treatment and Akt phosphorylation provoke merlin to aggregate in the ruffled plasma membrane and promote cell migration. Thus, these results suggest that PI3K signaling regulates the tumor-suppressive activity of merlin via both Akt phosphorylation and phosphatidylinositol lipids binding to merlin.
Collapse
Affiliation(s)
| | | | | | | | - Luca M. Neri
- Dipartimento di Morfologia ed Embriologia, Universita’ di Ferrara, Ferrara, Italy
| | | |
Collapse
|
6
|
Pino V, Ramsauer VP, Salas P, Carothers Carraway CA, Carraway KL. Membrane Mucin Muc4 Induces Density-dependent Changes in ERK Activation in Mammary Epithelial and Tumor Cells. J Biol Chem 2006; 281:29411-20. [PMID: 16891313 DOI: 10.1074/jbc.m604858200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The membrane mucin Muc4 has been shown to alter cellular behavior through both anti-adhesive effects on cell-cell and cell-extracellular matrix interactions and its ability to act as an intramembrane ligand for the receptor tyrosine kinase ErbB2. The ERK pathway is regulated by both cell-matrix and cell-cell adhesion. An analysis of the effects of Muc4 expression on ERK phosphorylation in mammary tumor and epithelial cells, which exhibit both adhesion-dependent growth and contact inhibition of growth, showed that the effects are density dependent, with opposing effects on proliferating cells and contact-inhibited cells. In these cells, cell-matrix interactions through integrins are required for activation of the ERK mitogenesis pathway. However, cell-cell interactions via cadherins inhibit the ERK pathway. Expression of Muc4 reverses both of these effects. In contact-inhibited cells, Muc4 appears to activate the ERK pathway at the level of Raf-1; this activation does not depend on Ras activation. The increase in ERK activity correlates with an increase in cyclin D(1) expression in these cells. This abrogation of contact inhibition is dependent on the number of mucin repeats in the mucin subunit of Muc4, indicative of an anti-adhesive effect. The mechanism by which Muc4 disrupts contact inhibition involves a Muc4-induced relocalization of E-cadherin from adherens junctions at the lateral membrane of the cells to the apical membrane. Muc4-induced abrogation of contact inhibition may be an important mechanism by which tumors progress from an early, more benign state to invasiveness.
Collapse
Affiliation(s)
- Vanessa Pino
- Departments of Cell Biology and Anatomy and Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida 33101, USA
| | | | | | | | | |
Collapse
|
7
|
Cheng G, Youssef BB, Markenscoff P, Zygourakis K. Cell population dynamics modulate the rates of tissue growth processes. Biophys J 2005; 90:713-24. [PMID: 16299082 PMCID: PMC1367098 DOI: 10.1529/biophysj.105.063701] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The development and testing of a discrete model describing the dynamic process of tissue growth in three-dimensional scaffolds is presented. The model considers populations of cells that execute persistent random walks on the computational grid, collide, and proliferate until they reach confluence. To isolate the effect of population dynamics on tissue growth, the model assumes that nutrient and growth factor concentrations remain constant in space and time. Simulations start either by distributing the seed cells uniformly and randomly throughout the scaffold, or from an initial condition designed to simulate the migration and cell proliferation phase of wound healing. Simulations with uniform seeding show that cell migration enhances tissue growth by counterbalancing the adverse effects of contact inhibition. This beneficial effect, however, diminishes and disappears completely for large migration speeds. By contrast, simulations with the "wound" seeding mode show a continual enhancement of tissue regeneration rates with increasing cell migration speeds. We conclude that cell locomotory parameters and the spatial distribution of seed cells can have profound effects on the dynamics of the process and, consequently, on the pattern and rates of tissue growth. These results can guide the design of experiments for testing the effectiveness of biomimetic modifications for stimulating tissue growth.
Collapse
Affiliation(s)
- Gang Cheng
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77251-1892, USA
| | | | | | | |
Collapse
|
8
|
Nair KS, Naidoo R, Chetty R. Expression of cell adhesion molecules in oesophageal carcinoma and its prognostic value. J Clin Pathol 2005; 58:343-51. [PMID: 15790695 PMCID: PMC1770622 DOI: 10.1136/jcp.2004.018036] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Oesophageal carcinoma remains a disease of poor prognosis. Surgical cure rates are compromised by the fact that most patients are diagnosed at a late stage of disease because of the delayed onset of symptoms, by which time metastases and organ infiltration may have already occurred. Thus, invasion and metastases play a key role in influencing patient survival, and the search for novel treatments may therefore hinge on gaining insight into the mechanisms controlling these processes. It has been established that the initial step in the metastatic cascade is the detachment of tumour cells from the primary tumour via dysregulation of normal cell-cell and cell-matrix interactions. Distinct proteins known as cell adhesion molecules (CAMs) mediate these interactions. In recent years, a plethora of information has contributed to the in depth understanding of these molecules. This review provides a brief description of five families of CAMs (cadherins, integrins, CD44, immunoglobulin superfamily, and selectins) and highlights their altered expression in relation both to prognosis and tumour behaviour in squamous cell carcinoma and adenocarcinoma of the oesophagus.
Collapse
Affiliation(s)
- K S Nair
- Pfizer Molecular Biology Research Facility, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of Natal, Congella 4013, Durban, South Africa
| | | | | |
Collapse
|
9
|
Motti ML, Califano D, Baldassarre G, Celetti A, Merolla F, Forzati F, Napolitano M, Tavernise B, Fusco A, Viglietto G. Reduced E-cadherin expression contributes to the loss of p27kip1-mediated mechanism of contact inhibition in thyroid anaplastic carcinomas. Carcinogenesis 2005; 26:1021-34. [PMID: 15718252 DOI: 10.1093/carcin/bgi050] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In the present study, we have characterized several human thyroid cancer cell lines of different histotypes for their responsiveness to contact inhibition. We found that cells derived from differentiated carcinoma (TPC-1, WRO) arrest in G(1) phase at confluence, whereas cells derived from anaplastic carcinoma (ARO, FRO and FB1) continue to grow after reaching confluence. Furthermore, we provide experimental evidence that the axis, E-cadherin/beta-catenin/p27(Kip1), represents an integral part of the regulatory mechanism that controls proliferation at a high cell density, whose disruption may play a key role in determining the clinical behaviour of thyroid cancer. This conclusion derives from the finding that: (i) the expression of p27(Kip1) is enhanced at high cell density only in cells responsive to contact inhibition (TPC-1, WRO), but not in contact-inhibition resistant cells (ARO, FRO or FB1 cells); (ii) the increase in p27(Kip1) also resulted in increased levels of p27(Kip1) bound to cyclin E-Cdk2 complex, a reduction in cyclin E-Cdk2 activity and dephosphorylation of the retinoblastoma protein; (iii) antisense inhibition of p27(Kip1) upregulation at high cell density in confluent-sensitive cells completely prevents the confluence-induced growth arrest; (iv) proper expression and/or membrane localization of E-cadherin is observed only in cells responsive to contact inhibition (TPC-1, NPA, WRO) but not in unresponsive cells (ARO, FRO or FB1); (v) disruption of E-cadherin-mediated cell-cell contacts at high cell density induced by an anti-E-cadherin neutralizing antibody, inhibits the induction of p27(kip1) and restores proliferation in contact-inhibited cells; (vi) re-expression of E-cadherin into cells unresponsive to contact inhibition (ARO, FB1) induces a p27(kip1) expression and growth arrest. In summary, our data indicate that the altered response to contact inhibition exhibited by thyroid anaplastic cancer cells is due to the failure to upregulate p27(Kip1) in response to cell-cell interactions.
