Field Of Vision Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Aug 14, 2012; 18(30): 3931-3935
Published online Aug 14, 2012. doi: 10.3748/wjg.v18.i30.3931
S100A4 in esophageal cancer: Is this the one to blame?
Jianyuan Chai, Laboratory of GI Injury and Cancer, VA Long Beach Healthcare System, Long Beach, CA 90822, United States
M Mazen Jamal, Division of Gastroenterology, Department of Medicine, University of California, Irvine, CA 92868, United States
Author contributions: Both authors contributed equally.
Supported by The Department of Veterans Affairs of the United States
Correspondence to: Jianyuan Chai, PhD, Research and Development Office (09-151), Laboratory of GI Injury and Cancer, VA Long Beach Healthcare System, 5901 E. Seventh Street, Long Beach, CA 90822, United States. jianyuan.chai@va.gov
Telephone: +1-562-8268000 Fax: +1-562-8265675
Received: May 31, 2012
Revised: June 15, 2012
Accepted: June 28, 2012
Published online: August 14, 2012

Abstract

Metastasis is the main reason for cancer-related death. S100A4 is one of the key molecules involved in this event. Several studies have shown that overexpression of S100A4 in non-metastatic cancer cells can make them become metastatic, and knockdown of S100A4 in metastatic cancer cells can curtail their invasive nature. A study by Chen et al[2] published in the World J Gastroenterol 18(9): 915-922, 2012 is a typical example. This study showed in vitro and in vivo evidence that S100A4 expression level determines the invasiveness of esophageal squamous carcinoma. Considering the fact that more than half of the cancer-related deaths are caused by malignancies derived from the digestive system and esophageal cancer is the 4th top contributor to this fraction, this study warrants more attention.

Key Words: Esophageal cancer, S100A4, Metastasis



INVITED COMMENTARY ON HOT ARTICLES

Cancer is the second leading cause of death in the world (18%), after heart disease (21%). Among about 18 million new cases of cancers diagnosed each year, about one third is skin cancer. However, 95% of skin cancer is either basal cell carcinoma or squamous cell carcinoma, which has a mortality of less than 0.5%. The majority of cancer-related deaths are actually caused by malignancies derived from the digestive system, including esophagus, stomach, small intestine, colon, rectum, anus, liver, gallbladder and pancreas[1]. The main feature that makes these cancers deadly is metastasis, a process that cancer cells break off from their original location and invade other parts of the organ. The majority of skin cancers do not have this capacity; therefore, they can be easily treated before becoming life threatening. Esophageal cancer, on the other hand, is highly metastatic. Therefore, understanding the molecular mechanisms behind its metastasis is of great values for developing better treatment strategies. A study by Chen et al[2] published in the World J Gastroenterol 18(9): 915-922, 2012 examined the role of S100A4, one of the well-known cancer metastatic markers, in esophageal squamous cell carcinoma (ESCC) in vitro and in vivo, in animal models as well as in clinical human specimens, and clearly demonstrated a reliance of the invasiveness of esophageal cancer on this small calcium-binding protein[2].

