1651
|
Henning W, Stürzbecher HW. Homologous recombination and cell cycle checkpoints: Rad51 in tumour progression and therapy resistance. Toxicology 2003; 193:91-109. [PMID: 14599770 DOI: 10.1016/s0300-483x(03)00291-9] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We provide an overview of the functional interrelationship between genes and proteins related to DNA repair by homologous recombination and cell cycle regulation in relation to the progression and therapy resistance of human tumours. To ensure the high-fidelity transmission of genetic information from one generation to the next, cells have evolved mechanisms to monitor genome integrity. Upon DNA damage, cells initiate complex response pathways including cell cycle arrest, activation of genes and gene products involved in DNA repair, and under some circumstances, the triggering of programmed cell death. Deregulation of this co-ordinated response leads to genetic instability and is fundamental to the aetiology of human cancer. Homologous recombination involved in DNA repair is induced by environmental damage as well as misreplication during the normal cell cycle. However, when not regulated properly, it can result in the loss of heterozygocity or genetic rearrangements, central to the process of carcinogenesis. The central step of homologous recombination is the DNA strand exchange reaction catalysed by the eukaryotic Rad51 protein. Here, we describe the recent progress in our understanding of how Rad51 is involved in the signalling and repair of DNA damage and how tumour suppressors, such as p53, ATM, BRCA1, BRCA2, BLM and FANCD2 are linked to Rad51-dependent pathways. An increased knowledge of the role of Rad51 in DNA repair by homologous recombination and its effects on cell cycle progression, tumour development and tumour resistance may provide opportunities for identifying improved diagnostic markers and developing more effective treatments for cancer.
Collapse
Affiliation(s)
- Wilhelm Henning
- Institute of Pathology, University Clinic Schleswig-Holstein, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| | | |
Collapse
|
1652
|
Muraoka RS, Koh Y, Roebuck LR, Sanders ME, Brantley-Sieders D, Gorska AE, Moses HL, Arteaga CL. Increased malignancy of Neu-induced mammary tumors overexpressing active transforming growth factor beta1. Mol Cell Biol 2003; 23:8691-703. [PMID: 14612410 PMCID: PMC262670 DOI: 10.1128/mcb.23.23.8691-8703.2003] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To determine if Neu is dominant over transforming growth factor beta (TGF-beta), we crossed mouse mammary tumor virus (MMTV)-Neu mice with MMTV-TGF-beta1(S223/225) mice expressing active TGF-beta1 in the mammary gland. Bigenic (NT) and Neu-induced mammary tumors developed with a similar latency. The bigenic tumors and their metastases were less proliferative than those occurring in MMTV-Neu mice. However, NT tumors exhibited less apoptosis and were more locally invasive and of higher histological grade. NT mice exhibited more circulating tumor cells and lung metastases than Neu mice, while NT tumors contained higher levels of phosphorylated (active) Smad2, Akt, mitogen-activated protein kinase (MAPK), and p38, as well as vimentin content and Rac1 activity in situ than tumors expressing Neu alone. Ex vivo, NT cells exhibited higher levels of P-Akt and P-MAPK than Neu cells. These were inhibited by the TGF-beta inhibitor-soluble TGF-beta type II receptor (TbetaRII:Fc), suggesting they were activated by autocrine TGF-beta. TGF-beta stimulated migration of Neu cells into surrounding matrix, while the soluble TGF-beta inhibitor abrogated motility and invasiveness of NT cells. These data suggest that (i) the antimitogenic and prometastatic effects of TGF-beta can exist simultaneously and (ii) Neu does not abrogate TGF-beta-mediated antiproliferative action but can synergize with TGF-beta in accelerating metastatic tumor progression.
Collapse
Affiliation(s)
- Rebecca S Muraoka
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
1653
|
Li W, Qiao W, Chen L, Xu X, Yang X, Li D, Li C, Brodie SG, Meguid MM, Hennighausen L, Deng CX. Squamous cell carcinoma and mammary abscess formation through squamous metaplasia inSmad4/Dpc4conditional knockout mice. Development 2003; 130:6143-53. [PMID: 14597578 DOI: 10.1242/dev.00820] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Smad4 is a central mediator for TGFβ signals, which play important functions in many biological processes. To study the role of Smad4 in mammary gland development and neoplasia, we disrupted this gene in mammary epithelium using a Cre-loxP approach. Smad4 is expressed in the mammary gland throughout development; however, its inactivation did not cause abnormal development of the gland during the first three pregnancies. Instead, lack of Smad4 gradually induced cell proliferation, alveolar hyperplasia and transdifferentiation of mammary epithelial cells into squamous epithelial cells. Consequently, all mutant mice developed squamous cell carcinoma and/or mammary abscesses between 5 and 16 months of age. We demonstrated that absence of Smad4 resulted in β-catenin accumulation at onset and throughout the process of transdifferentiation, implicating β-catenin, a key component of the Wnt signaling pathway, in the development of squamous metaplasia in Smad4-null mammary glands. We further demonstrated that TGFβ1 treatment degraded β-catenin and induced epithelial-mesenchymal transformation in cultured mammary epithelial cells. However, such actions were blocked in the absence of Smad4. These findings indicate that TGFβ/Smad4 signals play a role in cell fate maintenance during mammary gland development and neoplasia.
Collapse
Affiliation(s)
- Wenmei Li
- Genetics of Development and Disease Branch, NIDDK, NIH, 10/9N105, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1654
|
Abstract
We identified a new gene, called p8, which showed a strong induction during the acute phase of pancreatitis. Further experiments have shown that p8 mRNA is activated in response to several stresses and that its activation is not restricted to pancreatic cells. p8 is a nuclear protein and biochemical and biophysical studies showed that p8 was in many structural aspects very similar to the HMG (high mobility group) proteins, although sharing with them low amino acid sequence homology. Also, p8 was found overexpressed in many human cancers. Therefore, we wondered whether the p8-mediated response to cellular stress was necessary for tumor establishment. Subcutaneous or intraperitoneal injections of transformed p8-expressing fibroblasts led to tumor formation in nude mice, but no tumor was observed with transformed p8-deficient cells. Restoring p8 expression in transformed p8-deficient fibroblasts led to tumor formation demonstrating that p8 expression is necessary for tumor development and suggesting that the stress-response mechanisms governed by p8 are required for tumor establishment.
Collapse
Affiliation(s)
- Sophie Vasseur
- Inserm EMI 0116, 163, avenue de Luminy, Parc Scientifique et Technologique de Luminy, BP 172, 13009 Marseille, France
| | | |
Collapse
|
1655
|
Stuhlmeier KM, Pollaschek C. Differential effect of transforming growth factor beta (TGF-beta) on the genes encoding hyaluronan synthases and utilization of the p38 MAPK pathway in TGF-beta-induced hyaluronan synthase 1 activation. J Biol Chem 2003; 279:8753-60. [PMID: 14676202 DOI: 10.1074/jbc.m303945200] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Unfettered hyaluronan (HA) production is a hallmark of rheumatoid arthritis. The discovery of three genes encoding hyaluronan synthases (HASs) allows for the investigation of the signaling pathways leading to the activation of these genes. Our objective is to further understanding of the regulation of these genes as well as to find ways to prevent undesired gene activation. Human fibroblast-like synoviocytes were used in these experiments. mRNA levels of HAS were monitored by reverse transcriptase-PCR. A series of specific kinase inhibitors were used to investigate intracellular pathways leading to the up-regulation of HAS1. Our experiments, testing a series of stimuli including tumor necrosis factor alpha (TNFalpha), demonstrate that TGF-beta is the most potent stimulus for HAS1 transcription. TGF-beta activates HAS1 in a dose-dependent manner with a maximum effect at a concentration of 0.5-1 ng/ml. TGF-beta-induced HAS1 mRNA can be detected within 60 min and reaches maximal levels at 6 h. Furthermore, TGF-beta treatment leads to an increase in synthase activity as determined by HA ELISA and by in vitro HA synthase assays. In contrast to the activatory effect on HAS1, TGF-beta dose-dependently suppresses HAS3 mRNA. As to the mode of action of TGF-beta-induced HAS1 mRNA activation, our experiments reveal that blocking p38 MAPK inhibited the TGF-beta effect by 90%, blocking the MEK pathway led to an inhibition by 40%, and blocking the JNK pathway had no effect. The presented data might contribute to a better understanding of the role of TGF-beta and of HA in the pathology of diseases.
Collapse
Affiliation(s)
- Karl M Stuhlmeier
- Ludwig Boltzmann Institute for Rheumatology and Balneology, Kurbadstrasse 10, POB 78, A-1107 Vienna, Austria.
| | | |
Collapse
|
1656
|
Bliskovsky V, Ramsay ES, Scott J, DuBois W, Shi W, Zhang S, Qian X, Lowy DR, Mock BA. Frap, FKBP12 rapamycin-associated protein, is a candidate gene for the plasmacytoma resistance locus Pctr2 and can act as a tumor suppressor gene. Proc Natl Acad Sci U S A 2003; 100:14982-7. [PMID: 14634209 PMCID: PMC299869 DOI: 10.1073/pnas.2431627100] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2003] [Accepted: 10/06/2003] [Indexed: 11/18/2022] Open
Abstract
Susceptibility to mouse plasmacytomagenesis is a complex genetic trait controlled by several Pctr loci (Pctr1, Pctr2, etc). Congenic strain analysis narrowed the genetic interval surrounding the Pctr2 locus, and genes identified in the interval were sequenced from susceptible BALB/c and resistant DBA/2 mice. Frap (FKBP12 rapamycin-associated protein, mTOR, RAFT) was the only gene differing in amino acid sequence between alleles that correlated with strain sensitivity to tumor development. The in vitro kinase activity of the BALB/c FRAP allele was lower than the DBA/2 allele; phosphorylation of p53 and PHAS1/4EBP1 (properties of heat and acid stability/eukaryotic initiation factor 4E-binding protein) and autophosphorylation of FRAP were less efficient with the BALB/c allele. FRAP also suppressed transformation of NIH 3T3 cells by ras, with DBA/2 FRAP being more efficient than BALB/c FRAP. Rapamycin, a specific inhibitor of FRAP, did not inhibit growth of plasmacytoma cell lines. These studies identify Frap as a candidate tumor suppressor gene, in contrast to many reports that have focused on its prooncogenic properties. Frap may be similar to Tgfb and E2f in exerting both positive and negative growth-regulatory signals, depending on the timing, pathway, or tumor system involved. The failure of rapamycin to inhibit plasma cell tumor growth suggests that FRAP antagonists may not be appropriate for the treatment of plasma cell tumors. Pctr2 joins Pctr1 in possessing alleles that modify susceptibility to plasmacytomagenesis by encoding differences in efficiency of function (efficiency alleles), rather than all-or-none, gain-of-function, or loss-of-function alleles. By analogy, human cancer may also result from the combined effects of several inefficient alleles.