Collapse
Affiliation(s)
- Maria Letizia Motti
- Dipartimento di Biologia e Patologia Cellulare e Molecolare L.Califano Facoltà di Medicina e Chirurgia, Università Federico II, via S. Pansini 5, 80131, Napoli, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Samuel S, Bernstein LR. Adhesion, migration, transcriptional, interferon-inducible, and other signaling molecules newly implicated in cancer susceptibility and resistance of JB6 cells by cDNA microarray analyses. Mol Carcinog 2003; 39:34-60. [PMID: 14694446 DOI: 10.1002/mc.10163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Relative expression levels of 9500 genes were determined by cDNA microarray analyses in mouse skin JB6 cells susceptible (P+) and resistant (P-) to 12-O-tetradecanoyl phorbol-13 acetate (TPA)-induced neoplastic transformation. Seventy-four genes in 6 functional classes were differentially expressed: (I) extracellular matrix (ECM) and basement membrane (BM) proteins (20 genes). P+ cells express higher levels than P- cells of several collagens and proteases, and lower levels of protease inhibitors. Multiple genes encoding adhesion molecules are expressed preferentially in P- cells, including six genes implicated in axon guidance and adhesion. (II) Cytoskeletal proteins (13 genes). These include actin isoforms and regulatory proteins, almost all preferentially expressed in P- cells. (III) Signal transduction proteins (12 genes). Among these are Ras-GTPase activating protein (Ras-GAP), the deleted in oral cancer-1 and SLIT2 tumor suppressors, and connexin 43 (Cx43) gap junctional protein, all expressed preferentially in P- cells. (IV) Interferon-inducible proteins (3 genes). These include interferon-inducible protein (IFI)-16, an Sp1 transcriptional regulator expressed preferentially in P- cells. (V) Other transcription factors (4 genes). Paired related homeobox gene 2 (Prx2)/S8 homeobox, and retinoic acid (RA)-regulated nur77 and cellular retinoic acid-binding protein II (CRABPII) transcription factors are expressed preferentially in P- cells. The RIN-ZF Sp-transcriptional suppressor exhibits preferential P+ expression. (VI) Genes of unknown functions (22 sequences). Numerous mesenchymal markers are expressed in both cell types. Data for multiple genes were confirmed by real-time PCR. Overall, 26 genes were newly implicated in cancer. Detailed analyses of the functions of the genes and their interrelationships provided converging evidence for their possible roles in implementing genetic programs mediating cancer susceptibility and resistance. These results, in conjunction with cell wounding and phalloidin staining data, indicated that concerted genetic programs were implemented that were conducive to cell adhesion and tumor suppression in P- cells and that favored matrix turnover, cell motility, and abrogation of tumor suppression in P+ cells. Such genetic programs may in part be orchestrated by Sp-, RA-, and Hox-transcriptional regulatory pathways implicated in this study.
Collapse
Affiliation(s)
- Shaija Samuel
- Department of Pathology and Laboratory Medicine, Texas A & M University System Health Science Center, College Station, Texas, USA
| | | |
Collapse
|
11
|
Husmark J, Heldin NE, Nilsson M. Follicular growth of a thyroid carcinoma cell line (KAT-4) with abnormal E-cadherin and impaired epithelial barrier. Thyroid 2002; 12:781-90. [PMID: 12481943 DOI: 10.1089/105072502760339343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Loss of the epithelial phenotype is a well-established phenomenon during progression of carcinomas to a more malignant state. In the present study, we describe a human thyroid tumor cell line (KAT-4), established from a poorly differentiated carcinoma, which displays exceptional features. In culture, the KAT-4 cells had a fast proliferation rate that was not restricted by high cell density, resulting in multilayered growth. Unexpectedly, the cells expressed normal levels of epithelial markers, e.g., cytokeratin, occludin, and E-cadherin, showed apical-basolateral polarization of the plasma membrane including microvilli and junction complexes, and formed intercellular lumens resembling thyroid follicles. Yet, when grown on filter, the cells were unable to establish a tight paracellular barrier. Moreover, E-cadherin expressed at the cell surface consisted of two peptides with abnormal size (135 and 95 kd, respectively) as compared to mature E-cadherin (120 kd) in nonneoplastic thyrocytes. Northern blot analysis and examination of immunoreactivity, glycosylation, and catenin binding suggested that E-cadherin was aberrant because of altered posttranscriptional processing. Thus, the KAT-4 thyroid carcinoma cell line has a unique phenotype, with maintained epithelial morphology despite dysfunctioning tight junctions, abnormal E-cadherin, and loss of contact-inhibited growth, that is not previously identified in other wild-type tumor cell lines.
Collapse
Affiliation(s)
- Johanna Husmark
- Institute of Anatomy and Cell Biology, Göteborg University, Göteborg, Sweden
| | | | | |
Collapse
|
12
|
Potla L, Boghaert ER, Armellino D, Frost P, Damle NK. Reduced expression of EphrinA1 (EFNA1) inhibits three-dimensional growth of HT29 colon carcinoma cells. Cancer Lett 2002; 175:187-95. [PMID: 11741747 DOI: 10.1016/s0304-3835(01)00613-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ephrin A1 (EFNA1) is a GPI-anchored ligand that preferentially binds to the receptor tyrosine kinase, EphA2. EphA2 is over-expressed in malignant melanocytes and in prostate carcinoma cells. Whether activation of EphA2 by EFNA1 is involved in aberrant growth or differentiation of cancer cells is currently not known. We studied the effect of reducing EFNA1 on the growth of a colon carcinoma cell line (HT29). HT29 cells were transfected with EFNA1 antisense yielding clones that expressed less than 25% of EFNA1 found in vector controls. EFNA1-antisense transfectants grew slower than controls when cultured as three-dimensional spheroids. When grown as monolayers, the transfectants had a similar doubling time of the vector controls. These results indicated that autocrine stimulation of EphA2 by EFNA1 could trigger an indirect growth signal by overcoming 'contact inhibition'. Following addition of EFNA1-Fc to HT29 cells, tyrosine hyperphosphorylation of EphA2, E-cadherin, and beta-catenin were observed. Because the function of E-cadherin is associated with contact inhibition of HT29 cells, phosphorylation of E-cadherin and beta-catenin by activation of EphA1 is one possible mechanism by which HT29 cells alleviate contact inhibition.
Collapse
Affiliation(s)
- Lyka Potla
- Oncology/Immunology Division, Wyeth-Ayerst Research, 200/4604, 401 North Middletown Road, Pearl River, NY 10965-1299, USA
| | | | | | | | | |
Collapse
|
13
|
Morrison H, Sherman LS, Legg J, Banine F, Isacke C, Haipek CA, Gutmann DH, Ponta H, Herrlich P. The NF2 tumor suppressor gene product, merlin, mediates contact inhibition of growth through interactions with CD44. Genes Dev 2001; 15:968-80. [PMID: 11316791 PMCID: PMC312675 DOI: 10.1101/gad.189601] [Citation(s) in RCA: 399] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The neurofibromatosis-2 (NF2) gene encodes merlin, an ezrin-radixin-moesin-(ERM)-related protein that functions as a tumor suppressor. We found that merlin mediates contact inhibition of growth through signals from the extracellular matrix. At high cell density, merlin becomes hypo-phosphorylated and inhibits cell growth in response to hyaluronate (HA), a mucopolysaccharide that surrounds cells. Merlin's growth-inhibitory activity depends on specific interaction with the cytoplasmic tail of CD44, a transmembrane HA receptor. At low cell density, merlin is phosphorylated, growth permissive, and exists in a complex with ezrin, moesin, and CD44. These data indicate that merlin and CD44 form a molecular switch that specifies cell growth arrest or proliferation.
Collapse
Affiliation(s)
- H Morrison
- Forschungszentrum Karlsruhe, Institute of Toxicology and Genetics, and University of Karlsruhe, Institute of Genetics, 76021 Karlsruhe, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Corvera S, DiBonaventura C, Shpetner HS. Cell confluence-dependent remodeling of endothelial membranes mediated by cholesterol. J Biol Chem 2000; 275:31414-21. [PMID: 10903311 DOI: 10.1074/jbc.m001708200] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The plasma membranes of endothelial cells reaching confluence undergo profound structural and functional modifications, including the formation of adherens junctions, crucial for the regulation of vascular permeability and angiogenesis. Adherens junction formation is accompanied by the tyrosine dephosphorylation of adherens junctions proteins, which has been correlated with the strength and stability of adherens junctions. Here we show that cholesterol is a critical determinant of plasma membrane remodeling in cultures of growing cow pulmonary aortic endothelial cells. Membrane cholesterol increased dramatically at an early stage in the formation of confluent cow pulmonary aortic endothelial cell monolayers, prior to formation of intercellular junctions. This increase was accompanied by the redistribution of caveolin from a high density to a low density membrane compartment, previously shown to require cholesterol, and increased binding of the annexin II-p11 complex to membranes, consistent with other studies indicating cholesterol-dependent binding of annexin II to membranes. Furthermore, partial depletion of cholesterol from confluent cells with methyl-beta-cyclodextrin both induced tyrosine phosphorylation of multiple membrane proteins, including adherens junctions proteins, and disrupted adherens junctions. Both effects were dramatically reduced by prior complexing of methyl-beta-cyclodextrin with cholesterol. Our results reveal a novel physiological role for cholesterol regulating the formation of adherens junctions and other plasma membrane remodeling events as endothelial cells reach confluence.