A little biography of S100A4: Short but hot

S100A4 was discovered in the mid 1980s by several laboratories independently. One of these laboratories belonged to Daniel Nathans, MD (10/30/1928-11/16/1999) (Figure 1), the Nobel Prize winner in Physiology/Medicine 1978 for his landmark discovery of restriction enzymes. In 1983, one of his post-doctoral fellows, Daniel I Linzer, PhD, was constructing a cDNA library from serum-stimulated mouse 3T3 fibroblasts and found that a clone named 18A2 was highly up-regulated by serum exposure[3]. There seemed to be many laboratories in the late 70s and early 80s of the 20th century which were interested in the effect of serum on gene expression. That was also how and when serum response factor (SRF) was discovered[4,5]. In the following year, Linzer took a job at Northwest University in Illinois (now he is the Provost of this school) and continued his study on 18A2. He determined that 18A2 coded for a calcium binding protein of 101 amino acids[6], much similar to the members of S100 family, a group of small peptides that are known to be 100% soluble in saturated ammonia sulfate. He also compared the sequence of 18A2 with 2A9, a human clone that was published a year earlier[7], and found a 57% nucleotide and 62% amino acid homology between them. It might be due to the difference of species origin, Linzer was pretty sure that these two sequences represented different genes. Around that time and shortly thereafter, several other laboratories also published similar sequences and each of which was given a different name, including p9Ka from rat mammary cells[8], 42A from rat neuronal cells[9], pEL98 from mouse fibroblasts[10], CAPL from Aplysia neurons[11], mts1 from metastatic tumor cells[12], and FSP1 from mouse fibroblasts[13]. Despite the individuality of each of these studies, there were some common features shared among their discoveries: (1) serum inducibility; (2) around 100 amino acids; and (3) similarity to S100 calcium binding proteins. Although all of these sequences eventually turned out to be for a single molecule - S100A4, each of these studies made unique contributions to our knowledge today about S100A4. The last two studies warrant an extra attention, because one established the connection between S100A4 and cancer metastasis and the other associated it to fibroblast phenotype. Now we know that S100A4 is a prognostic marker for metastatic cancers as well as a marker for epithelial-mesenchymal transition. However, both of these studies went a little bit too far by calling this molecule metastatic-specific and fibroblast-specific, respectively. Now we know that is not entirely true, a lot of other cells (e.g., epithelial cells, endothelial cells, lymphocytes, smooth muscle cells, etc.) also express S100A4, just as our study reported[14].

Figure 1
Figure 1 Daniel Nathans, MD, Nobel Laureate (10/30/1928-11/16/1999), Department of Molecular Biology and Genetics, the Johns Hopkins University School of Medicine Baltimore, Baltimore, MD 21205, United States.
Functions of S100A4: Motivation to move

Up to date, S100 family includes 25 members with common characteristics such as low molecular weight, two calcium binding sites of the helix-loop-helix (“EF-hand type”) conformation, and complete solubility in ammonium sulfate at pH 7. They have been implicated in regulation of protein phosphorylation, transcription factor activation, calcium homeostasis, cytoskeleton reorganization, cell migration, cell growth and death[15].

S100A4 is naturally expressed in various cell types including both cancer and normal cells, and its elevation is usually associated with cell motility. It appears that wherever cell migration is required, such as wound healing[16], angiogenesis[17] and cancer metastasis[18], S100A4 is activated. Like other members of S100 family, S100A4 works like a calcium sensor. Upon calcium binding, S100A4 goes through a series of conformational changes, which allow the molecule to interact with its targets, such as nonmuscle myosin heavy chain (MHC IIA) and liprin β1, to facilitate cell migration[19,20]. For this reason, in motile cells, S100A4 is often found in complex with these cytoskeletal components at the migrating front where a high level of calcium is accumulated. It is interesting to know that S100A4 knock-out mice do not display developmental abnormalities in the postnatal period, but 10% of them develop tumors at age of 10-14 months, possibly due to destabilization of the tumor suppressor p53[21], as S100A4 has been shown capable to bind to the C-terminal of p53 and repress its transcriptional activity[22,23].

Yet, the story of S100A4 is not as straightforward as it might have been anticipated. In addition to being a cytoskeletal regulator in the cytoplasm, S100A4 has also been localized to the nucleus and extracellular matrix. How it gets there and what it does in these locations remain unclear. Nevertheless, its association with transcription factors like p53 might explain some of its roles in the nucleus. It has been postulated that S100A4 binding to the tetramerization domain of p53 favors p53 oligomerization and thereby facilitates p53 nuclear translocation[23]. On the other hand, extracellular S100A4 has been demonstrated to stimulate MMP-13 expression in chondrocytes in a receptor for advanced glycation end products (RAGE)-dependent manner[24], while its inductivity on neuron growth was found to be RAGE irrelevant[25]. More complicatedly, S100A4 has been found in association with cell death in a conflict way, it inhibits apoptosis in pancreatic cancer[26] but promotes it in osteosarcoma cells[27].