Collapse
Affiliation(s)
- Valery Bliskovsky
- Laboratory of Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4258, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1657
|
Sekiya T, Adachi S, Kohu K, Yamada T, Higuchi O, Furukawa Y, Nakamura Y, Nakamura T, Tashiro K, Kuhara S, Ohwada S, Akiyama T. Identification of BMP and activin membrane-bound inhibitor (BAMBI), an inhibitor of transforming growth factor-beta signaling, as a target of the beta-catenin pathway in colorectal tumor cells. J Biol Chem 2003; 279:6840-6. [PMID: 14660579 DOI: 10.1074/jbc.m310876200] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Wnt signaling pathway is activated in most human colorectal tumors. Mutational inactivation in the tumor suppressor adenomatous polyposis coli (APC), as well as activation of beta-catenin, causes the accumulation of beta-catenin, which in turn associates with the T cell factor/lymphoid enhancer factor (TCF/LEF) family of transcription factors and activates transcription of their target genes. Here we show that beta-catenin activates transcription of the BMP and activin membrane-bound inhibitor (BAMBI)/NMA gene. The expression level of BAMBI was found to be aberrantly elevated in most colorectal and hepatocellular carcinomas relative to the corresponding non-cancerous tissues. Expression of BAMBI in colorectal tumor cell lines was repressed by a dominant-negative mutant of TCF-4 or by an inhibitor of beta-catenin-TCF interaction, suggesting that beta-catenin is responsible for the aberrant expression of BAMBI in colorectal tumor cells. Furthermore, overexpression of BAMBI inhibited the response of tumor cells to transforming growth factor-beta signaling. These results suggest that beta-catenin interferes with transforming growth factor-beta-mediated growth arrest by inducing the expression of BAMBI, and this may contribute to colorectal and hepatocellular tumorigenesis.
Collapse
Affiliation(s)
- Takashi Sekiya
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1658
|
Kumar-Sinha C, Chinnaiyan AM. Molecular markers to identify patients at risk for recurrence after primary treatment for prostate cancer. Urology 2003; 62 Suppl 1:19-35. [PMID: 14747039 DOI: 10.1016/j.urology.2003.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Accurate prognostication is a prerequisite for accurate therapeutics and management of prostate cancer because indolent tumors may require no intervention, whereas aggressive tumors lead to patient mortality. There is a critical need to define these subgroups of patients with prostate cancer differing in clinical outcome. Prognostic nomograms based on clinical data provide useful predictions of clinical states and outcomes, but they need further refinements to improve accuracy and universality. Genomic and proteomic analyses have provided many novel markers that may help define prognostic parameters based on the underlying biology of prostate cancer progression at the molecular level. These molecular markers are likely to augment traditional prognostic modalities by providing a set of molecularly defined and quantifiable variables. Encompassing the genome, transcriptome, and proteome of prostate cancer will likely provide "molecular signatures" that will bridge prognostication, prediction, and treatment in a single continuum.
Collapse
Affiliation(s)
- Chandan Kumar-Sinha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA
| | | |
Collapse
|
1659
|
Colasante A, Aiello FB, Brunetti M, di Giovine FS. Gene expression of transforming growth factor β receptors I and II in non-small-cell lung tumors. Cytokine 2003; 24:182-9. [PMID: 14596814 DOI: 10.1016/j.cyto.2003.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transforming growth factor (TGF)beta inhibits normal epithelial cell proliferation. A decreased expression of TGFbeta receptors (TbetaR) has been associated with loss of TGFbeta sensitivity and enhanced tumor progression in many types of cancer. Although lung cancer is one of the leading causes of cancer death, a comparative analysis of TbetaR mRNA and protein expression in non-small-cell (NSC) lung tumors has not been performed. Lung tumor tissues and control non-lesional lung tissues were obtained from 17 patients undergoing thoracotomy for primary NSC lung tumors in clinical stage II. Each tissue sample was studied for TbetaRI and TbetaRII mRNA and immunoreactive protein expression, using a semi-quantitative reverse transcription-PCR method, and a quantitative immunohistochemistry method, respectively. TbetaRI protein expression was higher in tumors than in controls (p=0.0005) and a similar trend was present at the mRNA level. TbetaRII protein expression was not significantly different between tumors and controls, however an intense peri-nuclear staining for TbetaRII was observed in several tumor cells. TbetaRII mRNA levels were lower in tumors than in controls (p=0.005) and an inverse relation between TbetaRII mRNA and protein expression was detected in tumors (p=0.0013). Our findings suggest an altered function of the TbetaR system in NSC lung cancer.
Collapse
Affiliation(s)
- Antonella Colasante
- Department of Oncology and Neuroscience, G. D'Annunzio University, Anatomia Patologica, Ospedale SS. Annunziata, Via dei Vestini, 66013, Chieti, Italy.
| | | | | | | |
Collapse
|
1660
|
Kloeker S, Major MB, Calderwood DA, Ginsberg MH, Jones DA, Beckerle MC. The Kindler syndrome protein is regulated by transforming growth factor-beta and involved in integrin-mediated adhesion. J Biol Chem 2003; 279:6824-33. [PMID: 14634021 DOI: 10.1074/jbc.m307978200] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) contributes to tumor invasion and cancer progression by increasing the motility of tumor cells. To identify genes involved in TGF-beta-mediated cell migration, the transcriptional profiles of human mammary epithelial cells (HMEC) treated with TGF-beta were compared with untreated cells by cDNA microarray analysis. One gene up-regulated by TGF-beta was recently named kindlerin (Jobard, F., Bouadjar, B., Caux, F., Hadj-Rabia, S., Has, C., Matsuda, F., Weissenbach, J., Lathrop, M., Prud'homme, J. F., and Fischer, J. (2003) Hum. Mol. Genet. 12, 925-935). This gene is significantly overexpressed in some cancers (Weinstein, E. J., Bourner, M., Head, R., Zakeri, H., Bauer, C., and Mazzarella, R. (2003) Biochim. Biophys. Acta 1637, 207-216), and mutations in this gene lead to Kindler syndrome, an autosomal-recessive genodermatosis. TGF-beta stimulation of HMEC resulted in a marked induction of kindlerin RNA, and Western blotting demonstrated a corresponding increase in protein abundance. Kindlerin displays a putative FERM (four point one ezrin radixin moesin) domain that is closely related to the sequences in talin that interact with integrin beta subunit cytoplasmic domains. The critical residues in the talin FERM domain that mediate integrin binding show a high degree of conservation in kindlerin. Furthermore, kindlerin is recruited into a molecular complex with the beta1A and beta3 integrin cytoplasmic domains. Consistent with these biochemical findings, kindlerin is present at focal adhesions, sites of integrin-rich, membrane-substratum adhesion. Additionally, kindlerin is required for normal cell spreading. Taken together, these data suggest a role for kindlerin in mediating cell processes that depend on integrins.
Collapse
Affiliation(s)
- Susanne Kloeker
- Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah 84112-5550, USA
| | | | | | | | | | | |
Collapse
|
1661
|
Tang B, Vu M, Booker T, Santner SJ, Miller FR, Anver MR, Wakefield LM. TGF-beta switches from tumor suppressor to prometastatic factor in a model of breast cancer progression. J Clin Invest 2003; 112:1116-24. [PMID: 14523048 PMCID: PMC198530 DOI: 10.1172/jci18899] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The TGF-beta signaling network plays a complex role in carcinogenesis because it has the potential to act as either a tumor suppressor or a pro-oncogenic pathway. Currently, it is not known whether TGF-beta can switch from tumor suppressor to pro-oncogenic factor during the course of carcinogenic progression in a single cell lineage with a defined initiating oncogenic event or whether the specific nature of the response is determined by cell type and molecular etiology. To address this question, we have introduced a dominant negative type II TGF-beta receptor into a series of genetically related human breast-derived cell lines representing different stages in the progression process. We show that decreased TGF-beta responsiveness alone cannot initiate tumorigenesis but that it can cooperate with an initiating oncogenic lesion to make a premalignant breast cell tumorigenic and a low-grade tumorigenic cell line histologically and proliferatively more aggressive. In a high-grade tumorigenic cell line, however, reduced TGF-beta responsiveness has no effect on primary tumorigenesis but significantly decreases metastasis. Our results demonstrate a causal role for loss of TGF-beta responsiveness in promoting breast cancer progression up to the stage of advanced, histologically aggressive, but nonmetastatic disease and suggest that at that point TGF-beta switches from tumor suppressor to prometastatic factor.
Collapse
Affiliation(s)
- Binwu Tang
- Laboratory of Cell Regulation and Carcinogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
1662
|
Huang SS, Ling TY, Tseng WF, Huang YH, Tang FM, Leal SM, Huang JS. Cellular growth inhibition by IGFBP‐3 and TGF‐β1requires LRP‐1. FASEB J 2003; 17:2068-81. [PMID: 14597676 DOI: 10.1096/fj.03-0256com] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The type V TGF-beta receptor (TbetaR-V)/IGFBP-3 receptor mediates the IGF-independent growth inhibition induced by IGFBP-3. It also mediates the growth inhibitory response to TGF-beta1 in concert with other TGF-beta receptor types, and its loss may contribute to the malignant phenotype of human carcinoma cells. Here we demonstrate that TbetaR-V is identical to LRP-1/alpha2M receptor as shown by MALDI-TOF analysis of tryptic peptides of TbetaR-V purified from bovine liver. In addition, 125I-IGFBP-3 affinity-labeled TbetaR-V in Mv1Lu cells is immunoprecipitated by antibodies to LRP-1 and TbetaR-V. RAP, an LRP-1 antagonist, inhibits binding of 125I-TGF-beta1 and 125I-IGFBP-3 to TbetaR-V and diminishes IGFBP-3-induced growth inhibition in Mv1Lu cells. Absent or low levels of LRP-1, as with TbetaR-V, have been linked to the malignant phenotype of carcinoma cells. Mutagenized Mv1Lu cells selected for reduced expression of LRP-1 have an attenuated growth inhibitory response to TGF-beta1 and IGFBP-3. LRP-1-deficient mouse embryonic fibroblasts lack a growth inhibitory response to TGF-beta1 and IGFBP-3. On the other hand, stable transfection of H1299 human lung carcinoma cells with LRP-1 cDNA restores the growth inhibitory response. These results suggest that the LRP-1/TbetaR-V/IGFBP-3 receptor is required for the growth inhibitory response to IGFBP-3 and TGF-beta1.