Collapse
Affiliation(s)
- S Corvera
- Program in Molecular Medicine and Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | |
Collapse
|
15
|
Tatsuno I, Hirai A, Saito Y. Cell-anchorage, cell cytoskeleton, and Rho-GTPase family in regulation of cell cycle progression. PROGRESS IN CELL CYCLE RESEARCH 2000; 4:19-25. [PMID: 10740811 DOI: 10.1007/978-1-4615-4253-7_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It has been well known that cell-anchorage and the cell cytoskeleton are deeply involved in the regulation of cell proliferation and cell cycle. However, the precise molecular mechanism involved in cell-anchorage and the cell cytoskeleton have remained be to elucidated. The recent great volume of information regarding cell cycle regulators such as cyclin, cyclin-dependent kinases (CDKs) and CDK inhibitors (CKI) has facilitated the understanding of the cell cycle in mammalian cells. In this review, we will focus on these cell cycle regulators to discuss the regulation of cell proliferation controlled by cell-anchorage and the cytoskeleton, and especially the roles of Rho family GTPases.
Collapse
Affiliation(s)
- I Tatsuno
- Second Department of Internal Medicine, Chiba University School of Medicine, Japan
| | | | | |
Collapse
|
16
|
Abstract
Neural cell adhesion molecules (CAMs) of the immunoglobulin superfamily nucleate and maintain groups of cells at key sites during early development and in the adult. In addition to their adhesive properties, binding of CAMs can affect intracellular signaling. Their ability to influence developmental events, including cell migration, proliferation, and differentiation can therefore result both from their adhesive as well as their signaling properties. This review focuses on the two CAMs for which the most information is known, the neural CAM, N-CAM, and L1. N-CAM was the first CAM to be characterized and, therefore, has been studied extensively. The binding of N-CAM to cells leads to a number of signaling events, some of which result in changes in gene expression. Interest in L1 derives from the fact that mutations in its gene lead to human genetic diseases including mental retardation. Much is known about modifications of the L1 cytoplasmic domain and its interaction with cytoskeletal molecules. The study of CAM signaling mechanisms has been assay-dependent rather than molecule-dependent, with particular emphasis on assays of neurite outgrowth and gene expression, an emphasis that is maintained throughout the review. The signals generated following CAM binding that lead to alterations in cell morphology and gene expression have been linked directly in only a few cases. We also review information on other CAMs, giving special consideration to those that are anchored in the membrane by a phospholipid anchor. These proteins, including a form of N-CAM, are presumed to be localized in lipid rafts, membrane substructures that include distinctive subsets of cytoplasmic signaling molecules such as members of the src-family of nonreceptor protein tyrosine kinases. In the end, these studies may reveal that what CAMs do after they bind cells together may have as profound consequences for the cells as the adhesive interactions themselves. This area will therefore remain a rich ground for future studies.
Collapse
Affiliation(s)
- K L Crossin
- Department of Neurobiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | |
Collapse
|
17
|
Prévôt D, Voeltzel T, Birot AM, Morel AP, Rostan MC, Magaud JP, Corbo L. The leukemia-associated protein Btg1 and the p53-regulated protein Btg2 interact with the homeoprotein Hoxb9 and enhance its transcriptional activation. J Biol Chem 2000; 275:147-53. [PMID: 10617598 DOI: 10.1074/jbc.275.1.147] [Citation(s) in RCA: 121] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BTG1 and BTG2 belong to a family of functionally related genes involved in the control of the cell cycle. As part of an ongoing attempt to understand their biological functions, we used a yeast two-hybrid screening to look for possible functional partners of Btg1 and Btg2. Here we report the physical and functional association between these proteins and the homeodomain protein Hoxb9. We further show that Btg1 and Btg2 enhance Hoxb9-mediated transcription in transfected cells, and we report the formation of a Hoxb9.Btg2 complex on a Hoxb9-responsive target, and the fact that this interaction facilitates the binding of Hoxb9 to DNA. The transcriptional activity of the Hoxb9.Btg complex is essentially dependent on the activation domain of Hoxb9, located in the N-terminal portion of the protein. Our data indicate that Btg1 and Btg2 act as transcriptional cofactors of the Hoxb9 protein, and suggest that this interaction may mediate their antiproliferative function.
Collapse
Affiliation(s)
- D Prévôt
- Unité INSERM U453, Centre Léon Bérard, 69373 Lyon Cedex 08, France
| | | | | | | | | | | | | |
Collapse
|
18
|
Sasaki T, Endo T. Evidence for the presence of N‐CAM 180 on astrocytes from rat cerebellum and differences in glycan structures between N‐CAM 120 and N‐CAM 140. Glia 1999. [DOI: 10.1002/(sici)1098-1136(199912)28:3<236::aid-glia7>3.0.co;2-#] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Tasuku Sasaki
- Department of Glycobiology, Tokyo Metropolitan Institute of Gerontology, Sakaecho, Itabashi‐ku, Tokyo, Japan
| | - Tamao Endo
- Department of Glycobiology, Tokyo Metropolitan Institute of Gerontology, Sakaecho, Itabashi‐ku, Tokyo, Japan
| |
Collapse
|
19
|
Fiore M, Degrassi F. Dimethyl sulfoxide restores contact inhibition-induced growth arrest and inhibits cell density-dependent apoptosis in hamster cells. Exp Cell Res 1999; 251:102-10. [PMID: 10438575 DOI: 10.1006/excr.1999.4542] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Most nontransformed cell lines respond to confluence by arresting the cell cycle in a viable G(1) phase, whereas immortalized cell lines growing in monolayer do not stop cell cycle progression in response to high cell density and are subjected to density-dependent apoptosis. We have examined the effects, in terms of cell growth, apoptosis, and expression of adhesion molecules of culturing contact inhibition-deficient hamster cells in the presence of dimethyl sulfoxide (DMSO). Addition of 1.5% DMSO to the growth medium for 96 h arrested Chinese hamster ovary (CHO) cells in the G(1) phase as a confluent monolayer, associated with a remarkable increase in the expression of the cyclin-dependent kinase inhibitor p27. Cells cultured in DMSO-containing medium showed increased levels of cadherins and alpha5beta1 and beta1 integrin complexes. Cell exposure to DMSO also reduced both cell density-dependent apoptosis and necrosis and resulted in increased Bcl-2 expression. These results converge to indicate that DMSO restores contact inhibition-induced growth arrest and prevents high-density-dependent apoptosis and suggest that the effect of DMSO may be mediated by intracellular signaling triggered by cell-extracellular matrix and cell-cell interactions. Both p27 and bcl-2 appear to be involved in the resumption of growth control accompanying cell adhesion in DMSO-exposed CHO cells.
Collapse
Affiliation(s)
- M Fiore
- Centre for Evolutionary Genetics, C.N.R., c/o Department of Genetics and Molecular Biology, University "La Sapienza,", Rome, Italy
| | | |
Collapse
|
20
|
St Croix B, Sheehan C, Rak JW, Flørenes VA, Slingerland JM, Kerbel RS. E-Cadherin-dependent growth suppression is mediated by the cyclin-dependent kinase inhibitor p27(KIP1). J Biophys Biochem Cytol 1998; 142:557-71. [PMID: 9679152 PMCID: PMC2133056 DOI: 10.1083/jcb.142.2.557] [Citation(s) in RCA: 346] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Recent studies have demonstrated the importance of E-cadherin, a homophilic cell-cell adhesion molecule, in contact inhibition of growth of normal epithelial cells. Many tumor cells also maintain strong intercellular adhesion, and are growth-inhibited by cell- cell contact, especially when grown in three-dimensional culture. To determine if E-cadherin could mediate contact-dependent growth inhibition of nonadherent EMT/6 mouse mammary carcinoma cells that lack E-cadherin, we transfected these cells with an exogenous E-cadherin expression vector. E-cadherin expression in EMT/6 cells resulted in tighter adhesion of multicellular spheroids and a reduced proliferative fraction in three-dimensional culture. In addition to increased cell-cell adhesion, E-cadherin expression also resulted in dephosphorylation of the retinoblastoma protein, an increase in the level of the cyclin-dependent kinase inhibitor p27(kip1) and a late reduction in cyclin D1 protein. Tightly adherent spheroids also showed increased levels of p27 bound to the cyclin E-cdk2 complex, and a reduction in cyclin E-cdk2 activity. Exposure to E-cadherin-neutralizing antibodies in three-dimensional culture simultaneously prevented adhesion and stimulated proliferation of E-cadherin transfectants as well as a panel of human colon, breast, and lung carcinoma cell lines that express functional E-cadherin. To test the importance of p27 in E-cadherin-dependent growth inhibition, we engineered E-cadherin-positive cells to express inducible p27. By forcing expression of p27 levels similar to those observed in aggregated cells, the stimulatory effect of E-cadherin-neutralizing antibodies on proliferation could be inhibited. This study demonstrates that E-cadherin, classically described as an invasion suppressor, is also a major growth suppressor, and its ability to inhibit proliferation involves upregulation of the cyclin-dependent kinase inhibitor p27.
