Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Pathophysiol. Mar 22, 2023; 14(2): 12-20
Published online Mar 22, 2023. doi: 10.4291/wjgp.v14.i2.12
Role of T-box transcription factor 3 in gastric cancers
Naoki Asano, Akira Imatani, Akio Takeuchi, Masashi Saito, Xiao-Yi Jin, Waku Hatta, Kaname Uno, Tomoyuki Koike, Atsushi Masamune, Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
ORCID number: Naoki Asano (0000-0003-4452-8459); Akira Imatani (0000-0003-1885-9983); Xiao-Yi Jin (0000-0003-4690-9570); Waku Hatta (0000-0001-9717-0281); Kaname Uno (0000-0002-4739-8795); Tomoyuki Koike (0000-0001-6472-3257); Atsushi Masamune (0000-0001-7184-7282).
Author contributions: Asano N performed the majority of the writing, prepared the figure and table, and acquired funding; Imatani A performed the writing review and the editing, and acquired funding; Takeuchi A, Saito M, and Jin XY performed data acquisition; Hatta W and Uno K performed writing review and editing; Koike T performed writing review and editing, and supervision; Masamune A performed supervision, writing review and editing.
Supported by Grants-in-Aid for Scientific Research from the Japanese Society for the Promotion of Science, No. 18K07928, No. 19K08411, and No. 22K08025.
Conflict-of-interest statement: Dr. Asano reports grants from Japan Society for the Promotion of Science, during the conduct of the study.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Naoki Asano, MD, PhD, Lecturer, Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi Aoba-ku, Sendai 980-8574, Japan. asanon@med.tohoku.ac.jp
Received: December 14, 2022
Peer-review started: December 14, 2022
First decision: February 18, 2023
Revised: February 22, 2023
Accepted: March 10, 2023
Article in press: March 10, 2023
Published online: March 22, 2023
Processing time: 97 Days and 1.3 Hours

Abstract

The expression of T-box transcription factor 3 (TBX3) has been identified in various cancers, including gastric cancers. Its role in breast cancers and melanomas has been intensively studied, and its contribution to the progression of cancers through suppressing senescence and promoting epithelial-mesenchymal transition has been reported. Recent reports on the role of TBX3 in gastric cancers have implied its involvement in gastric carcinogenesis. Considering its pivotal role in the initiation and progression of cancers, TBX3 could be a promising therapeutic target for gastric cancers.

Key Words: Aging, Wnt, β-catenin, Transforming growth factor-β, Stomach, Carcinogenesis

Core Tip: Expression of T-box transcription factor 3 (TBX3) has been reported in a variety of cancers. Preceding reports have shown that TBX3 contributes to the progression of cancers by suppressing cellular senescence and promoting epithelial-mesenchymal transition. Recent reports on the role of TBX3 in gastric cancers have implied its involvement in aging-related gastric carcinogenesis.



INTRODUCTION

The T-box gene family is involved in embryonic development[1] and is conserved among species[2]. Currently, seventeen transcription factors have been identified as members of the T-box gene family in mammals. The T-box gene family consists of five subfamilies, namely, T, Tbx1, Tbx2, Tbx6, and Tbr1 (Table 1).

Table 1 T-box gene family.
T subfamily
Tbx1 subfamily
Tbx2 subfamily
Tbx6 subfamily
Tbr1 subfamily
TTbx1Tbx2Tbx6Tbr1
Tbx19 (Tpit)Tbx10Tbx3MgaTbr2 (Eomes)
Tbx15Tbx4Tbx21 (Tbet)
Tbx18Tbx5
Tbx20
Tbx22

T-box transcription factor 3 (TBX3), which belongs to the Tbx2 subfamily, was initially reported as the gene responsible for ulnar-mammary syndrome, an autosomal dominant human development disorder that affects limb, apocrine gland, tooth, hair, and genital development[3]. Studies with genetically engineered mice revealed that Tbx3 homozygous mutant mice were embryonic lethal and exhibited yolk sac defects, lack of mammary glands, and limb defects[4]. Subsequent studies have revealed the involvement of TBX3 in the development of numerous organs, including the heart[5], retina[6], ureter[7], and inner ear[8].