S100A4 in cancers: A facilitator, not a generator

Elevation of S100A4 has been found in almost every metastatic cancer known, including breast[28], ovarian[29], prostate[30], urinary bladder[31], lung[32], esophageal[33], gastric[34], colon[35], pancreatic[36], liver[37], gallbladder[38] and thyroid carcinomas[39]. More direct evidence for the essential role of S100A4 in cancer metastasis perhaps comes from in vitro studies and animal models, which have shown that overexpression of S100A4 in non-metastatic tumor cells confers a metastatic phenotype, just as demonstrated in the study by Chen et al[2] as well as several others[40,41]; whereas, knockdown of S100A4 in metastatic tumor cells curtails their invasive capability[2,42,43].

It should be pointed out though that S100A4 is not an oncogene product. As shown by transgenic studies[17,44], mice carrying extra copies of S100A4 gene develop normally as wild-type and have no increased risk of cancer. However, when these mice mated with cancer mice, their offspring showed increased number of tumors distant from their primary location[45]. Therefore, S100A4 is not a cancer generator but a metastatic facilitator.

S100A4 has been studied extensively in other cancers, especially in breast cancer. In esophageal cancer, there are about a dozen of publications so far, mostly focusing on squamous cell carcinoma. The earliest study that can be found was done by a Japanese group[33], showing an elevated expression of S100A4 protein in surgically resected ESCC, and a possible association with esophageal cancer progression. However, a later study reported an opposite result, showing that 11 out of 16 S100 family members examined, including S100A4, were down-regulated at transcriptional level in tumor tissues compared with adjacent normal tissues[46]. In 2010, a Chinese research team used RNA interference technology to knock down S100A4 in metastatic esophageal tumor cells and grafted them in nude mice[47]. They noticed that tumor growth was significantly inhibited by S100A4 deficiency, and E-cadherin expression was reciprocal to the level of S100A4. Unfortunately, the study had little impact because it was published in a local journal in Chinese. However, the idea of xenografting has recently advanced to a new cancer treatment strategy - the “avatar” mice. Principally, it is to take tumor tissue from a patient and graft it in nude mice to create a personalized colony of mice carrying exact that patient’s cancer, and then test every potential treatment combinations in mice before selecting the best one to treat that patient. Manuel Hidalgo, the Director of the Spanish National Cancer Research Center in Madrid, has been practicing this approach for pancreatic cancer patients over years and showed a clear advantage in drug responses[48,49], and now more and more researchers believe that this idea holds a great promise in cancer treatment in the future.

In the study by Chen et al[2], the research team cleverly used two ESCC cell lines, EC109 (highly invasive) and TE13 (non-invasive), and successfully made these cells switch characters by down-regulation of S100A4 in EC109 and up-regulation of S100A4 in TE13. They provided in vitro and in vivo evidence that the level of S100A4 determines the metastatic status of the cancer.

There are two main subtypes of esophageal cancer: ESCC and esophageal adenocarcinoma (EAC). Although nearly 95% of esophageal cancer is ESCC, EAC has been rising by 6-fold annually in Americans and now its increase rate exceeds the rate for any other type of cancers. Overexpression of S100A4 was also reported in EAC and its correlation with lymph node metastasis was found significant[50].

Although the exact molecular mechanisms how S100A4 promotes cancer metastasis still need to be further examined, based on various studies, one possible explanation could be that S100A4 binding to liprin β1 inhibits its phosphorylation[19], and thereby prevents its interaction with liprin α1. As a result, liprin α1 fails to recruit leukocyte common antigen-related (LAR) protein[51], a phosphatase, to focal adhesions. Without LAR to dephosphorylate β-catenin[52], β-catenin becomes activated to leave E-cadherin and results in the collapse of adherens junctions, allowing cells to migrate. As found in our study, the dissociation of β-catenin from E-cadherin causes E-cadherin ubiquitination and degradation[53], which might at least in part explains why S100A4 elevation is often found in association of E-cadherin loss, as shown in the study by Chen et al[2].