Collapse
Affiliation(s)
- Shuan Shian Huang
- Department of Biochemistry, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, Missouri 63104, USA.
| | | | | | | | | | | | | |
Collapse
|
1663
|
Affiliation(s)
- Peter M Siegel
- Cancer Biology and Genetics Program, Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | |
Collapse
|
1664
|
Robert F, Busby E, Marques MB, Reynolds RE, Carey DE. Phase II study of docetaxel plus enoxaparin in chemotherapy-naive patients with metastatic non-small cell lung cancer: preliminary results. Lung Cancer 2003; 42:237-45. [PMID: 14568692 DOI: 10.1016/s0169-5002(03)00354-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Activation of coagulation appears to play a role in tumor progression. This report describes the preliminary results of a phase II study using docetaxel plus enoxaparin in 15 patients with stage IV non-small cell lung cancer (NSCLC). Time to progression was the primary endpoint. Several surrogate markers of coagulation and angiogenesis were evaluated. Enoxaparin was administered at a daily dose of 1 mg/kg (subcutaneously). The initial dose of docetaxel was 100 mg/m2, given as a 60 min infusion every 21 days with prophylactic dexamethasone. Eight patients achieved an objective response (53%) and four had stable disease, with a median duration of 3.5 months. The median time to progression was 5 months (range, 2 to >15 months). The median survival was 11 months. The most frequent toxicities were neutropenia and asthenia. No significant bleeding or thrombotic events were observed. Eleven patients had elevated D-dimer plasma levels prior to therapy, and seven of these patients with a response or stable disease had a significant decline of the D-dimer during therapy. There were no consistent changes of the plasma levels of the angiogenic factors, except for transforming growth factor-beta-1 (TGF-beta1). The median baseline level of TGF-beta1 prior to therapy was 34,867 pg/ml. Twelve out of 13 patients who achieved a response or stable disease had a significant reduction of the TGF-beta1 levels during therapy. Enoxaparin in combination with chemotherapy was safe and well tolerated in patients with advanced NSCLC. This preliminary data suggests that enoxaparin may prolong the time to progression, and therefore justify the continuation of this trial.
Collapse
Affiliation(s)
- Francisco Robert
- Department of Medicine, Division of Hematology/Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, 1824 6th Avenue South, Birmingham, AL 35294-3300, USA.
| | | | | | | | | |
Collapse
|
1665
|
Lack J, O'Leary JM, Knott V, Yuan X, Rifkin DB, Handford PA, Downing AK. Solution Structure of the Third TB Domain from LTBP1 Provides Insight into Assembly of the Large Latent Complex that Sequesters Latent TGF-β. J Mol Biol 2003; 334:281-91. [PMID: 14607119 DOI: 10.1016/j.jmb.2003.09.053] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Almost all TGF-beta is secreted as part of a large latent complex. This complex is formed from three molecules, a latent transforming growth factor-beta binding protein (LTBP), which plays roles in targeting and activation, a latency associated peptide (LAP), which regulates latency, and the TGF-beta cytokine. LAP is the TGF-beta pro-peptide that is cleaved intracellularly prior to secretion, and TGF-beta binds non-covalently to LAP. Formation of the large latent complex is important for the efficient secretion of TGF-beta. Previous studies have revealed that the LTBP-LAP interaction is mediated by intracellular exchange of a single disulphide bond within the third, and only the third, TB domain (TB3) with LAP. We have previously reported the structure of a homologous TB domain from fibrillin-1. However, TB3 contains a two amino acid insertion, not found in fibrillin-1 TB domains, which is not amenable to molecular modelling. In order to clarify the basis of TB domain function, we have determined the solution NMR structure of TB3(LTBP1). Comparison with the fibrillin-1 TB domain reveals that the two-residue insertion is associated with a significant increase in solvent accessibility of one of the disulphide bonds (linking the second and sixth cysteine residues). Site-directed mutagenesis and NMR studies indicate that this is the only disulphide bond that can be removed without perturbing the TB domain fold. Furthermore, a ring of negatively charged residues has been identified that surrounds this disulphide bond. Homology modelling suggests that the surface properties of TB3 domains from different LTBP isoforms correlate with binding activities. This research provides testable hypotheses regarding the molecular basis of complex formation between LTBPs and LAPs.
Collapse
Affiliation(s)
- Jeremy Lack
- Division of Structural Biology, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | | | | | | | | | | | | |
Collapse
|
1666
|
Waite KA, Eng C. From developmental disorder to heritable cancer: it's all in the BMP/TGF-beta family. Nat Rev Genet 2003; 4:763-73. [PMID: 14526373 DOI: 10.1038/nrg1178] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transforming growth factor-beta (TGF-beta) regulates many cellular processes through complex signal-transduction pathways that have crucial roles in normal development. Disruption of these pathways can lead to a range of diseases, including cancer. Mutations in the genes that encode members of the TGF-beta pathway are involved in vascular diseases as well as gastrointestinal neoplasia. More recently, they have been implicated in Cowden syndrome, which is normally associated with mutations in the phosphatase and tensin homologue gene PTEN. Molecular studies of TGF-beta signalling are now showing why mutations in genes that encode components of this pathway result in inherited cancer and developmental diseases.
Collapse
Affiliation(s)
- Kristin A Waite
- Human Cancer Genetics and Clinical Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
1667
|
Bravo SB, Pampín S, Cameselle-Teijeiro J, Carneiro C, Domínguez F, Barreiro F, Alvarez CV. TGF-β-induced apoptosis in human thyrocytes is mediated by p27kip1 reduction and is overridden in neoplastic thyrocytes by NF-κB activation. Oncogene 2003; 22:7819-30. [PMID: 14586408 DOI: 10.1038/sj.onc.1207029] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Millions of people worldwide suffer goiter, a proliferative disease of the follicular cells of the thyroid that may become neoplastic. Thyroid neoplasms have low proliferative index, low apoptotic index and a high incidence of metastasis. TGF-beta is overexpressed in thyroid follicular tumor cells. To investigate the role of TGF-beta in thyroid tumor progression, we established cultures of human thyrocytes from different proliferative pathologies (Grave's disease, multinodular goiter, follicular adenoma, papillary carcinoma), lymph node metastasis, and a normal thyroid sample. All cultures maintained the thyrocyte phenotype. TGF-beta induced cell-cycle arrest in all cultures, in contrast with results reported for other epithelial tumors. In deprived medium, TGF-beta induced apoptosis in normal thyrocyte cultures and all neoplastic cultures except the metastatic cultures. This apoptosis was mediated by a reduction in p27kip1 levels, inducing cell-cycle initiation. Antisense p27 expression induced apoptosis in the absence of TGF-beta. By contrast, in cells in which p27 was overexpressed, TGF-beta had a survival effect. In growth medium, a net survival effect occurs in neoplastic thyrocytes only, not normal thyrocytes, due to activation of the NF-kappaB survival program. Together, these findings suggest that (a) thyroid neoplasms are due to reduced apoptosis, not increased division, in line with the low proliferative index of these pathologies, and (b) TGF-beta induces apoptosis in normal thyrocytes via p27 reduction, but that in neoplastic thyrocytes this effect is overridden by activation of the NF-kappaB program.
Collapse
Affiliation(s)
- Susana B Bravo
- Department of Physiology, School of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|
1668
|
Gorska AE, Jensen RA, Shyr Y, Aakre ME, Bhowmick NA, Moses HL. Transgenic mice expressing a dominant-negative mutant type II transforming growth factor-beta receptor exhibit impaired mammary development and enhanced mammary tumor formation. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1539-49. [PMID: 14507660 PMCID: PMC1868288 DOI: 10.1016/s0002-9440(10)63510-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We have previously shown that expression of a dominant-negative type II transforming growth factor-beta receptor (DNIIR) in mammary epithelium under control of the MMTV promoter/enhancer causes alveolar hyperplasia and differentiation in virgin mice. Here we show that MMTV-DNIIR female mice have accelerated mammary gland differentiation during early pregnancy with impaired development during late pregnancy and lactation followed by delayed postlactational involution. Mammary tumors, mostly carcinoma in situ, developed spontaneously in the MMTV-DNIIR mice with a long median latency (27.5 months). Crossbreeding to MMTV-transforming growth factor (TGF)-alpha mice to obtain mice expressing both transgenes resulted in mammary tumor formation with a much shorter latency more similar to those expressing only the MMTV-TGF-alpha transgene (<10 months median latency). The major difference in mammary tumors arising in MMTV-TGF-alpha compared to bigenic MMTV-DNIIR/MMTV-TGF-alpha was the marked suppression of tumor invasion by DNIIR transgene expression. Invading carcinoma cells in both MMTV-DNIIR and bigenic animals showed loss of DNIIR transgene expression as determined by in situ hybridization. The data indicate that signaling from endogenous TGF-betas not only plays an important role in normal mammary gland physiology but also can also suppress the early stage of tumor formation and contribute to tumor invasion once carcinomas have developed.