Collapse
Affiliation(s)
- B St Croix
- Division of Cancer Biology Research, Sunnybrook Health Science Center, University of Toronto, Toronto-Sunnybrook Regional Cancer Center, Toronto, Ontario, Canada M4N 3M5
| | | | | | | | | | | |
Collapse
|
21
|
Krushel LA, Tai MH, Cunningham BA, Edelman GM, Crossin KL. Neural cell adhesion molecule (N-CAM) domains and intracellular signaling pathways involved in the inhibition of astrocyte proliferation. Proc Natl Acad Sci U S A 1998; 95:2592-6. [PMID: 9482931 PMCID: PMC19425 DOI: 10.1073/pnas.95.5.2592] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/1997] [Indexed: 02/06/2023] Open
Abstract
The neural cell adhesion molecule (N-CAM) inhibits astrocyte proliferation in vitro and in vivo, and this effect is partially reversed by the glucocorticoid antagonist RU-486. The present studies have tested the hypothesis that N-CAM-mediated inhibition of astrocyte proliferation is caused by homophilic binding and involves the activation of glucocorticoid receptors. It was observed that all N-CAM Ig domains inhibited astrocyte proliferation in parallel with their ability to influence N-CAM binding. The proliferation of other N-CAM-expressing cells also was inhibited by the addition of N-CAM. In contrast, the proliferation of astrocytes from knockout mice lacking N-CAM was not inhibited by added N-CAM. These findings support the hypothesis that it is binding of soluble N-CAM to N-CAM on the astrocyte surface that leads to decreased proliferation. Signaling pathways stimulated by growth factors include activation of mitogen-activated protein (MAP) kinase. Addition of N-CAM inhibited MAP kinase activity induced by basic fibroblast growth factor in astrocytes. In accord with previous findings that RU-486 could partially prevent the proliferative effects of N-CAM, inhibition of MAP kinase activity by N-CAM was reversed by RU-486. The ability of N-CAM to inhibit astrocyte proliferation was unaffected, however, by agents that block the ability of N-CAM to promote neurite outgrowth. Together, these findings indicate that homophilic N-CAM binding leads to inhibition of astrocyte proliferation via a pathway involving the glucocorticoid receptor and that the ability of N-CAM to influence astrocyte proliferation and neurite outgrowth involves different signal pathways.
Collapse
Affiliation(s)
- L A Krushel
- Department of Neurobiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
22
|
Edvardsen K, Bock E, Jirus S, Frandsen TL, Holst-Hansen C, Moser C, Spang-Thomsen M, Pedersen N, Walsh FS, Vindeløv LL, Brünner N. Effect of NCAM-transfection on growth and invasion of a human cancer cell line. APMIS 1997; 105:919-30. [PMID: 9463510 DOI: 10.1111/j.1699-0463.1997.tb05103.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A cDNA encoding the human transmembrane 140 kDa isoform of the neural cell adhesion molecule (NCAM) was transfected into the highly invasive MDA-MB-231 human breast cancer cell line. Transfectants with a homogeneous expression of NCAM showed a restricted capacity for penetration of an artificial basement membrane. However, when injected into nude mice, both control and NCAM-expressing cell lines produced equally invasive tumors. Tumors generated from NCAM-transfected cells were heterogeneous, containing NCAM-positive as well as NCAM-negative areas, indicating the existence of host factors capable of modulating NCAM expression in vivo. In nude mice, NCAM-transfected cells developed tumors with longer latency periods and slower growth rates than tumors induced by NCAM-negative control cells, implying that NCAM may be involved not only in adhesive and motile behavior of tumor cells but also in their growth regulation. There was no indication of differences in cell proliferative characteristics between the different NCAM-transfected and the control transfected cells as determined by flow cytometric DNA analysis, suggesting an increased cell loss as the reason for decreased in vivo growth rate of the NCAM-transfected cells. The fact that NCAM expression influences growth regulation attributes a pivotal role to this cell adhesion molecule during ontogenesis and tumor development.
Collapse
Affiliation(s)
- K Edvardsen
- Research Center for Medical Biotechnology, University of Copenhagen, Panum Institute, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Roesler J, Srivatsan E, Moatamed F, Peters J, Livingston EH. Tumor suppressor activity of neural cell adhesion molecule in colon carcinoma. Am J Surg 1997; 174:251-7. [PMID: 9324132 DOI: 10.1016/s0002-9610(97)00142-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Neural Cell Adhesion Molecule (NCAM) is a well-characterized member of the immunoglobin superfamily. The structure of NCAM is similar to the tumor suppressor Deleted in Colon Carcinoma (DCC). NCAM has been found in some epithelial tissues and plays a role in tumorigenesis of some cancers. The purpose of the present study was to determine if NCAM is present in normal human colon. Once its presence was established, its function as a tumor suppressor was investigated. METHODS Colon tumors and normal proximal margins were processed for reverse transcription-polymerase chain reaction (RT-PCR) of the NCAM-180 message. Immunohistochemistry of the tissue was performed to determine the distribution of NCAM. RESULTS RT-PCR analysis demonstrated the presence of the NCAM-180 kD isoform in normal colonic epithelia. Immunohistochemistry showed NCAM on the basolateral surface of colonic epithelial cells of the villous tips. Tumors from 15 patients followed up for 4 years were studied. All seven tumors expressing NCAM-180 were from patients having a benign clinical course. Seven of eight tumors that lacked NCAM-180 were associated with aggressive clinical behaviors (presenting with obstruction, perforation or metastatic disease, or patient death within 18 months of presentation). The sole exception was in a villous adenoma excised from a patient who has had multiply recurrent polyps on follow-up. CONCLUSION We conclude that like DCC, NCAM is an important colonic adhesion molecule that functions as a tumor suppressor.
Collapse
Affiliation(s)
- J Roesler
- Surgical, Pathology and Research Services, West Los Angeles VAMC, and the University of California, Los Angeles, School of Medicine, 90095-6904, USA
| | | | | | | | | |
Collapse
|
24
|
Kang JS, Gao M, Feinleib JL, Cotter PD, Guadagno SN, Krauss RS. CDO: an oncogene-, serum-, and anchorage-regulated member of the Ig/fibronectin type III repeat family. J Cell Biol 1997; 138:203-13. [PMID: 9214393 PMCID: PMC2139939 DOI: 10.1083/jcb.138.1.203] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Cell adhesion molecules of the Ig superfamily are implicated in a wide variety of biological processes, including cell migration, axon guidance and fasciculation, and growth control and tumorigenesis. Expression of these proteins can be highly dynamic and cell type specific, but little is known of the signals that regulate such specificity. Reported here is the molecular cloning and characterization of rat CDO, a novel cell surface glycoprotein of the Ig superfamily that contains five Ig-like repeats, followed by three fibronectin type III-like repeats in its extracellular region, and a 256-amino acid intracellular region that does not resemble other known proteins. In rat embryo fibroblasts, cdo mRNA expression is maximal in confluent, quiescent cells. It is rapidly and transiently down-regulated by serum stimulation of such cells, and is constitutively down-regulated in oncogene-transformed derivatives of these cells. CDO protein levels are also dramatically regulated by cell-substratum adhesion, via a mechanism that is independent of cdo mRNA expression. The amount of CDO produced at the surface of a cell may therefore be governed by a complex balance of signals, including mitogenic stimuli that regulate cdo mRNA levels, and substratum-derived signals that regulate CDO protein production. cdo mRNA is expressed at low levels in most adult rat tissues. A closely related human gene maps to chromosome 11q23-24, a region that displays frequent loss of heterozygosity in human lung, breast, and ovarian tumors. Taken together, these data suggest that loss of CDO function could play a role in oncogenesis.