S100A4 in normal situation: An innocent bystander

As discussed above, S100A4 is expressed wherever cell migration is required, regardless normal or pathological situation. However, most of S100A4 studies focus on its bad side, such as cancer metastasis and organ fibrosis. Its good side has been continually overlooked. If we go back to the story that S100A4 was discovered in an experiment of serum stimulated fibroblasts, we know that S100A4 is innocent. Cells, including fibroblasts, in our body normally do not come into a direct contact with serum unless there is an injury. Therefore, when cells are suddenly exposed to serum, as the experiment done in Nathans’ lab, they naturally interpret it as a signal of a wound. Therefore, a transcriptional program for wound healing gets activated immediately to battle against injury. S100A4 is just one of the players in this battle. So is SRF, and so are many SRF-regulated genes (e.g., C-FOS, EGR-1, CCN1, CTGF, FGF10, etc.)[54]. All these genes contain a common regulatory element CArG box, which SRF recognizes to bind. S100A4 gene also contains such element in its promoter region[55], suggesting a possible regulation by SRF. In vivo, S100A4 activation has been found in various wound healings, and its contributions to tissue repair and modification are indisputable[16,56].

Footnotes

Peer reviewer: Ferruccio Bonino, MD, PhD, Professor of Gastroenterology, Director of Liver and Digestive Disease Division, Department of Internal Medicine, University of Pisa, Director of General Medicine 2 Unit University Hospital of Pisa, Via Roma 67, 56124 Pisa, Italy