Collapse
MESH Headings
- Animals
- Carcinoma/genetics
- Carcinoma/pathology
- Female
- Gene Expression
- Genes, Dominant
- Genes, Viral/genetics
- Lactation
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Transgenic
- Mutation
- Neoplasm Invasiveness
- Pregnancy
- Promoter Regions, Genetic
- Receptors, Transforming Growth Factor beta/genetics
- Transforming Growth Factor alpha/genetics
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta2
Collapse
Affiliation(s)
- Agnieszka E Gorska
- Vanderbilt-Ingram Cancer Center and the Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
1669
|
Tang B, Vu M, Booker T, Santner SJ, Miller FR, Anver MR, Wakefield LM. TGF-β switches from tumor suppressor to prometastatic factor in a model of breast cancer progression. J Clin Invest 2003. [DOI: 10.1172/jci200318899] [Citation(s) in RCA: 291] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
1670
|
Untergasser G, Gander R, Rumpold H, Heinrich E, Plas E, Berger P. TGF-β cytokines increase senescence-associated beta-galactosidase activity in human prostate basal cells by supporting differentiation processes, but not cellular senescence. Exp Gerontol 2003; 38:1179-88. [PMID: 14580871 DOI: 10.1016/j.exger.2003.08.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The family of transforming growth factors betas (TGF-betas) comprises molecules involved in growth inhibition, stress-induced premature senescence, epithelial mesenchymal transition and differentiation processes. The aim of this study was to clarify the effect of long term exposure of human prostate basal cells to TGF-betas, which are found in high concentrations in prostatic fluid and areas of benign prostatic hyperplasia (BPH). Basal cell cultures established from prostate explants (n=3) were either grown into cellular senescence, or stimulated with TGF-beta1, beta2 and beta3. Similar to cellular senescence, TGF-beta stimulation resulted in an increase of SA-beta galactosidase (SA-beta-gal) activity, flattened and enlarged cell morphology, and down-regulation of the inhibitor of differentiation Id-1. TGF-beta-treated prostate epithelial cells neither showed terminal growth arrest nor induction of important senescence-relevant genes, such as p16(INK4A), IFI-6-16, IGFBP-3 or Dkk-3. Cells stained positive for cytokeratins 8/18, but did not express other lumenal markers, such as prostate-specific antigen and androgen-receptors. TGF-betas increased also the expression of the mesenchymal marker vimentin, indicating that basal epithelial cells underwent differentiation with lumenal and mesenchymal features. In contrast, in vitro-differentiated neuroendocrine-like cells from prostate organoide cultures, expressing chromogranin A and cytokeratin 18, strongly stained positive for SA-beta-gal. Thus, SA-beta-gal activity is not only a marker for senescence, but also for differentiation of human prostate epithelial cells. With regard to the in vivo situation, in addition to cellular senescence, TGF-beta could contribute to the increased number of SA-beta-gal positive epithelial cells in BPH.
Collapse
Affiliation(s)
- G Untergasser
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, Innsbruck A6020, Austria
| | | | | | | | | | | |
Collapse
|
1671
|
Abstract
The thymus is a complex epithelial organ in which thymocyte development is dependent upon the sequential contribution of morphologically and phenotypically distinct stromal cell compartments. It is these microenvironments that provide the unique combination of cellular interactions, cytokines, and chemokines to induce thymocyte precursors to undergo a differentiation program that leads to the generation of functional T cells. Despite the indispensable role of thymic epithelium in the generation of T cells, the mediators of this process and the differentiation pathway undertaken by the primordial thymic epithelial cells are not well defined. There is a lack of lineage-specific cell-surface-associated markers, which are needed to characterize putative thymic epithelial stem cell populations. This review explores the role of thymic stromal cells in T-cell development and thymic organogenesis, as well as the molecular signals that contribute to the growth and expansion of primordial thymic epithelial cells. It highlights recent advances in these areas, which have allowed for a lineage relationship amongst thymic epithelial cell subsets to be proposed. While many fundamental questions remain to be addressed, collectively these works have broadened our understanding of how the thymic epithelium becomes specialized in the ability to support thymocyte differentiation. They should also facilitate the development of novel, rationally based therapeutic strategies for the regeneration and manipulation of thymic function in the treatment of many clinical conditions in which defective T cells have an important etiological role.
Collapse
Affiliation(s)
- Jason Gill
- Department of Pathology and Immunology, Monash University, Faculty of Medicine, Nursing and Health Sciences, Alfred Medical Research and Education Precinct, Prahran, Australia.
| | | | | | | | | | | |
Collapse
|
1672
|
Park CC, Henshall-Powell RL, Erickson AC, Talhouk R, Parvin B, Bissell MJ, Barcellos-Hoff MH. Ionizing radiation induces heritable disruption of epithelial cell interactions. Proc Natl Acad Sci U S A 2003; 100:10728-33. [PMID: 12960393 PMCID: PMC196872 DOI: 10.1073/pnas.1832185100] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Indexed: 02/07/2023] Open
Abstract
Ionizing radiation (IR) is a known human breast carcinogen. Although the mutagenic capacity of IR is widely acknowledged as the basis for its action as a carcinogen, we and others have shown that IR can also induce growth factors and extracellular matrix remodeling. As a consequence, we have proposed that an additional factor contributing to IR carcinogenesis is the potential disruption of critical constraints that are imposed by normal cell interactions. To test this hypothesis, we asked whether IR affected the ability of nonmalignant human mammary epithelial cells (HMEC) to undergo tissue-specific morphogenesis in culture by using confocal microscopy and imaging bioinformatics. We found that irradiated single HMEC gave rise to colonies exhibiting decreased localization of E-cadherin, beta-catenin, and connexin-43, proteins necessary for the establishment of polarity and communication. Severely compromised acinar organization was manifested by the majority of irradiated HMEC progeny as quantified by image analysis. Disrupted cell-cell communication, aberrant cell-extracellular matrix interactions, and loss of tissue-specific architecture observed in the daughters of irradiated HMEC are characteristic of neoplastic progression. These data point to a heritable, nonmutational mechanism whereby IR compromises cell polarity and multicellular organization.
Collapse
Affiliation(s)
- Catherine C Park
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | | | | | | | | | |
Collapse
|
1673
|
Owens DM, Romero MR, Gardner C, Watt FM. Suprabasal alpha6beta4 integrin expression in epidermis results in enhanced tumourigenesis and disruption of TGFbeta signalling. J Cell Sci 2003; 116:3783-91. [PMID: 12902406 DOI: 10.1242/jcs.00725] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inappropriate alpha6beta4 integrin expression correlates with a high risk of tumour progression in stratified squamous epithelia. Targeted expression of alpha6beta4 in the suprabasal layers of transgenic mouse epidermis dramatically increased the frequency of papillomas, carcinomas and metastases induced by chemical carcinogenesis, independent of the beta4 cytoplasmic domain. Suprabasal alpha6beta4 also perturbed transforming growth factor beta (TGFbeta) signalling as demonstrated by decreased nuclear Smad2 in transgenic epidermis and tumours. In cultured keratinocytes, suprabasal alpha6beta4 relieved TGFbeta-mediated growth inhibition and blocked nuclear translocation of activated Smad2/3. Responsiveness to TGFbeta could be restored by inhibiting cadherin-mediated cell-cell adhesion or phosphoinositide 3-kinase (PI3-K) activity, but not by inhibiting mitogen-activated protein kinase (MAPK) activity. These data suggest that suprabasal alpha6beta4 promotes tumourigenesis by preventing TGFbeta from suppressing clonal expansion of initiated cells in the epidermal basal layer.
Collapse
Affiliation(s)
- David M Owens
- Keratinocyte Laboratory, CR-UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | | | | | |
Collapse
|
1674
|
Siegel PM, Shu W, Massagué J. Mad upregulation and Id2 repression accompany transforming growth factor (TGF)-beta-mediated epithelial cell growth suppression. J Biol Chem 2003; 278:35444-50. [PMID: 12824180 DOI: 10.1074/jbc.m301413200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The growth inhibitory cytokine TGF-beta enforces homeostasis of epithelia by activating processes such as cell cycle arrest and apoptosis. Id2 expression is often highest in proliferating epithelial cells and declines during differentiation. Recently, Id2 expression has been found to depend on Myc-Max transcriptional complexes. We observed that TGF-beta signaling inhibits Id2 expression in human and mouse epithelial cell lines from different tissue origins. Furthermore, the observed Id2 down-regulation by TGF-beta in mouse mammary epithelial cells occurs without a concurrent drop in c-Myc levels. However, sustained Id2 repression in these cells and in human keratinocytes coincides with induction of the Myc antagonistic repressors Mad2 and Mad4, decreased formation of Myc-Max heterodimers and the replacement of Myc-Max complexes with Mad-Max complexes on the Id2 promoter. These results argue that induction of Mad expression and Id2 down-regulation are important events during the TGF-beta cytostatic program in epithelial cells.
Collapse
Affiliation(s)
- Peter M Siegel
- Cell Biology Program and Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| | | | | |
Collapse
|
1675
|
Schwarte-Waldhoff I, Schmiegel W. Smad4 transcriptional pathways and angiogenesis. INTERNATIONAL JOURNAL OF GASTROINTESTINAL CANCER 2003; 31:47-59. [PMID: 12622415 DOI: 10.1385/ijgc:31:1-3:47] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Smad4/DPC4 is a tumor suppressor gene frequently inactivated in gastrointestinal carcinomas. Smad4 encodes a key intracellular transmitter for signals of the TGF-beta superfamily of cytokines. TGF-beta potently inhibits the growth of normal epithelial cells but tumor cells are frequently resistant; thus, it has been assumed that loss of Smad4 during tumor progression relieves this inhibition. Mediating TGF-beta responses is only one of the many putative functions of Smad4 as a signaling molecule. Smad proteins are versatile transcriptional co-modulators whose activities depend on the genetic makeup of a cell. We have used restoration of Smad4 in deficient cancer cells as an unbiased approach to decipher Smad4's tumor suppressor functions. Stable reexpression of Smad4 in human colon and pancreatic cancer cells potently suppressed tumor growth in vivo in nude mice. Surprisingly, it was not adequate to suppress tumor cell growth in vitro, nor did it restore TGF-beta responsiveness. Rather, Smad4 restoration influenced angiogenesis, decreasing expression of vascular endothelial growth factor and increasing expression of thrombospondin-1. These findings suggest that the acquisition of TGF-beta resistance and loss of Smad4 may be independent consecutive events in the tumorigenic process. They define the control of an angiogenic switch as a novel alternative mechanism of tumor suppression for Smad4.