Collapse
Affiliation(s)
- J S Kang
- Department of Biochemistry, Mount Sinai School of Medicine, New York 10029, USA
| | | | | | | | | | | |
Collapse
|
25
|
Zhang M, Wang MH, Singh RK, Wells A, Siegal GP. Epidermal growth factor induces CD44 gene expression through a novel regulatory element in mouse fibroblasts. J Biol Chem 1997; 272:14139-46. [PMID: 9162042 DOI: 10.1074/jbc.272.22.14139] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Growth factors coordinately regulate a variety of genes associated with pathological states including tumor invasion and metastasis. Overexpressed epidermal growth factor receptor (EGFR) on tumor cell surfaces is associated with enhanced cell attachment and migration into extracellular matrices, which promotes tumor aggressiveness. We have demonstrated that epidermal growth factor (EGF) up-regulates the cell surface adhesion molecule CD44 at both the mRNA and protein levels on mouse fibroblasts expressing full-length wild-type EGFR (NR6-WT) but not on EGFR-deficient cells (NR6-P). This increases cell attachment to hyaluronic acid. In this investigation, transcriptional regulation of CD44 by EGF was confirmed by defining an EGF-regulatory element. By employing human CD44 gene promoter-chloramphenicol acetyltransferase (CAT) constructs transfected into NR6-WT cells, EGF inducibility was observed within a 120-base pair (bp) DNA fragment located 450 bp upstream of the RNA initiation site. Differential EGF inducibility was found among different cell lines chosen, indicating a 3.2- and 1.8-fold enhancement in DU145 cells carrying exogenous wild-type EGFR and in MCF-7 cells, respectively, while minimal EGF induction was found in cervical cancer HeLa cells. Utilizing gel shift assays, a time-dependent increase of DNA-protein complex formation was found upon EGF stimulation in NR6-WT cells but not in NR6-P cells. Based upon these observations, a novel 22-bp EGF regulatory element (ERE) (5'--604CCCTCTCTCCAGCTCCTCTCCC-583-3') was isolated from the CD44 gene promoter. This ERE conferred DNA-protein binding ability in vitro, as well as the full functional recovery of EGF inducibility of CAT activity when linked to a homologous CD44 promoter or a SV40 promoter driving a CAT reporter gene. A two-base mutation of the ERE completely eliminated its binding activity as well as its EGF inducibility of CAT expression. Our studies indicate that EGF induces CD44 gene expression through an interaction between a specific ERE and putative novel transcriptional factor so as to regulate cell attachment to extracellular matrix.
Collapse
Affiliation(s)
- M Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35233-1924, USA
| | | | | | | | | |
Collapse
|
26
|
Crossin KL, Tai MH, Krushel LA, Mauro VP, Edelman GM. Glucocorticoid receptor pathways are involved in the inhibition of astrocyte proliferation. Proc Natl Acad Sci U S A 1997; 94:2687-92. [PMID: 9122257 PMCID: PMC20150 DOI: 10.1073/pnas.94.6.2687] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In earlier studies, the neural cell adhesion molecule, N-CAM, was found to inhibit the proliferation of rat astrocytes both in vitro and in vivo. To identify the gene targets involved, we used subtractive hybridization to examine changes in gene expression that occur after astrocytes are exposed to N-CAM in vitro. While the mRNA levels for N-CAM decreased after such treatment, the levels of mRNAs for glutamine synthetase and calreticulin increased. Since glutamine synthetase and calreticulin are known to be involved in glucocorticoid receptor pathways, we tested a number of steroids for their effects on astrocyte proliferation. Dexamethasone, corticosterone, and aldosterone were all found to inhibit rat cortical astrocyte proliferation in culture in a dose-dependent manner. RU-486, a potent glucocorticoid antagonist, reversed the inhibitory effects of dexamethasone. These observations prompted the hypothesis that the inhibition of proliferation by N-CAM might be mediated through the glucocorticoid receptor pathway. Consistent with this hypothesis, the inhibition of astrocyte proliferation by N-CAM was reversed in part by a number of glucocorticoid antagonists, including RU-486, dehydroepiandrosterone, and progesterone. In transfection experiments with cultured astrocytes, N-CAM treatment increased the expression of a luciferase reporter gene under the control of a minimal promoter linked to a glucocorticoid response element. The enhanced activity of this construct stimulated by N-CAM was abolished in the presence of RU-486. The combined data suggest that astrocyte proliferation is in part regulated by alterations in glucocorticoid receptor pathways.
Collapse
Affiliation(s)
- K L Crossin
- Department of Neurobiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
Cultured skin fibroblasts, among other non-neuronal cells (e.g. platelets, lymphocytes, red blood cells), provide an advantageous system for investigating dynamic molecular regulatory processes underlying abnormal cell growth, metabolism, and receptor-mediated signal transduction, without the confounding effects of disease state and its treatment in a variety of brain disorders, including schizophrenia, and are useful for studies of systemic biochemical defects with predominant consequences for brain function. These cells are also useful for studying aspects of neurotransmitter functions because the cells express enzymes involved in their metabolism, as well as their receptors with complete machinery for signal transduction. These processes also function predictably with receptors that are transfected in fibroblasts. This review will focus on the use of cultured skin of which have also been studied in post-mortem brains. These mechanisms might involve DNA processing and mitogenesis, cell-cell adhesion molecules, actions of growth factors, oxidative damage, and membrane phospholipid derived second messengers. This review will further discuss the implications of these processes to clinical and structural brain abnormalities. An understanding of these biochemical processes might help establish therapeutic implications and identify the risk for illness through experimental strategies such as epidemiology, family pedigree and high risk populations. Finally, despite some methodological limitations, skin fibroblasts are relatively easy to grow and maintain as primary cultures or as immortalized cell lines for long periods of time for use in investigating newly identified biochemical abnormalities.
Collapse
Affiliation(s)
- S P Mahadik
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta, USA
| | | |
Collapse
|
28
|
Zhang M, Singh RK, Wang MH, Wells A, Siegal GP. Epidermal growth factor modulates cell attachment to hyaluronic acid by the cell surface glycoprotein CD44. Clin Exp Metastasis 1996; 14:268-76. [PMID: 8674281 DOI: 10.1007/bf00053900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cell adhesion to and migration through extracellular matrices (ECM) are critical events in tumor invasion and metastasis. Previous work by us had demonstrated that signaling of epidermal growth factor receptor (EGFR) confers an oncogenic phenotype on NR6 cells and that these cells when transfected with holo EGFR demonstrate greater motility and invasiveness than cells carrying a carboxy-terminal truncated EGFR. Recently, a cell surface glycoprotein, CD44, has been implicated in cell-ECM adhesion involved in tumor cell migration, signal transduction, and metastasis. We investigated whether EGF regulates cellular interactions with ECM components, and in particular, hyaluronate, by modulating CD44 expression. In vitro cell attachment assays on hyaluronate-coated plates demonstrated similar basal level of binding (approximately 33%) for murine NR6 parental cells devoid of endogenous EGFR (P) or expressing wild-type EGFR (WT), while a time-dependent increase in binding was observed in WT cells stimulated with EGF. Additionally, utilizing monoclonal antibody blocking assays, CD44, but not EGFR, was shown to be directly involved in this attachment. Both WT and P cells possessed equivalent 95 kDa bands on immunoblots, corresponding to CD44. The existence of CD44 mRNA was verified by RT-PCR using synthetic oligonucleotides in which a 1.1 kb cDNA was detected in both cell lines and confirmed by DNA sequencing. After 24-h exposure to exogenous EGF, an increase in CD44 protein and mRNA expression was found in WT cells, but not in P cells, supporting the contention that a functional EGFR signaling pathway is required for CD44 regulation. Thus, EGF stimulates cell binding to hyaluronate in vitro by regulating CD44 expression.