S- Editor Cheng JX L- Editor Ma JY E- Editor Xiong L

References
1.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Chen D, Zheng XF, Yang ZY, Liu DX, Zhang GY, Jiao XL, Zhao H. S100A4 silencing blocks invasive ability of esophageal squamous cell carcinoma cells. World J Gastroenterol. 2012;18:915-922.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Linzer DI, Nathans D. Growth-related changes in specific mRNAs of cultured mouse cells. Proc Natl Acad Sci USA. 1983;80:4271-4275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 166]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
4.  Treisman R. Identification of a protein-binding site that mediates transcriptional response of the c-fos gene to serum factors. Cell. 1986;46:567-574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 558]  [Cited by in F6Publishing: 753]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
5.  Treisman R. Journey to the surface of the cell: Fos regulation and the SRE. EMBO J. 1995;14:4905-4913.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Jackson-Grusby LL, Swiergiel J, Linzer DI. A growth-related mRNA in cultured mouse cells encodes a placental calcium binding protein. Nucleic Acids Res. 1987;15:6677-6690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 97]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
7.  Calabretta B, Battini R, Kaczmarek L, de Riel JK, Baserga R. Molecular cloning of the cDNA for a growth factor-inducible gene with strong homology to S-100, a calcium-binding protein. J Biol Chem. 1986;261:12628-12632.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Barraclough R, Savin J, Dube SK, Rudland PS. Molecular cloning and sequence of the gene for p9Ka. A cultured myoepithelial cell protein with strong homology to S-100, a calcium-binding protein. J Mol Biol. 1987;198:13-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 92]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
9.  Masiakowski P, Shooter EM. Nerve growth factor induces the genes for two proteins related to a family of calcium-binding proteins in PC12 cells. Proc Natl Acad Sci USA. 1988;85:1277-1281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 112]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
10.  Goto K, Endo H, Fujiyoshi T. Cloning of the sequences expressed abundantly in established cell lines: identification of a cDNA clone highly homologous to S-100, a calcium binding protein. J Biochem. 1988;103:48-53.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Beushausen S, Bergold P, Sturner S, Elste A, Roytenberg V, Schwartz JH, Bayley H. Two catalytic subunits of cAMP-dependent protein kinase generated by alternative RNA splicing are expressed in Aplysia neurons. Neuron. 1988;1:853-864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 41]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
12.  Ebralidze A, Tulchinsky E, Grigorian M, Afanasyeva A, Senin V, Revazova E, Lukanidin E. Isolation and characterization of a gene specifically expressed in different metastatic cells and whose deduced gene product has a high degree of homology to a Ca2+-binding protein family. Genes Dev. 1989;3:1086-1093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 238]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
13.  Strutz F, Okada H, Lo CW, Danoff T, Carone RL, Tomaszewski JE, Neilson EG. Identification and characterization of a fibroblast marker: FSP1. J Cell Biol. 1995;130:393-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 786]  [Cited by in F6Publishing: 851]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
14.  Chai JY, Modak C, Mouazzen W, Narvaez R, Pham J. Epithelial or mesenchymal: Where to draw the line? Biosci Trends. 2010;4:130-142.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Lukanidin E, Sleeman JP. Building the niche: the role of the S100 proteins in metastatic growth. Semin Cancer Biol. 2012;22:216-225.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Schneider M, Kostin S, Strøm CC, Aplin M, Lyngbaek S, Theilade J, Grigorian M, Andersen CB, Lukanidin E, Lerche Hansen J. S100A4 is upregulated in injured myocardium and promotes growth and survival of cardiac myocytes. Cardiovasc Res. 2007;75:40-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
17.  Ambartsumian N, Klingelhöfer J, Grigorian M, Christensen C, Kriajevska M, Tulchinsky E, Georgiev G, Berezin V, Bock E, Rygaard J. The metastasis-associated Mts1(S100A4) protein could act as an angiogenic factor. Oncogene. 2001;20:4685-4695.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Boye K, Maelandsmo GM. S100A4 and metastasis: a small actor playing many roles. Am J Pathol. 2010;176:528-535.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Kriajevska M, Fischer-Larsen M, Moertz E, Vorm O, Tulchinsky E, Grigorian M, Ambartsumian N, Lukanidin E. Liprin beta 1, a member of the family of LAR transmembrane tyrosine phosphatase-interacting proteins, is a new target for the metastasis-associated protein S100A4 (Mts1). J Biol Chem. 2002;277:5229-5235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 103]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