Collapse
|
1676
|
Quintanilla M, Ramirez JR, Pérez-Gómez E, Romero D, Velasco B, Letarte M, López-Novoa JM, Bernabéu C. Expression of the TGF-beta coreceptor endoglin in epidermal keratinocytes and its dual role in multistage mouse skin carcinogenesis. Oncogene 2003; 22:5976-85. [PMID: 12955076 DOI: 10.1038/sj.onc.1206841] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endoglin is an integral membrane glycoprotein primarily expressed in the vascular endothelium, but also found on macrophages and stromal cells. It binds several members of the transforming growth factor (TGF)-beta family of growth factors and modulates TGF-beta(1)-dependent cellular responses. However, it lacks cytoplasmic signaling motifs and is considered as an auxiliary receptor for TGF-beta. We show here that endoglin is expressed in mouse and human epidermis and in skin appendages, such as hair follicles and sweat glands, as determined by immunohistochemistry. In normal interfollicular epidermis, endoglin was restricted to basal keratinocytes and absent in differentiating cells of suprabasal layers. Follicular expression of endoglin was high in hair bulb keratinocytes, but decreased in parts distal from the bulb. To address the role of endoglin in skin carcinogenesis in vivo, Endoglin heterozygous mice were subjected to long-term chemical carcinogenesis treatment. Reduction in endoglin had a dual effect during multistage carcinogenesis, by inhibiting the early appearance of benign papillomas, but increasing malignant progression to highly undifferentiated carcinomas. Our results are strikingly similar to those previously reported for transgenic mice overexpressing TGF-beta(1) in the epidermis. These data suggest that endoglin might attenuate TGF-beta(1) signaling in normal epidermis and interfere with progression of skin carcinogenesis.
Collapse
Affiliation(s)
- Miguel Quintanilla
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier 4, Madrid 28029, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
1677
|
Yang YC, Piek E, Zavadil J, Liang D, Xie D, Heyer J, Pavlidis P, Kucherlapati R, Roberts AB, Böttinger EP. Hierarchical model of gene regulation by transforming growth factor beta. Proc Natl Acad Sci U S A 2003; 100:10269-74. [PMID: 12930890 PMCID: PMC193550 DOI: 10.1073/pnas.1834070100] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2003] [Indexed: 11/18/2022] Open
Abstract
Transforming growth factor betas (TGF-betas) regulate key aspects of embryonic development and major human diseases. Although Smad2, Smad3, and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinases (MAPKs) have been proposed as key mediators in TGF-beta signaling, their functional specificities and interactivity in controlling transcriptional programs in different cell types and (patho)physiological contexts are not known. We investigated expression profiles of genes controlled by TGF-beta in fibroblasts with ablations of Smad2, Smad3, and ERK MAPK. Our results suggest that Smad3 is the essential mediator of TGF-beta signaling and directly activates genes encoding regulators of transcription and signal transducers through Smad3/Smad4 DNA-binding motif repeats that are characteristic for immediate-early target genes of TGF-beta but absent in intermediate target genes. In contrast, Smad2 and ERK predominantly transmodulated regulation of both immediate-early and intermediate genes by TGF-beta/Smad3. These results suggest a previously uncharacterized hierarchical model of gene regulation by TGF-beta in which TGF-beta causes direct activation by Smad3 of cascades of regulators of transcription and signaling that are transmodulated by Smad2 and/or ERK.
Collapse
Affiliation(s)
- Yaw-Ching Yang
- Departments of Medicine and Molecular Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1678
|
Melchionda F, Fry TJ, Mackall CL. Harnessing the immune modulatory effects of IL7 for immunotherapy. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1529-1049(03)00046-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
1679
|
Tarakanova VL, Wold WSM. Transforming growth factor beta1 receptor II is downregulated by E1A in adenovirus-infected cells. J Virol 2003; 77:9324-36. [PMID: 12915548 PMCID: PMC187388 DOI: 10.1128/jvi.77.17.9324-9336.2003] [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: 12/14/2022] Open
Abstract
Transforming growth factor beta1 (TGF-beta1) signaling is compromised in many tumors, thereby allowing the tumor to escape the growth-inhibitory and proapoptotic activities of the cytokine. Human adenoviruses interfere with a number of cellular pathways involved in cell cycle regulation and apoptosis, initially placing the cell in a "tumor-like" state by forcing quiescent cells into the cell cycle and also inhibiting apoptosis. We report that adenovirus-infected cells resemble tumor cells in that TGF-beta1 signaling is inhibited. The levels of TGF-beta1 receptor II (TbetaRII) in adenovirus-infected cells were decreased, and this decrease was mapped, by using virus mutants, to the E1A gene and to amino acids 2 to 36 and the C-terminal binding protein binding site in the E1A protein. The decrease in the TbetaRII protein was accompanied by a decrease in TbetaRII mRNA. The decrease in TbetaRII protein levels in adenovirus-infected cells was greater than the decrease in TbetaRII mRNA, suggesting that downregulation of the TbetaRII protein may occur through more than one mechanism. Surprisingly in this context, the half-lives of the TbetaRII protein in infected and uninfected cells were similar. TGF-beta1 signaling was compromised in cells infected with wild-type adenovirus, as measured with 3TP-lux, a TGF-beta-sensitive reporter plasmid expressing luciferase. Adenovirus mutants deficient in TbetaRII downregulation did not inhibit TGF-beta1 signaling. TGF-beta1 pretreatment reduced the relative abundance of adenovirus structural proteins in infected cells, an effect that was potentiated when cells were infected with mutants incapable of modulating the TGF-beta signaling pathway. These results raise the possibility that inhibition of TGF-beta signaling by E1A is a means by which adenovirus counters the antiviral defenses of the host.
Collapse
Affiliation(s)
- Vera L Tarakanova
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA.
| | | |
Collapse
|
1680
|
Yasuda J, Tsuchiya A, Yamada T, Sakamoto M, Sekiya T, Hirohashi S. Nemo-like kinase induces apoptosis in DLD-1 human colon cancer cells. Biochem Biophys Res Commun 2003; 308:227-33. [PMID: 12901858 DOI: 10.1016/s0006-291x(03)01343-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Deregulation of Wnt/beta-catenin signaling is thought to play a critical role in human carcinogenesis. Nemo-like kinase (NLK) is an evolutionarily conserved serine/threonine kinase that suppresses beta-catenin/T-cell factor (TCF) complex transcriptional activity through phosphorylation of TCF. Since NLK may be a tumor suppressor as a negative regulator of Wnt/beta-catenin pathway, we established tetracycline-inducible NLK and its kinase-negative mutant expression in DLD-1 human colon cancer cells to analyze the effect of NLK on cell growth and viability. The induction of wild-type NLK in DLD-1 cells caused suppression of cell growth whereas the kinase-negative mutant did not. Flow cytometry indicated that NLK expression increased the number of apoptotic cells but did not induce obvious cell cycle arrest. Apoptosis induction by wild-type NLK was confirmed using TUNEL assays. Our results suggest that overexpression of NLK may have targets other than TCF for induction of apoptosis in human colon carcinoma cells.
Collapse
Affiliation(s)
- Jun Yasuda
- Cancer Transcriptome Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | | | | | | | | | | |
Collapse
|
1681
|
Adkins HB, Bianco C, Schiffer SG, Rayhorn P, Zafari M, Cheung AE, Orozco O, Olson D, De Luca A, Chen LL, Miatkowski K, Benjamin C, Normanno N, Williams KP, Jarpe M, LePage D, Salomon D, Sanicola M. Antibody blockade of the Cripto CFC domain suppresses tumor cell growth in vivo. J Clin Invest 2003; 112:575-87. [PMID: 12925698 PMCID: PMC171388 DOI: 10.1172/jci17788] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cripto, a cell surface-associated protein belonging to the EGF-CFC family of growth factor-like molecules, is overexpressed in many human solid tumors, including 70-80% of breast and colon tumors, yet how it promotes cell transformation is unclear. During embryogenesis, Cripto complexes with Alk4 via its unique cysteine-rich CFC domain to facilitate signaling by the TGF-beta ligand Nodal. We report, for the first time to our knowledge, that Cripto can directly bind to another TGF-beta ligand, Activin B, and that Cripto overexpression blocks Activin B growth inhibition of breast cancer cells. This result suggests a novel mechanism for antagonizing Activin signaling that could promote tumorigenesis by deregulating growth homeostasis. We show that an anti-CFC domain antibody, A8.G3.5, both disrupts Cripto-Nodal signaling and reverses Cripto blockade of Activin B-induced growth suppression by blocking Cripto's association with either Alk4 or Activin B. In two xenograft models, testicular and colon cancer, A8.G3.5 inhibited tumor cell growth by up to 70%. Both Nodal and Activin B expression was found in the xenograft tumor, suggesting that either ligand could be promoting tumorigenesis. These data validate that functional blockade of Cripto inhibits tumor growth and highlight antibodies that block Cripto signaling mediated through its CFC domain as an important class of antibodies for further therapeutic development.
Collapse
Affiliation(s)
- Heather B Adkins
- Biogen Inc., 14 Cambridge Center, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1682
|
Abstract
The cell-surface associated molecule Cripto is overexpressed in a wide range of epithelial cancers, yet little is known about potential mechanisms by which Cripto expression might enhance tumorigenesis. A new study reveals that binding of Cripto to the TGF-beta ligand Activin B can block Activin B-mediated suppression of cell proliferation. Furthermore, this study also demonstrates that antibody blockade of Cripto function may prove useful in the inhibition of tumorigenesis.
Collapse
Affiliation(s)
- Michael M Shen
- Center for Advanced Biotechnology and Medicine, 679 Hoes Lane, Piscataway, New Jersey 08854, USA.
| |
Collapse
|
1683
|
Grünert S, Jechlinger M, Beug H. Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol 2003; 4:657-65. [PMID: 12923528 DOI: 10.1038/nrm1175] [Citation(s) in RCA: 522] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In contrast to the aberrant control of proliferation, apoptosis, angiogenesis and lifespan, the cellular mechanisms that cause local invasion and metastasis of tumour cells are still poorly understood. New experimental approaches have identified different types of epithelial-plasticity changes in tumour cells towards fibroblastoid phenotypes as crucial events that occur during metastasis, and many molecules and signalling pathways cooperate to trigger these processes.