Collapse
Affiliation(s)
- M Zhang
- Department of Pathology, University of Alabama at Birmingham, USA
| | | | | | | | | |
Collapse
|
29
|
Moreton K, Turner R, Blake N, Paton A, Groome N, Rumsby M. Protein expression of the alpha, gamma, delta and epsilon subspecies of protein kinase C changes as C6 glioma cells become contact inhibited and quiescent in the presence of serum. FEBS Lett 1995; 372:33-8. [PMID: 7556638 DOI: 10.1016/0014-5793(95)00941-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Total protein kinase C (PKC) activity and protein expression of the alpha and delta subspecies of PKC increases markedly as C6 glioma cells grow from low cell density to the contact-inhibited quiescent state (also known as G(o)) in the presence of serum. At the same time protein expression of PKC subspecies gamma and epsilon decreases while the beta I, beta II, iota and zeta subspecies did not change. Serum deprivation of growing C6 glioma cells does not induce the same changes in PKC subspecies protein expression. The findings support the growing view that there are significant differences between the G(o) states brought about by contact inhibition or serum deprivation.
Collapse
Affiliation(s)
- K Moreton
- Department of Biology, University of York, England, UK
| | | | | | | | | | | |
Collapse
|
30
|
Sporns O, Edelman GM, Crossin KL. The neural cell adhesion molecule (N-CAM) inhibits proliferation in primary cultures of rat astrocytes. Proc Natl Acad Sci U S A 1995; 92:542-6. [PMID: 7831326 PMCID: PMC42777 DOI: 10.1073/pnas.92.2.542] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cell proliferation is a key primary process during neural development and also plays an important role in the regenerative response of neural tissue to injury. It has been reported that glial cell proliferation is, at least in part, controlled by a neuronal signal, possibly involving cell surface molecules. We report here that the addition of purified rat neural cell adhesion molecule (N-CAM) to primary cultures of rat forebrain astrocytes inhibits their proliferation. This inhibitory effect can be elicited in cultures grown in chemically defined serum-free medium or in medium that had been supplemented with growth factors. Polyclonal antibodies to N-CAM or their Fab' fragments elicited a similar inhibitory effect. The magnitude of the inhibitory effect of N-CAM was dependent on cell density: it was maximal at low cell densities and weakened progressively as cells approached confluency. Synthetic peptides with sequences identical to a putative homophilic binding region of N-CAM mimicked the effect of purified N-CAM, while peptides of the same length and amino acid composition but with a randomized sequence did not. The addition of N-CAM antisense oligonucleotides to primary astrocyte cultures for 48 h resulted in reduced levels of N-CAM expression. After N-CAM levels on astrocytes were diminished by this treatment, the antiproliferative effect of N-CAM added to the medium was significantly reduced. The combined results suggest that N-CAM homophilic binding may be involved in the control of glial cell proliferation.
Collapse
Affiliation(s)
- O Sporns
- Department of Neurobiology, Scripps Research Institute, La Jolla, CA 92037
| | | | | |
Collapse
|
31
|
Ostman A, Yang Q, Tonks NK. Expression of DEP-1, a receptor-like protein-tyrosine-phosphatase, is enhanced with increasing cell density. Proc Natl Acad Sci U S A 1994; 91:9680-4. [PMID: 7937872 PMCID: PMC44880 DOI: 10.1073/pnas.91.21.9680] [Citation(s) in RCA: 169] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
cDNA encoding a receptor-like protein-tyrosine-phosphatase (PTP) termed DEP-1 was isolated from a HeLa cell library. The cDNA predicts an enzyme consisting of an extracellular segment containing eight fibronectin type III repeats, a single transmembrane segment, and a single intracellular PTP domain. Following expression of DEP-1 cDNA in COS cells a glycoprotein of 180 kDa was detected and PTP activity was demonstrated in immunocomplexes with a C-terminal peptide antiserum. Endogenous DEP-1 was detected in WI-38 human embryonic lung fibroblasts by immunoblotting and immunocomplex PTP assays. Immunoblot analysis of DEP-1 expression in WI-38 cells revealed dramatically increased levels and activity of the PTP in dense cultures relative to sparse cultures. Also, DEP-1 activity, detected in PTP assays of immunocomplexes, was increased in dense cell cultures. In contrast, the expression levels of PTP-1B did not change with cell density. This enhancement of DEP-1 expression with increasing cell density was also observed in another fibroblast cell line, AG1518. The increase in DEP-1 occurs gradually with increasing cell contact and is initiated before saturation cell density is reached. These observations suggest that DEP-1 may contribute to the mechanism of contact inhibition of cell growth.
Collapse
Affiliation(s)
- A Ostman
- Cold Spring Harbor Laboratory, NY 11724-2208
| | | | | |
Collapse
|
32
|
Shih IM, Elder DE, Hsu MY, Herlyn M. Regulation of Mel-CAM/MUC18 expression on melanocytes of different stages of tumor progression by normal keratinocytes. THE AMERICAN JOURNAL OF PATHOLOGY 1994; 145:837-45. [PMID: 7943174 PMCID: PMC1887332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The cell-cell adhesion receptor, Mel-CAM/MUC18, is highly expressed on metastatic melanoma cells and is also detectable on primary melanomas but not on normal melanocytes. Previous studies have shown that increased Mel-CAM/MUC18 expression correlates with tumor thickness and metastatic potential. We show here that normal melanocytes and nevus cells in culture express Mel-CAM/MUC18, but expression is down-regulated when cells are co-cultured with keratinocytes. Such keratinocyte-mediated regulation of Mel-CAM/MUC18 expression on melanocytes, nevus cells, and early melanomas can also be demonstrated in situ in patients' specimens. On the other hand, melanoma cells from primary and metastatic lesions constitutively express Mel-CAM/MUC18, and keratinocytes have no modulatory effect. These results suggest that contact between keratinocytes and human melanocytic cells modulates Mel-CAM/MUC18 expression, raising the possibility that escape from keratinocyte control during melanoma development leads to expression of antigens that contribute to the malignant phenotype.
Collapse
Affiliation(s)
- I M Shih
- Wistar Institute, Philadelphia, PA 19104-4268
| | | | | | | |
Collapse
|
33
|
Abstract
Cell adhesion is a key process, elementary in the establishment of tissue architecture and differentiation. In neoplasia, in which there is a disruption of tissue architecture and a derangement in differentiation, it has been postulated that changes in cell-cell and cell-matrix interactions account for the ability of cancer cells to transgress normal tissue boundaries and disperse to distant sites. Complex and coordinated reductions and increases in adhesion have been proposed to be necessary for tumor invasion and metastasis. This hypothesis has fueled the interest of cancer research teams to evaluate the expression of various adhesion molecules in a wide range of human malignancies in the hope of pinpointing some of the cell adhesion alterations underlying tumor behavior. To date, a multitude of transmembrane glycoproteins, including cell-cell adhesion molecules (CAMs) and cell-matrix or substratum adhesion molecules (SAMs), have been identified; their structure, molecular genetics, and biochemistry have been elucidated, and we are beginning to understand their normal function. A few of these, on the basis of current evidence, seem to be promising candidate molecules for a role in neoplasia. This article aims to summarize recent developments in this field of adhesion research as well as the clinical applications in diagnostic pathology arising from it. First, by way of introduction, a summary of the biochemical and functional characterization of each family of adhesion receptors will be presented, followed by a presentation of the experimental data implicating them in the control of invasion, metastasis, and differentiation.
Collapse
Affiliation(s)
- M Pignatelli
- Department of Histopathology, Royal Postgraduate Medical School, London, United Kingdom
| | | |
Collapse
|
34
|
Chuong CM, Jiang TX, Yin E, Widelitz RB. cDCC (chicken homologue to a gene deleted in colorectal carcinoma) is an epithelial adhesion molecule expressed in the basal cells and involved in epithelial-mesenchymal interaction. Dev Biol 1994; 164:383-97. [PMID: 8045341 DOI: 10.1006/dbio.1994.1208] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cloning of human DCC (deleted in colorectal carcinoma, Fearon et al., 1990) showed that it is an immunoglobulin superfamily member homologous to neural cell adhesion molecules (N-CAM). To explore the normal function of this molecule, we have cloned a chicken homologue to DCC (cDCC) and raised an antibody to DCC. cDCC is a protein of 160 kDa with an expression pattern distinct from those of other immunoglobulin family members including N-CAM and Ng-CAM. Transgene expression of cDCC in fibroblasts led to increased cell-cell adhesion. Localization studies in chicken and mouse embryos showed that DCC is expressed in the epithelia of skin, gut, lung, and bladder. In adult, the expression of DCC is limited to the basal layer of stratified epithelium in skin, crypt regions of intestinal villi, and stem cells in mammary duct. Cell aggregation assay using embryonic chicken skin epithelial cells and antibody to DCC showed it is a Ca2+ independent cell adhesion molecule. In epithelial-mesenchymal interactions during feather morphogenesis, antibody to DCC suppressed the formation of dermal condensations and the polarized localization of N-CAM and fibronectin. These results implied that DCC is an epithelial cell adhesion molecule required for mediating critical functions in epithelial-epithelial and epithelial-mesenchymal interactions.