20.  Badyal SK, Basran J, Bhanji N, Kim JH, Chavda AP, Jung HS, Craig R, Elliott PR, Irvine AF, Barsukov IL. Mechanism of the Ca²+-dependent interaction between S100A4 and tail fragments of nonmuscle myosin heavy chain IIA. J Mol Biol. 2011;405:1004-1026.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  EL Naaman C, Grum-Schwensen B, Mansouri A, Grigorian M, Santoni-Rugiu E, Hansen T, Kriajevska M, Schafer BW, Heizmann CW, Lukanidin E. Cancer predisposition in mice deficient for the metastasis-associated Mts1(S100A4) gene. Oncogene. 2004;23:3670-3680.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Grigorian M, Andresen S, Tulchinsky E, Kriajevska M, Carlberg C, Kruse C, Cohn M, Ambartsumian N, Christensen A, Selivanova G. Tumor suppressor p53 protein is a new target for the metastasis-associated Mts1/S100A4 protein: functional consequences of their interaction. J Biol Chem. 2001;276:22699-22708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 238]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
23.  Fernandez-Fernandez MR, Veprintsev DB, Fersht AR. Proteins of the S100 family regulate the oligomerization of p53 tumor suppressor. Proc Natl Acad Sci USA. 2005;102:4735-4740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 145]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
24.  Yammani RR, Carlson CS, Bresnick AR, Loeser RF. Increase in production of matrix metalloproteinase 13 by human articular chondrocytes due to stimulation with S100A4: Role of the receptor for advanced glycation end products. Arthritis Rheum. 2006;54:2901-2911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 161]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
25.  Kiryushko D, Novitskaya V, Soroka V, Klingelhofer J, Lukanidin E, Berezin V, Bock E. Molecular mechanisms of Ca(2+) signaling in neurons induced by the S100A4 protein. Mol Cell Biol. 2006;26:3625-3638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
26.  Mahon PC, Baril P, Bhakta V, Chelala C, Caulee K, Harada T, Lemoine NR. S100A4 contributes to the suppression of BNIP3 expression, chemoresistance, and inhibition of apoptosis in pancreatic cancer. Cancer Res. 2007;67:6786-6795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
27.  Pedersen KB, Andersen K, Fodstad Ø, Maelandsmo GM. Sensitization of interferon-gamma induced apoptosis in human osteosarcoma cells by extracellular S100A4. BMC Cancer. 2004;4:52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
28.  Rudland PS, Platt-Higgins A, Renshaw C, West CR, Winstanley JH, Robertson L, Barraclough R. Prognostic significance of the metastasis-inducing protein S100A4 (p9Ka) in human breast cancer. Cancer Res. 2000;60:1595-1603.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kikuchi N, Horiuchi A, Osada R, Imai T, Wang C, Chen X, Konishi I. Nuclear expression of S100A4 is associated with aggressive behavior of epithelial ovarian carcinoma: an important autocrine/paracrine factor in tumor progression. Cancer Sci. 2006;97:1061-1069.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Saleem M, Kweon MH, Johnson JJ, Adhami VM, Elcheva I, Khan N, Bin Hafeez B, Bhat KM, Sarfaraz S, Reagan-Shaw S. S100A4 accelerates tumorigenesis and invasion of human prostate cancer through the transcriptional regulation of matrix metalloproteinase 9. Proc Natl Acad Sci USA. 2006;103:14825-14830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 153]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
31.  Davies BR, O'Donnell M, Durkan GC, Rudland PS, Barraclough R, Neal DE, Mellon JK. Expression of S100A4 protein is associated with metastasis and reduced survival in human bladder cancer. J Pathol. 2002;196:292-299.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Grum-Schwensen B, Klingelhöfer J, Grigorian M, Almholt K, Nielsen BS, Lukanidin E, Ambartsumian N. Lung metastasis fails in MMTV-PyMT oncomice lacking S100A4 due to a T-cell deficiency in primary tumors. Cancer Res. 2010;70:936-947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
33.  Ninomiya I, Ohta T, Fushida S, Endo Y, Hashimoto T, Yagi M, Fujimura T, Nishimura G, Tani T, Shimizu K. Increased expression of S100A4 and its prognostic significance in esophageal squamous cell carcinoma. Int J Oncol. 2001;18:715-720.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Cho YG, Nam SW, Kim TY, Kim YS, Kim CJ, Park JY, Lee JH, Kim HS, Lee JW, Park CH. Overexpression of S100A4 is closely related to the aggressiveness of gastric cancer. APMIS. 2003;111:539-545.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Takenaga K, Nakanishi H, Wada K, Suzuki M, Matsuzaki O, Matsuura A, Endo H. Increased expression of S100A4, a metastasis-associated gene, in human colorectal adenocarcinomas. Clin Cancer Res. 1997;3:2309-2316.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Ai KX, Lu LY, Huang XY, Chen W, Zhang HZ. Prognostic significance of S100A4 and vascular endothelial growth factor expression in pancreatic cancer. World J Gastroenterol. 2008;14:1931-1935.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Komatsu K, Murata K, Kameyama M, Ayaki M, Mukai M, Ishiguro S, Miyoshi J, Tatsuta M, Inoue M, Nakamura H. Expression of S100A6 and S100A4 in matched samples of human colorectal mucosa, primary colorectal adenocarcinomas and liver metastases. Oncology. 2002;63:192-200.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Nakamura T, Ajiki T, Murao S, Kamigaki T, Maeda S, Ku Y, Kuroda Y. Prognostic significance of S100A4 expression in gallbladder cancer. Int J Oncol. 2002;20:937-941.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Zou M, Famulski KS, Parhar RS, Baitei E, Al-Mohanna FA, Farid NR, Shi Y. Microarray analysis of metastasis-associated gene expression profiling in a murine model of thyroid carcinoma pulmonary metastasis: identification of S100A4 (Mts1) gene overexpression as a poor prognostic marker for thyroid carcinoma. J Clin Endocrinol Metab. 2004;89:6146-6154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 41]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