Collapse
Affiliation(s)
- Stefan Grünert
- Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria.
| | | | | |
Collapse
|
1684
|
Gillanders E, Hank Juo SH, Holland EA, Jones M, Nancarrow D, Freas-Lutz D, Sood R, Park N, Faruque M, Markey C, Kefford RF, Palmer J, Bergman W, Bishop DT, Tucker MA, Bressac-de Paillerets B, Hansson J, Stark M, Gruis N, Bishop JN, Goldstein AM, Bailey-Wilson JE, Mann GJ, Hayward N, Trent J. Localization of a novel melanoma susceptibility locus to 1p22. Am J Hum Genet 2003; 73:301-13. [PMID: 12844286 PMCID: PMC1180369 DOI: 10.1086/377140] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2003] [Accepted: 05/20/2003] [Indexed: 11/03/2022] Open
Abstract
Over the past 20 years, the incidence of cutaneous malignant melanoma (CMM) has increased dramatically worldwide. A positive family history of the disease is among the most established risk factors for CMM; it is estimated that 10% of CMM cases result from an inherited predisposition. Although mutations in two genes, CDKN2A and CDK4, have been shown to confer an increased risk of CMM, they account for only 20%-25% of families with multiple cases of CMM. Therefore, to localize additional loci involved in melanoma susceptibility, we have performed a genomewide scan for linkage in 49 Australian pedigrees containing at least three CMM cases, in which CDKN2A and CDK4 involvement has been excluded. The highest two-point parametric LOD score (1.82; recombination fraction [theta] 0.2) was obtained at D1S2726, which maps to the short arm of chromosome 1 (1p22). A parametric LOD score of 4.65 (theta=0) and a nonparametric LOD score of 4.19 were found at D1S2779 in nine families selected for early age at onset. Additional typing yielded seven adjacent markers with LOD scores >3 in this subset, with the highest parametric LOD score, 4.95 (theta=0) (nonparametric LOD score 5.37), at D1S2776. Analysis of 33 additional multiplex families with CMM from several continents provided further evidence for linkage to the 1p22 region, again strongest in families with the earliest mean age at diagnosis. A nonparametric ordered sequential analysis was used, based on the average age at diagnosis in each family. The highest LOD score, 6.43, was obtained at D1S2779 and occurred when the 15 families with the earliest ages at onset were included. These data provide significant evidence of a novel susceptibility gene for CMM located within chromosome band 1p22.
Collapse
Affiliation(s)
- Elizabeth Gillanders
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Suh-Hang Hank Juo
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Elizabeth A. Holland
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - MaryPat Jones
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Derek Nancarrow
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Diana Freas-Lutz
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Raman Sood
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Naeun Park
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Mezbah Faruque
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Carol Markey
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Richard F. Kefford
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Jane Palmer
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Wilma Bergman
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - D. Timothy Bishop
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Margaret A. Tucker
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Brigitte Bressac-de Paillerets
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Johan Hansson
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - The Lund Melanoma Study Group
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Mitchell Stark
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Nelleke Gruis
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Julia Newton Bishop
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Alisa M. Goldstein
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Joan E. Bailey-Wilson
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Graham J. Mann
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Nicholas Hayward
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | - Jeffrey Trent
- Cancer Genetics Branch, National Human Genome Research Institute, and Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda; Columbia Genome Center and Department of Epidemiology, Columbia University, New York; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney; Queensland Cancer Fund Research Unit, Brisbane, Australia; Department of Dermatology and Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands; Genetic Epidemiology Division, Cancer Research UK Clinical Centre, Leeds, United Kingdom; Institut Gustave Rousy, Villejuif, France; Department of Oncology-Pathology, Karolinska Hospital, Stockholm; Lund Cancer Center Department of Oncology, University Hospital, Lund, Sweden; Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore; and Translational Genomics Research Institute, Phoenix
| | | |
Collapse
|
1685
|
Fjellbirkeland L, Cambier S, Broaddus VC, Hill A, Brunetta P, Dolganov G, Jablons D, Nishimura SL. Integrin alphavbeta8-mediated activation of transforming growth factor-beta inhibits human airway epithelial proliferation in intact bronchial tissue. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:533-42. [PMID: 12875973 PMCID: PMC1868219 DOI: 10.1016/s0002-9440(10)63681-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Transforming growth factor (TGF)-beta is a potent multifunctional cytokine that is an essential regulator of epithelial proliferation. Because TGF-beta is expressed almost entirely in a latent state in vivo, a major source of regulation of TGF-beta function is its activation. A subset of integrins, alphavbeta8 and alphavbeta6, which are expressed in the human airway, has recently been shown to activate latent TGF-beta in vitro, suggesting a regulatory role for integrins in TGF-beta function in vivo. Here we have developed a novel, biologically relevant experimental model of human airway epithelium using intact human bronchial tissue. We have used this model to determine the function of integrin-mediated activation of TGF-beta in the airway. In human bronchial fragments cultured in vitro, authentic epithelial-stromal interactions were maintained and integrin and TGF-beta expression profiles correlated with profiles found in normal lung. In addition, in this model, we found that either the integrin alphavbeta8 or TGF-beta could inhibit airway epithelial cell proliferation. Furthermore, we found that one mechanism of integrin-alphavbeta8-dependent inhibition of cell proliferation was through activation of TGF-beta because anti-beta8 antibody blocked the majority (76%) of active TGF-beta released from bronchial fragments. These data provide compelling evidence for a functional role for integrin-mediated activation of TGF-beta in control of human airway epithelial proliferation in vivo.
Collapse
Affiliation(s)
- Lars Fjellbirkeland
- Department of Anatomic Pathology and Lung Biology Center, San Francisco General Hospital, University of California at San Francisco/Mt. Zion Cancer Center, San Francisco, California 94110, USA
| | | | | | | | | | | | | | | |
Collapse
|
1686
|
Affiliation(s)
- Yansong Bian
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Medical School, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | | | | | | |
Collapse
|
1687
|
Lee PSW, Chang C, Liu D, Derynck R. Sumoylation of Smad4, the common Smad mediator of transforming growth factor-beta family signaling. J Biol Chem 2003; 278:27853-63. [PMID: 12740389 DOI: 10.1074/jbc.m301755200] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transforming growth factor-beta (TGF-beta) and TGF-beta-related factors regulate cell growth, differentiation, and apoptosis, and play key roles in normal development and tumorigenesis. TGF-beta family-induced changes in gene expression are mediated by serine/threonine kinase receptors at the cell surface and Smads as intracellular effectors. Receptor-activated Smads combine with a common Smad4 to translocate into the nucleus where they cooperate with other transcription factors to activate or repress transcription. The activities of the receptor-activated Smads are controlled by post-translational modifications such as phosphorylation and ubiquitylation. Here we show that Smad4 is modified by sumoylation. Sumoylation of Smad4 was enhanced by the conjugating enzyme Ubc9 and members of the PIAS family of SUMO ligases. A major sumoylation site in Smad4 was localized to Lys-159 in its linker segment with an additional site at Lys-113 in the MH-1 domain. Increased sumoylation in the presence of the PIASy E3 ligase correlated with targeting of Smad4 to subnuclear speckles that contain SUMO-1 and PIASy. Replacement of lysines 159 and 113 by arginines or increased sumoylation enhanced the stability of Smad4, and transcription in mammalian cells and Xenopus embryos. These observations suggest a role for Smad4 sumoylation in the regulation of TGF-beta signaling through Smads.
Collapse
Affiliation(s)
- Pierre S W Lee
- Department of Growth and Development, University of California, San Francisco, California 94143-0640, USA
| | | | | | | |
Collapse
|
1688
|
Prunier C, Pessah M, Ferrand N, Seo SR, Howe P, Atfi A. The oncoprotein Ski acts as an antagonist of transforming growth factor-beta signaling by suppressing Smad2 phosphorylation. J Biol Chem 2003; 278:26249-57. [PMID: 12732634 DOI: 10.1074/jbc.m304459200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The phosphorylation of Smad2 and Smad3 by the transforming growth factor (TGF)-beta-activated receptor kinases and their subsequent heterodimerization with Smad4 and translocation to the nucleus form the basis for a model how Smad proteins work to transmit TGF-beta signals. The transcriptional activity of Smad2-Smad4 or Smad3-Smad4 complexes can be limited by the corepressor Ski, which is believed to interact with Smad complexes on TGF-beta-responsive promoters and represses their ability to activate TGF-beta target genes by assembling on DNA a repressor complex containing histone deacetylase. Here we show that Ski can block TGF-beta signaling by interfering with the phosphorylation of Smad2 and Smad3 by the activated TGF-beta type I receptor. Furthermore, we demonstrate that overexpression of Ski induces the assembly of Smad2-Smad4 and Smad3-Smad4 complexes independent of TGF-beta signaling. The ability of Ski to engage Smad proteins in nonproductive complexes provides new insights into the molecular mechanism used by Ski for disabling TGF-beta signaling.
Collapse
Affiliation(s)
- Celine Prunier
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
1689
|
Ju HR, Jung U, Sonn CH, Yoon SR, Jeon JH, Yang Y, Lee KN, Choi I. Aberrant signaling of TGF-beta1 by the mutant Smad4 in gastric cancer cells. Cancer Lett 2003; 196:197-206. [PMID: 12860278 DOI: 10.1016/s0304-3835(03)00237-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
TGF-beta1 has been known to suppress the growth of gastric cancer cells. Interestingly, TGF-beta1 treatment increased the proliferation of human gastric cancer cell line, SNU-216 cells, while it reduced the proliferation of other tumor cells including SNU-620 cells. TGF-beta1-mediated down-regulation of c-Myc and induction of p21CIP1 were observed in SNU-620, but there was no change in SNU-216 in response to TGF-beta1. Similarly, TGF-beta1 receptors were upregulated by TGF-beta1 treatment in SNU-620, but they were not responded in SNU-216. By a single strand conformation polymorphism analysis, a repeated insertion of 37 nucleotides in the exon 8 of Smad4, resulting in premature termination at codon 362, was found in SNU-216. Furthermore, this truncated Smad4 functioned as a dominant negative form in TGF-beta1-mediated reporter activity and TGF-beta1 receptor expression. However, the proliferation of tumor cells was not affected by Smad4 mutation, but it was modulated by PD98059. Taken together, a mutation in Smad4 in addition to mitogen-activated protein kinase altered the TGF-beta1-mediated signaling, which is one of key events of gastric tumorigenesis.