Collapse
Affiliation(s)
- C M Chuong
- Department of Pathology, School of Medicine, University of Southern California, Los Angeles 90033
| | | | | | | |
Collapse
|
35
|
Edvardsen K, Pedersen PH, Bjerkvig R, Hermann GG, Zeuthen J, Laerum OD, Walsh FS, Bock E. Transfection of glioma cells with the neural-cell adhesion molecule NCAM: effect on glioma-cell invasion and growth in vivo. Int J Cancer 1994; 58:116-22. [PMID: 8014007 DOI: 10.1002/ijc.2910580119] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The tumor growth and the invasive capacity of a rat glioma cell line (BT4Cn) were studied after transfection with the human transmembrane 140-kDa isoform of the neural-cell adhesion molecule, NCAM. After s.c. injection, the NCAM-transfected cells showed a slower growth rate than the parent cell line (BT4Cn). Upon intracerebral implantation with BT4Cn cells and different clones of NCAM-transfected cells, all animals developed neurological symptoms within 13-16 days. However, the tumors showed different growth characteristics. The NCAM-transfected BT4Cn cells were localized in the region of the injection site, with a sharply demarcated border between the tumor and brain tissue. In contrast, the parental cell line showed single-cell infiltration and more pronounced destruction of normal brain tissue. Using a 51Cr-release assay, spleen cells from rats transplanted with BT4Cn tumor cells generally showed a lower cytotoxic response than the spleen cells from rats transplanted with the transfected variants of BT4Cn cells, indicating that the transfection procedure in itself mediated an activation of the immune system. The present data suggest that NCAM may influence the malignant behavior of rat glioma cells in vivo.
Collapse
Affiliation(s)
- K Edvardsen
- Research Center for Medical Biotechnology, University of Copenhagen, Panum Institute, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hueber AO, Raposo G, Pierres M, He HT. Thy-1 triggers mouse thymocyte apoptosis through a bcl-2-resistant mechanism. J Exp Med 1994; 179:785-96. [PMID: 7906706 PMCID: PMC2191406 DOI: 10.1084/jem.179.3.785] [Citation(s) in RCA: 57] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Programmed cell death plays an important role during thymocyte development, since a vast majority (97%) of mouse cortical thymocytes die in thymus, whereas only 3% of these cells are rescued from cell death and positively selected. Although it seems well established that thymocyte fate depends upon appropriate surface-expressed T cell receptor, little is known about the molecular mechanism(s) responsible for the massive thymocyte elimination that occurs in the thymus. We report here that Thy-1 is capable of triggering mouse thymocyte death in vitro through a bcl-2-resistant mechanism. We have previously shown that Thy-1 is involved in mouse thymocyte adhesion to thymic stroma through interaction with an epithelial cell ligand. To examine the Thy-1 signaling function in thymocytes, we have mimicked its interaction with stromal cells by culturing mouse thymocytes onto tissue culture plates coated with monoclonal antibodies (mAb) directed at distinct Thy-1 epitope regions. mAb recognizing determinants in a defined Thy-1 structural domain, but not others, were found to induce marked thymocyte apoptosis as evidenced by morphological and biochemical data. Use of a quantitative DNA dot blot assay indicated that Thy-1-mediated thymocyte apoptosis was not blocked by RNA or protein synthesis inhibitors, EGTA, or by cyclosporin A, and differed, therefore, from "activation-driven cell death". Moreover, Thy-1(+)-transfected, but not wild-type AKR1 (Thy-1-d) thymoma cells underwent apoptosis after ligation with apoptosis-inducing, Thy-1-specific mAb. In contrast to thymocytes, the latter event was inhibitable by RNA and protein synthesis inhibitors, an indication that thymocytes, but not thymoma cells, contain the molecular components necessary for Thy-1-driven apoptosis. We further showed that Thy-1-triggered thymocyte death is a developmentally regulated process operative in fetal thymocytes from day 17 of gestation, but not in peripheral T cells. Indeed, the target of apoptosis by anti-Thy-1 was found to reside mainly within the CD4+8+3- and CD4+8+3lo double positive immature thymocyte subsets. Finally, it is of major interest that Thy-1-mediated apoptosis, which was found to be readily detectable in thymocytes from bcl-2-transgenic mice, represents a thus far unique experimental system for studying bcl-2-resistant thymocyte death mechanism(s).
Collapse
Affiliation(s)
- A O Hueber
- Centre d'Immunologie Institut National de la Santé et de la Recherche Médicale-Centre National de la Recherche Scientifique de Marseille Luminy, France
| | | | | | | |
Collapse
|
37
|
Edvardsen K, Brünner N, Spang-Thomsen M, Walsh FS, Bock E. Migratory, invasive and metastatic capacity of NCAM transfected rat glioma cells. Int J Dev Neurosci 1993; 11:681-90. [PMID: 8116479 DOI: 10.1016/0736-5748(93)90055-i] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
A cDNA encoding a transmembrane 140 kDa isoform of the neural cell adhesion molecule, NCAM, was transfected into the rat glioma cell line BT4Cn. Transfectants with a homogeneously high expression of NCAM-B showed a decreased capacity for penetration of an artificial basement membrane when compared to cells transfected with expression-vector alone or untransfected cells. However, when injected subcutaneously into nude mice, both NCAM expressing cells and control cells produced invasive tumors. Nude mice injected with NCAM positive cells developed tumors with slower growth rates as compared to those induced by NCAM negative cells. This implies that NCAM may not only be involved in adhesive and motile behaviour of glioma cells, but also in their growth regulation.
Collapse
Affiliation(s)
- K Edvardsen
- Research Centre for Medical Biotechnology, University of Copenhagen, Panum Institute, Denmark
| | | | | | | | | |
Collapse
|
38
|
Seki T, Arai Y. Distribution and possible roles of the highly polysialylated neural cell adhesion molecule (NCAM-H) in the developing and adult central nervous system. Neurosci Res 1993; 17:265-90. [PMID: 8264989 DOI: 10.1016/0168-0102(93)90111-3] [Citation(s) in RCA: 318] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The neural cell adhesion molecule (NCAM) is a cell surface glycoprotein which is thought to mediate cell adhesion and recognition. During developmental stages, NCAM is highly polysialylated (NCAM-H) by a unique alpha-2,8-linked polysialic acid chain (PSA), and this PSA portion of NCAM-H has been found to be closely associated with various developmental processes of the nervous system. Further, recent immunohistochemical investigations have revealed that even in the adult nervous system, a persistent PSA expression has been found confined to several regions: the olfactory bulb, the piriform cortex, the hippocampal dentate gyrus, the hypothalamus, some nuclei of the medulla and the dorsal horn of the spinal cord, which are related directly or indirectly to sensory systems. Moreover, in the dentate gyrus and olfactory bulb the expression is connected with adult neurogenesis that may add new neuronal circuits to the adult neural tissue. Therefore, the possible role of NCAM-H in the central nervous system may be associated not only with neural development, but also with adult functions, such as the processing system of sensory information and neuronal plasticity.