40.  Davies BR, Davies MP, Gibbs FE, Barraclough R, Rudland PS. Induction of the metastatic phenotype by transfection of a benign rat mammary epithelial cell line with the gene for p9Ka, a rat calcium-binding protein, but not with the oncogene EJ-ras-1. Oncogene. 1993;8:999-1008.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Grigorian M, Ambartsumian N, Lykkesfeldt AE, Bastholm L, Elling F, Georgiev G, Lukanidin E. Effect of mts1 (S100A4) expression on the progression of human breast cancer cells. Int J Cancer. 1996;67:831-841.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Maelandsmo GM, Hovig E, Skrede M, Engebraaten O, Flørenes VA, Myklebost O, Grigorian M, Lukanidin E, Scanlon KJ, Fodstad O. Reversal of the in vivo metastatic phenotype of human tumor cells by an anti-CAPL (mts1) ribozyme. Cancer Res. 1996;56:5490-5498.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Takenaga K, Nakamura Y, Sakiyama S. Expression of antisense RNA to S100A4 gene encoding an S100-related calcium-binding protein suppresses metastatic potential of high-metastatic Lewis lung carcinoma cells. Oncogene. 1997;14:331-337.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Davies MP, Rudland PS, Robertson L, Parry EW, Jolicoeur P, Barraclough R. Expression of the calcium-binding protein S100A4 (p9Ka) in MMTV-neu transgenic mice induces metastasis of mammary tumours. Oncogene. 1996;13:1631-1637.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Xue C, Plieth D, Venkov C, Xu C, Neilson EG. The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. Cancer Res. 2003;63:3386-3394.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Ji J, Zhao L, Wang X, Zhou C, Ding F, Su L, Zhang C, Mao X, Wu M, Liu Z. Differential expression of S100 gene family in human esophageal squamous cell carcinoma. J Cancer Res Clin Oncol. 2004;130:480-486.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Zhang HY, Zheng XZ, Xuan XY, Wang XH, Wang F, Li SS. [Inhibitory effect and molecular mechanism of silencing S100A4 gene on the growth of transplanted tumor of human esophageal carcinoma EC-1 cells in nude mice]. Sichuan Da Xue Xue Bao Yi Xue Ban. 2010;41:755-759.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Jimeno A, Feldmann G, Suárez-Gauthier A, Rasheed Z, Solomon A, Zou GM, Rubio-Viqueira B, García-García E, López-Ríos F, Matsui W. A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Mol Cancer Ther. 2009;8:310-314.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Morelli MP, Calvo E, Ordoñez E, Wick MJ, Viqueira BR, Lopez-Casas PP, Bruckheimer E, Calles-Blanco A, Sidransky D, Hidalgo M. Prioritizing phase I treatment options through preclinical testing on personalized tumorgraft. J Clin Oncol. 2012;30:e45-e48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
50.  Lee OJ, Hong SM, Razvi MH, Peng D, Powell SM, Smoklin M, Moskaluk CA, El-Rifai W. Expression of calcium-binding proteins S100A2 and S100A4 in Barrett's adenocarcinomas. Neoplasia. 2006;8:843-850.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Serra-Pagès C, Kedersha NL, Fazikas L, Medley Q, Debant A, Streuli M. The LAR transmembrane protein tyrosine phosphatase and a coiled-coil LAR-interacting protein co-localize at focal adhesions. EMBO J. 1995;14:2827-2838.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Müller T, Choidas A, Reichmann E, Ullrich A. Phosphorylation and free pool of beta-catenin are regulated by tyrosine kinases and tyrosine phosphatases during epithelial cell migration. J Biol Chem. 1999;274:10173-10183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 231]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
53.  Chai J, Norng M, Modak C, Reavis KM, Mouazzen W, Pham J. CCN1 induces a reversible epithelial-mesenchymal transition in gastric epithelial cells. Lab Invest. 2010;90:1140-1151.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Chai J. Gastric ulcer healing - Role of serum response factor. Peptic Ulcer Disease. Rijeka, Croatia: InTech 2011; 143-164.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Venkov CD, Link AJ, Jennings JL, Plieth D, Inoue T, Nagai K, Xu C, Dimitrova YN, Rauscher FJ, Neilson EG. A proximal activator of transcription in epithelial-mesenchymal transition. J Clin Invest. 2007;117:482-491.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Ryan DG, Taliana L, Sun L, Wei ZG, Masur SK, Lavker RM. Involvement of S100A4 in stromal fibroblasts of the regenerating cornea. Invest Ophthalmol Vis Sci. 2003;44:4255-4262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]