Collapse
Affiliation(s)
- Hyang Ran Ju
- Laboratory of Immunology, Korea Research Institute of Bioscience and Biotechnology, Eoun-Dong 52, Yusong, Taejon 305-333, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
1690
|
Siegel PM, Shu W, Cardiff RD, Muller WJ, Massagué J. Transforming growth factor beta signaling impairs Neu-induced mammary tumorigenesis while promoting pulmonary metastasis. Proc Natl Acad Sci U S A 2003; 100:8430-5. [PMID: 12808151 PMCID: PMC166246 DOI: 10.1073/pnas.0932636100] [Citation(s) in RCA: 339] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The influence of transforming growth factor beta (TGF-beta) signaling on Neu-induced mammary tumorigenesis and metastasis was examined with transgenic mouse models. We generated mice expressing an activated TGF-beta type I receptor or dominant negative TGF-beta type II receptor under control of the mouse mammary tumor virus promoter. When crossed with mice expressing activated forms of the Neu receptor tyrosine kinase that selectively couple to the Grb2 or Shc signaling pathways the activated type I receptor increased the latency of mammary tumor formation but also enhanced the frequency of extravascular lung metastasis. Conversely, expression of the dominant negative type II receptor decreased the latency of Neu-induced mammary tumor formation while significantly reducing the incidence of extravascular lung metastases. These observations argue that TGF-beta can promote the formation of lung metastases while impairing Neu-induced tumor growth and suggest that extravasation of breast cancer cells from pulmonary vessels is a point of action of TGF-beta in the metastatic process.
Collapse
MESH Headings
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/physiology
- Adaptor Proteins, Signal Transducing
- Adaptor Proteins, Vesicular Transport
- Animals
- Apoptosis
- Drosophila Proteins
- Female
- GRB2 Adaptor Protein
- Genes, Dominant
- Genes, erbB-2
- Humans
- Lactation
- Ligases/antagonists & inhibitors
- Ligases/physiology
- Lung Neoplasms/etiology
- Lung Neoplasms/secondary
- Mammary Glands, Animal/growth & development
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Transgenic
- Mitotic Index
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/physiology
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/physiology
- Pregnancy
- Protein Serine-Threonine Kinases
- Proteins/antagonists & inhibitors
- Proteins/physiology
- Receptor, Transforming Growth Factor-beta Type I
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/physiology
- Sequence Deletion
- Shc Signaling Adaptor Proteins
- Signal Transduction
- Src Homology 2 Domain-Containing, Transforming Protein 1
- Time Factors
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta/physiology
- Tumor Cells, Cultured
- Ubiquitin-Protein Ligases
Collapse
Affiliation(s)
- Peter M. Siegel
- Cell Biology Program and Howard Hughes Medical
Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021;
Department of Pathology, School of Medicine,
University of California, Davis, CA 95616; and
Departments of Biochemistry and Medicine,
Molecular Oncology Group, McGill University Health Centre, McGill University,
Montreal, QC H3A 1A1, Canada
| | - Weiping Shu
- Cell Biology Program and Howard Hughes Medical
Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021;
Department of Pathology, School of Medicine,
University of California, Davis, CA 95616; and
Departments of Biochemistry and Medicine,
Molecular Oncology Group, McGill University Health Centre, McGill University,
Montreal, QC H3A 1A1, Canada
| | - Robert D. Cardiff
- Cell Biology Program and Howard Hughes Medical
Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021;
Department of Pathology, School of Medicine,
University of California, Davis, CA 95616; and
Departments of Biochemistry and Medicine,
Molecular Oncology Group, McGill University Health Centre, McGill University,
Montreal, QC H3A 1A1, Canada
| | - William J. Muller
- Cell Biology Program and Howard Hughes Medical
Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021;
Department of Pathology, School of Medicine,
University of California, Davis, CA 95616; and
Departments of Biochemistry and Medicine,
Molecular Oncology Group, McGill University Health Centre, McGill University,
Montreal, QC H3A 1A1, Canada
| | - Joan Massagué
- Cell Biology Program and Howard Hughes Medical
Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021;
Department of Pathology, School of Medicine,
University of California, Davis, CA 95616; and
Departments of Biochemistry and Medicine,
Molecular Oncology Group, McGill University Health Centre, McGill University,
Montreal, QC H3A 1A1, Canada
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
1691
|
Abstract
TGF-beta1 induces cell cycle activation in mouse embryonic fibroblasts by down regulation of p27(Kip1) but it can also induce delay of EGF-induced cell cycle activation in these cells under similar conditions. In an attempt to determine the basis for these responses, the study of early TGF-beta1-induced signal transduction pathways in the presence and absence of EGF was undertaken. It is proposed that a likely target for the inhibition by TGF-beta1 of the early EGF-induced p42/p44 MAPK is at the c-Raf locus. The finding that the catalytic subunits of PKA are associated with Raf-1 within minutes following application of TGF-beta1 but not EGF in fibroblasts arrested in early G1 is suggestive of a role of PKA-Raf-1 interaction in TGF-beta1 induced delay of EGF-induced cell cycle kinetics. A model for TGF-beta1 induced translocation to the plasma membrane-associated Raf-1 is proposed. Reports that Rho-like GTPase activity is critical for the activation of TGF-beta1 downstream pathways raises the question as to whether Rho proteins are involved in these observed TGF-beta1-induced responses. Post-receptor signaling mechanisms for TGF-beta1 and cross-talk with PKA-mediated pathways are examined in an effort to explain the modulation by TGF-beta1 of mitogen-induced cell proliferation in mesenchymal cells.
Collapse
Affiliation(s)
- Charles E Wenner
- Department of Cell and Molecular Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | | |
Collapse
|
1692
|
Kurisaki K, Kurisaki A, Valcourt U, Terentiev AA, Pardali K, Ten Dijke P, Heldin CH, Ericsson J, Moustakas A. Nuclear factor YY1 inhibits transforming growth factor beta- and bone morphogenetic protein-induced cell differentiation. Mol Cell Biol 2003; 23:4494-510. [PMID: 12808092 PMCID: PMC164850 DOI: 10.1128/mcb.23.13.4494-4510.2003] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2002] [Revised: 12/19/2002] [Accepted: 04/07/2003] [Indexed: 12/19/2022] Open
Abstract
Smad proteins transduce transforming growth factor beta (TGF-beta) and bone morphogenetic protein (BMP) signals that regulate cell growth and differentiation. We have identified YY1, a transcription factor that positively or negatively regulates transcription of many genes, as a novel Smad-interacting protein. YY1 represses the induction of immediate-early genes to TGF-beta and BMP, such as the plasminogen activator inhibitor 1 gene (PAI-1) and the inhibitor of differentiation/inhibitor of DNA binding 1 gene (Id-1). YY1 inhibits binding of Smads to their cognate DNA elements in vitro and blocks Smad recruitment to the Smad-binding element-rich region of the PAI-1 promoter in vivo. YY1 interacts with the conserved N-terminal Mad homology 1 domain of Smad4 and to a lesser extent with Smad1, Smad2, and Smad3. The YY1 zinc finger domain mediates the association with Smads and is necessary for the repressive effect of YY1 on Smad transcriptional activity. Moreover, downregulation of endogenous YY1 by antisense and small interfering RNA strategies results in enhanced transcriptional responses to TGF-beta or BMP. Ectopic expression of YY1 inhibits, while knockdown of endogenous YY1 enhances, TGF-beta- and BMP-induced cell differentiation. In contrast, overexpression or knockdown of YY1 does not affect growth inhibition induced by TGF-beta or BMP. Accordingly, YY1 does not interfere with the regulation of immediate-early genes involved in the TGF-beta growth-inhibitory response, the cell cycle inhibitors p15 and p21, and the proto-oncogene c-myc. In conclusion, YY1 represses Smad transcriptional activities in a gene-specific manner and thus regulates cell differentiation induced by TGF-beta superfamily pathways.
Collapse
Affiliation(s)
- Keiko Kurisaki
- Ludwig Institute for Cancer Research, Biomedical Center, SE-751 24 Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
1693
|
Abstract
Matrix metalloproteinases (MMPs) are a growing family of metalloendopeptidases that cleave the protein components of the extracellular matrix and thereby play a central role in tissue remodelling. For many years following their discovery, MMPs were believed to function primarily as regulators of ECM composition and to facilitate cell migration simply by removing barriers such as collagen. It is becoming increasingly clear, however, that MMPs are implicated in the functional regulation of a host of non-ECM molecules that include growth factors and their receptors, cytokines and chemokines, adhesion receptors and cell surface proteoglycans, and a variety of enzymes. MMPs therefore play an important role in the control of cellular interactions with and response to their environment in conditions that promote tissue turnover, be they physiological, such as normal development, or pathological, such as inflammation and cancer. This review summarizes some of the recent discoveries that have shed new light on the role of MMPs in physiology and disease.
Collapse
Affiliation(s)
- Ivan Stamenkovic
- Experimental Pathology Division, Institut Universitaire de Pathologie, Université de Lausanne, 25 Rue du Bagnon, CH-1011 Lausanne, Switzerland
| |
Collapse
|
1694
|
Abstract
Maintenance of epithelial tissues needs the stroma. When the epithelium changes, the stroma inevitably follows. In cancer, changes in the stroma drive invasion and metastasis, the hallmarks of malignancy. Stromal changes at the invasion front include the appearance of myofibroblasts, cells sharing characteristics with fibroblasts and smooth muscle cells. The main precursors of myofibroblasts are fibroblasts. The transdifferentiation of fibroblasts into myofibroblasts is modulated by cancer cell-derived cytokines, such as transforming growth factor-beta (TGF-beta). TGF-beta causes cancer progression through paracrine and autocrine effects. Paracrine effects of TGF-beta implicate stimulation of angiogenesis, escape from immunosurveillance and recruitment of myofibroblasts. Autocrine effects of TGF-beta in cancer cells with a functional TGF-beta receptor complex may be caused by a convergence between TGF-beta signalling and beta-catenin or activating Ras mutations. Experimental and clinical observations indicate that myofibroblasts produce pro-invasive signals. Such signals may also be implicated in cancer pain. N-Cadherin and its soluble form act as invasion-promoters. N-Cadherin is expressed in invasive cancer cells and in host cells such as myofibroblasts, neurons, smooth muscle cells, and endothelial cells. N-Cadherin-dependent heterotypic contacts may promote matrix invasion, perineural invasion, muscular invasion, and transendothelial migration; the extracellular, the juxtamembrane and the beta-catenin binding domain of N-cadherin are implicated in positive invasion signalling pathways. A better understanding of stromal contributions to cancer progression will likely increase our awareness of the importance of the combinatorial signals that support and promote growth, dedifferentiation, invasion, and ectopic survival and eventually result in the identification of new therapeutics targeting the stroma.