Collapse
Affiliation(s)
- T Seki
- Department of Anatomy, Juntendo University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
39
|
Lafrenie RM, Buchanan MR, Orr FW. Adhesion molecules and their role in cancer metastasis. CELL BIOPHYSICS 1993; 23:3-89. [PMID: 7895250 DOI: 10.1007/bf02796507] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
This article describes various adhesion molecules and reviews evidence to support a mechanistic role for adhesion molecules in the process of cancer metastasis. A variety of evidence supports the involvement of specific adhesion molecules in metastasis. 1. For example, some cancer cells metastasize to specific organs, irrespective of the first organ encountered by the circulating cancer cells. This ability to colonize a specific organ has been correlated with the preferential adhesion of the cancer cells to endothelial cells derived from the target organ. This suggests that cancer cell/endothelial cell adhesion is involved in cancer cell metastasis and that adhesion molecules are expressed on the endothelium in an organ-specific manner. 2. Further, inclusion of peptides that inhibit cell adhesion, such as the YIGSR- or RGD-containing peptides, is capable of inhibiting experimental metastasis. 3. Metastasis can be enhanced by acute or chronic inflammation of target vessels, or by treatment of animals with inflammatory cytokines, such as interleukin-1. In vitro, cancer cell/endothelial cell adhesion can be enhanced by pretreating the endothelial cell monolayer with cytokines, such as interleukin-1 or tumor necrosis factor-alpha. This suggests that, in addition to organ-specific adhesion molecules, a population of inducible endothelial adhesion molecules is involved and is relevant to metastasis. 4. Further support for this model is found in the comparison to leukocyte/endothelial adhesion during leukocyte trafficking. Convincing evidence exists, both in vivo and in vitro, to demonstrate an absolute requirement for leukocyte/endothelial adhesion before leukocyte extravasation can occur. The relevance of this comparison to metastasis is reinforced by the observation that some of the adhesion molecules involved in leukocyte/endothelial adhesion are also implicated in cancer cell/endothelial adhesion. The involvement of adhesion molecules suggests a potential therapy for metastasis based on interrupting adhesive interactions that would augment other treatments for primary tumors.
Collapse
Affiliation(s)
- R M Lafrenie
- Department of Pathology, McMaster University, Hamilton, Ontario
| | | | | |
Collapse
|
40
|
Brady-Kalnay SM, Boghaert ER, Zimmer S, Brackenbury R. Increasing N-CAM-mediated cell-cell adhesion does not reduce invasion of RSV-transformed WC5 rat cerebellar cells. Clin Exp Metastasis 1993; 11:313-24. [PMID: 8391406 DOI: 10.1007/bf00058051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The WC5 rat cerebellar cell line, infected with a Rous sarcoma virus (RSV) that is temperature-sensitive for pp60v-src transformation, expresses high levels of the neural cell adhesion molecule, N-CAM, when grown at the non-permissive temperature for pp60v-src activity. At the permissive temperature, N-CAM expression is 4- to 10-fold reduced and the cells aggregate poorly. To evaluate the effects of variations in N-CAM expression, we compared the invasive ability of transformed WC5 cells that express low levels of N-CAM with transformed cells in which N-CAM-mediated adhesion was restored. WC5 cells were transfected with expression vectors containing cDNAs encoding the 120 or 180 kDa forms of chicken N-CAM linked to constitutive promoters. Several permanently transfected lines that expressed chicken N-CAM at the cell surface were isolated. These cell lines showed enhanced aggregation at the permissive temperature relative to untransfected WC5 cells or cells transfected with control constructs. By comparing the ability of control and transfected WC5 cells to invade reconstituted extracellular matrix, we tested the effect of variations in N-CAM-mediated adhesion on invasion. Clones that expressed high levels of N-CAM showed invasion rates that were similar to control cells, indicating that increasing N-CAM-mediated adhesion does not inhibit the invasiveness of RSV-transformed WC5 cells.
Collapse
Affiliation(s)
- S M Brady-Kalnay
- Department of Anatomy and Cell Biology, University of Cincinnati Medical Center, OH 45267-0521
| | | | | | | |
Collapse
|
41
|
Bally-Cuif L, Goridis C, Santoni MJ. The mouse NCAM gene displays a biphasic expression pattern during neural tube development. Development 1993; 117:543-52. [PMID: 8330525 DOI: 10.1242/dev.117.2.543] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The neural cell adhesion molecule (NCAM) is one of the most abundant cell adhesion molecules expressed in vertebrates and it is thought to play important roles as a regulator of morphogenetic processes, but little is known of its expression pattern in mammalian embryos. In this study, we have examined the developmental profile of NCAM gene expression in mouse embryos from gestational day 7.5 to 12.5, focusing on the developing neural tube. NCAM transcripts were first detected around day 8.5 in the somites and the forming neural tube. At this stage, NCAM transcripts were expressed in the neuroepithelium throughout the width of the neural groove and tube up to a rostral boundary within the hindbrain, whereas NCAM mRNA levels were very low or undetectable in the neuroepithelium of the head region. The positional restriction of NCAM expression was confirmed by immunohistochemistry at the protein, and by polymerase chain reaction analysis at the RNA level. Expression in the neuroepithelium was transient as the level of NCAM transcripts declined in the germinal layer beyond day 8.5. By day 9.5, strong NCAM expression had appeared on the earliest postmitotic neurones along the entire neuraxis, and this pattern of expression in all regions with differentiating neurones was maintained until day 12.5. We conclude that NCAM expression in the neural tube occurs in two spatiotemporal distinct waves: a first wave in the proliferating neuroepithelium showing positional dependence along the rostrocaudal axis, and a second wave on essentially all neurones that have become postmitotic.
Collapse
Affiliation(s)
- L Bally-Cuif
- INSERM U106, Hôpital de la Salpêtrière, Paris, France
| | | | | |
Collapse
|
42
|
Lee YS, Chuong CM. Adhesion molecules in skeletogenesis: I. Transient expression of neural cell adhesion molecules (NCAM) in osteoblasts during endochondral and intramembranous ossification. J Bone Miner Res 1992; 7:1435-46. [PMID: 1481729 DOI: 10.1002/jbmr.5650071211] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We report that neural cell adhesion molecules (NCAM) are expressed transiently in developing chicken osteoblasts during osteogenesis using immunostaining on cryostat sections. NCAM is strongly expressed in most osteoblasts along bone trabeculae that coincide with the presence of collagen I and alkaline phosphatase activity. In endochondral ossification, NCAM is highly expressed in osteogenic buds as seen in the epiphysis and diaphysis of tibia and vertebrae. In intramembranous ossification, NCAM is seen in osteogenic condensation of calvaria and in the periosteum of tibial diaphysis. The expression is transient because NCAM is not expressed in mesenchymal cells before osteogenic condensation and NCAM expression is lost in osteocytes in later stages. The staining pattern suggests that NCAM is present on the cell membrane of osteoblasts. Using a specific monoclonal antibody, the osteoblast NCAM is shown to contain polysialic acid, which is enriched in embryonic brain. Northern blot analysis using chicken brain NCAM cDNA as probes showed two major sizes of mRNA at 6.4 and 4.2 kb in calvarial mRNA as opposed to bands at 7.2, 6.4, and 4.2 kb in the brain. An immunoblot showed major proteins at Mr 165 and 110 kd, unlike brain NCAM, which are 180, 140, and 120 kD. That NCAM is involved in bone morphogenesis is consistent with the general hypothesis that NCAM plays pivotal roles in mesenchymal condensation, as shown in the formation of muscle, kidney, skin, and cartilage. The results establish NCAM as a cell surface molecule expressed transiently during osteoblast lineage. The implication that NCAM may mediate osteoblast interaction and regulate skeletal morphogenesis is discussed.
Collapse
Affiliation(s)
- Y S Lee
- Department of Pathology, School of Medicine, University of Southern California, Los Angeles
| | | |
Collapse
|
43
|
Abstract
Several aspects of the interactions between growth factors and cell adhesion are described. Recent advances in the field come from the identification of molecules resembling growth factors or growth factor receptors, which bear cell adhesion motifs as well as molecules participating in both cell growth control and adhesion.
Collapse
Affiliation(s)
- J P Thiery
- Laboratoire de Physiopathologie du Développement URA CNRS, Ecole Normale Supérieure, Paris, France
| | | |
Collapse
|
44
|
Goridis C, Brunet JF. NCAM: structural diversity, function and regulation of expression. SEMINARS IN CELL BIOLOGY 1992; 3:189-97. [PMID: 1623208 DOI: 10.1016/s1043-4682(10)80015-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
NCAM is a large family of structurally closely related proteins with cell-cell adhesive properties and a temporo-spatially regulated expression throughout development. This review covers recent work on NCAM with an emphasis on the still open questions of the full extent of structural diversity and the mechanism whereby it arises, the chemistry and functional consequences of the binding event and the intricacies of the developmental regulation of NCAM, all of which have ramifications in its likely role as an effector of morphogenesis.
Collapse
Affiliation(s)
- C Goridis
- Centre d'Immunologie INSERM-CNRS de Marseille-Luminy, France
| | | |
Collapse
|