Collapse
Affiliation(s)
- Olivier De Wever
- Laboratory of Experimental Cancerology, Department of Radiotherapy and Nuclear Medicine, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | | |
Collapse
|
1695
|
Warburton D, Bellusci S, Del Moral PM, Kaartinen V, Lee M, Tefft D, Shi W. Growth factor signaling in lung morphogenetic centers: automaticity, stereotypy and symmetry. Respir Res 2003; 4:5. [PMID: 12818006 PMCID: PMC185249 DOI: 10.1186/1465-9921-4-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2002] [Revised: 01/28/2003] [Accepted: 02/17/2003] [Indexed: 12/16/2022] Open
Abstract
Lung morphogenesis is stereotypic, both for lobation and for the first several generations of airways, implying mechanistic control by a well conserved, genetically hardwired developmental program. This program is not only directed by transcriptional factors and peptide growth factor signaling, but also co-opts and is modulated by physical forces. Peptide growth factors signal within repeating epithelial-mesenchymal temporospatial patterns that constitute morphogenetic centers, automatically directing millions of repetitive events during both stereotypic branching and nonstereotypic branching as well as alveolar surface expansion phases of lung development. Transduction of peptide growth factor signaling within these centers is finely regulated at multiple levels. These may include ligand expression, proteolytic activation of latent ligand, ligand bioavailability, ligand binding proteins and receptor affinity and presentation, receptor complex assembly and kinase activation, phosphorylation and activation of adapter and messenger protein complexes as well as downstream events and cross-talk both inside and outside the nucleus. Herein we review the critical Sonic Hedgehog, Fibroblast Growth Factor, Bone Morphogenetic Protein, Vascular Endothelial Growth Factor and Transforming Growth Factorbeta signaling pathways and propose how they may be functionally coordinated within compound, highly regulated morphogenetic gradients that drive first stereotypic and then non-stereotypic, automatically repetitive, symmetrical as well as asymmetrical branching events in the lung.
Collapse
Affiliation(s)
- David Warburton
- Developmental Biology Program, Childrens Hospital Los Angeles Research Institute and the Center for Craniofacial Molecular Biology, Keck School of Medicine and School of Dentistry, University of Southern California, USA.
| | | | | | | | | | | | | |
Collapse
|
1696
|
Nicolás FJ, Hill CS. Attenuation of the TGF-beta-Smad signaling pathway in pancreatic tumor cells confers resistance to TGF-beta-induced growth arrest. Oncogene 2003; 22:3698-711. [PMID: 12802277 DOI: 10.1038/sj.onc.1206420] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We have investigated the mechanism whereby tumor cells become resistant to the antiproliferative effects of transforming growth factor (TGF)-beta, while maintaining other responses that can lead to increased malignancy and invasiveness. TGF-beta signaling results in nuclear accumulation of active Smad complexes which regulate transcription of target genes. Here we show that in two pancreatic carcinoma cell lines, PT45 and Panc-1, that are resistant to TGF-beta-induced growth arrest, the TGF-beta-Smad signaling pathway is attenuated compared with epithelial cells that are sensitive to the antiproliferative effects of TGF-beta (HaCaT and Colo-357). In PT45 and Panc-1 cells, active Smad complexes remain nuclear for only 1-2 h compared with more than 6 h in HaCaT and Colo-357 cells. The attenuated pathway in PT45 and Panc-1 cells correlates with low levels of TGF-beta type I receptor and results in an altered expression profile of TGF-beta-inducible genes required for cell cycle arrest. Most significantly, expression of the CDK inhibitor, p21(Cip1/WAF1), which is required for TGF-beta-induced growth arrest in these cells, is not maintained. Moreover, we show that artificially attenuating the TGF-beta-Smad signaling pathway in HaCaT cells is sufficient to prevent TGF-beta-induced growth arrest. Our results demonstrate that the duration of TGF-beta-Smad signaling is a critical determinant of the specificity of the TGF-beta response.
Collapse
Affiliation(s)
- Francisco J Nicolás
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, UK
| | | |
Collapse
|
1697
|
Yamamoto T, Kimura T, Ueta E, Tatemoto Y, Osaki T. Characteristic cytokine generation patterns in cancer cells and infiltrating lymphocytes in oral squamous cell carcinomas and the influence of chemoradiation combined with immunotherapy on these patterns. Oncology 2003; 64:407-15. [PMID: 12759539 DOI: 10.1159/000070300] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Cytokines produced by tumor cells and tumor-infiltrating lymphocytes (TIL) appear to regulate tumor cell growth and the cytotoxic activity of TIL. The objectives of the present study were to investigate cytokine generation patterns in tumor cells and TIL and to examine the influence of cancer therapy on this cytokine production and the cytotoxic activity of TIL. METHODS We determined the levels of cytokines produced by tumor cells and TIL in vitro and measured the cytotoxic activity of TIL against Daudi cells in patients with oral squamous cell carcinoma (OSC) before and 1 week after the start of concomitant chemo-radio-immunotherapy. RESULTS Before the therapy, OSC cells generated higher levels of granulocyte-macrophage colony-stimulating factor, tumor necrosis factor-alpha (TNF-alpha) and transforming growth factor-beta (TGF-beta) than did oral keratinocytes isolated from the noninflamed gingivae of healthy individuals, but both kinds of cells generated similar levels of interleukin (IL)-1beta and IL-6. Compared with peripheral blood mononuclear cells (PBMC) of the patients, TIL produced higher levels of IL-1beta, IL-6, IL-10, TNF-alpha and TGF-beta, whereas their production of IL-12 and interferon-gamma (IFN-gamma) was only slightly higher than that in PBMC. After 1 week of therapy, the cytokine production by OSC cells had largely decreased, while the production of TNF-alpha, IFN-gamma, TGF-beta and IL-12 by TIL had increased greatly, although other cytokine levels were almost constant during the investigations. The cytotoxic activity of TIL was higher than that of PBMC before the therapy, and this activity was strongly increased by 1 week of therapy. CONCLUSIONS These results suggest that the cytokine productivities of TIL and tumor cells differ from those of PBMC and normal keratinocytes, respectively, and that chemo-radio-immunotherapy modulates in situ cytokine generation, which is advantageous for inhibition of tumor cell growth and activation of TIL.
Collapse
Affiliation(s)
- Tetsuya Yamamoto
- Department of Oral Surgery, Kochi Medical School, Nankoku City, Japan.
| | | | | | | | | |
Collapse
|
1698
|
Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003; 3:537-49. [PMID: 12842083 DOI: 10.1016/s1535-6108(03)00132-6] [Citation(s) in RCA: 1915] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We investigated the molecular basis for osteolytic bone metastasis by selecting human breast cancer cell line subpopulations with elevated metastatic activity and functionally validating genes that are overexpressed in these cells. These genes act cooperatively to cause osteolytic metastasis, and most of them encode secreted and cell surface proteins. Two of these genes, interleukin-11 and CTGF, encode osteolytic and angiogenic factors whose expression is further increased by the prometastatic cytokine TGF beta. Overexpression of this bone metastasis gene set is superimposed on a poor-prognosis gene expression signature already present in the parental breast cancer population, suggesting that metastasis requires a set of functions beyond those underlying the emergence of the primary tumor.
Collapse
Affiliation(s)
- Yibin Kang
- Cell Biology Program and Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
1699
|
Affiliation(s)
- Nancy Dumont
- Department of Cancer Biology, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN 37232, USA
| | | |
Collapse
|
1700
|
Jeruss JS, Santiago JY, Woodruff TK. Localization of activin and inhibin subunits, receptors and SMADs in the mouse mammary gland. Mol Cell Endocrinol 2003; 203:185-96. [PMID: 12782414 DOI: 10.1016/s0303-7207(02)00291-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Activin and inhibin, two closely related protein hormones, are members of the transforming growth factor beta (TGF beta) superfamily of growth factors. Activin and TGF beta have been associated with mouse mammary gland development and human breast carcinogenesis. TGF beta expression in the mammary gland has been previously described, and was found to be expressed in nonparous tissue and during pregnancy, down-regulated during lactation, and then up-regulated during involution. The expression pattern of activin subunits, receptors and cytoplasmic signaling molecules has not been thoroughly described in post-natal mammary gland development. We hypothesize that activin signaling components are dynamically regulated during mammary gland development, thereby permitting activin to have distinct temporal growth regulatory actions on this tissue. To examine the activin signal transduction system in the mammary gland, tissue from CD1 female mice was dissected from nonparous, lactating day 1, 10, and 20 and post-weaning day 4 animals. The expression of the activin receptors (ActRIIA, ActRIIB and ActRIB), the inhibin co-receptor (betaglycan), and ligand subunit (alpha, beta A and beta B), mRNA was measured by semi-quantitative RT-PCR in these tissues. In addition, the cellular compartmentalization of the activin signaling proteins, including the cytoplasmic signaling co-activators, Smads 2, 3 and 4, were examined by immunohistochemistry. Generally, mRNA abundance of activin signaling components was greatest in the nonparous tissue, and then decreased, whereas protein immunoreactivity for activin signaling components increased during lactation and decreased during involution. The alpha-subunit protein was detected in nonparous and lactating day 1 tissue only. Importantly, Smad 3, but not Smad 2, was detected in epithelial cell nuclei during all time points examined, indicating that activin signaling is mediated by Smad 3 at these times. These findings suggest that activin's growth regulatory role during lactation may be distinguished from that of TGF beta during post-natal mammary development. Future studies will focus on determining the exact role this ligand plays in mammary tissue differentiation and neoplasia.
Collapse
Affiliation(s)
- Jacqueline S Jeruss
- Department of Neurobiology and Physiology, Northwestern University, O.T. Hogan 4-150, 2153 N. Campus Dr., Evanston, IL 60208, USA
| | | | | |
Collapse
|