The TBX3 protein consists of 723 amino acids and is encoded by 2169 bp nucleotides in 7 exons. Differential splicing of the second intron leads to the addition of the 2a exon, resulting in the production of the TBX3+2a isoform. Both TBX3 and TBX3+2a are widely expressed in humans and mice, and alternative splicing of TBX3 was shown to be tissue- and species-specific[9]. TBX3 contains a DNA-binding T-domain[10], two repression domains, and an activation domain[11]. The protein is recruited to the T-box binding sites in the promoter regions of its downstream genes and acts both as a repressor and an activator. The functional similarity between TBX3 and its isoform is still controversial. Fan et al[9] reported that the TBX3+2a isoform lacked the ability to bind to the T-box binding site, and that while TBX3 immortalized mouse embryonic fibroblasts, the TBX3+2a isoform accelerated the senescence in those cells. On the other hand, Hoogaars et al[12] reported that both TBX3 and TBX3+2a were able to bind to the T-box binding site and inhibit cardiac chamber formation in mouse embryonic hearts. Another report from Zhao et al[13] showed that overexpression of either Tbx3 or Tbx3+2a induced the differentiation of mouse embryonic stem cells, but only Tbx3+2a was able to interact directly with Nanog. This discrepancy could be due to the difference in tissues and cells, and future studies are needed to elucidate this issue.

THE FUNCTION AND REGULATION OF TBX3

As expected from its broad expression, TBX3 has important functions. As mentioned earlier, it plays a crucial role in development. TBX3 binds to DNA through its T-domain, and functions as a repressor or an activator owing to its repression domains and an activation domain. Regarding cell cycle-related molecules, Tbx3 has been reported to repress p19ARF (p14ARF in humans) and inhibit cellular senescence[14,15]. This repression was either regulated through direct binding of TBX3 to the p14ARF promoter[15] or through interactions of TBX3 with histone deacetylases (HDAC) 1, 2, 3, and 5[16]. Tbx3 has also been shown to suppress p53[17], while another preceding report demonstrated that it repressed p21CIP1/WAF in a p53-independent manner[18]. In addition, Burgucu et al[19] reported that TBX3 suppressed phosphatase and tensin homolog by repressing its promoter activity, which led to augmented phosphatidylinositol-3-kinase activity. Collectively, these findings indicate that TBX3 possesses the ability to enhance cellular proliferation by regulating these molecules.

Several studies have reported that TBX3 suppresses apoptosis in addition to cellular senescence. Huang et al[20] showed that knocking down TBX3 in hypopharyngeal cancer cells increased annexin V-positive cells and the level of cleaved caspase 3. Ito et al[21] demonstrated that transfection of anti-sense Tbx3 into a rat bladder cancer cell line increased annexin V-positive cells, and the floating cells in the transfected culture exhibited DNA ladders on gel electrophoresis. These two previous studies reported that suppressing TBX3 led to increased apoptosis. On the other hand, Wensing and Campos[22] showed that overexpressing TBX3 and TBX3+2a reduced apoptosis in mesangial cells as assessed by caspase 3 activity. Carlson et al[17] also reported the anti-apoptotic function of TBX3 in overexpression experiments showing that transfection of TBX3 rescued primary mouse embryonic fibroblasts from Myc-induced apoptosis. Taken together, these preceding studies demonstrated that TBX3 possesses an anti-apoptotic function.

Another reported function of TBX3 is the repression of E-cadherin, which contributes to the promotion of epithelial-mesenchymal transition (EMT). Rodriguez et al[23] showed that TBX3 bound to the T-box binding site in the promoter of the E-cadherin gene and repressed E-cadherin expression, which resulted in enhanced invasiveness of melanomas. Dong et al[24] also reported that TBX3 repressed E-cadherin expression in hepatocellular carcinomas (HCC), but the repression occurred through the interaction of TBX3 with HDAC5. Peres et al[25] demonstrated that phosphorylation of TBX3 by AKT serine/threonine kinase 3 (AKT3) stabilized and promoted the nuclear translocation of TBX3, which was essential for E-cadherin repression. Although the effects were exerted through different mechanisms, these reports demonstrated that TBX3 negatively regulates E-cadherin expression, which can promote tumor invasion and metastasis.

For the regulation of TBX3, TBX3 has been regarded as one of the target genes of the Wnt/β-catenin signaling pathway[7,26-28], and a preceding report showed that β-catenin directly bound to the Tcf binding site in the promoter region of Tbx3 and induced Tbx3 expression[29]. However, the Wnt/β-catenin signaling pathway is not the only signaling pathway that regulates Tbx3 expression. Transforming growth factor-β (TGF-β) is another signaling molecule that has been shown to induce Tbx3. Li et al[30] showed that Smad3 and Smad4, downstream signaling molecules of TGF-β, bound to the Smad-binding element in the TBX3 promoter together with JunB and enhanced TBX3 promoter activity. Lee et al[31] also reported that TBX3 was upregulated by TGF-β, although they demonstrated that this induction was dependent on the MAPKK-like protein kinase TOPK. Notch signaling has been shown to facilitate the nuclear translocation of Smad3 and activate TGF-β signaling[32,33], and considering that Notch signaling activates Tbx5[34], another member of the Tbx2 subfamily, it is possible that Notch signaling also regulates Tbx3 expression, but further studies are needed to clarify whether Notch signaling regulates TBX3 expression (Figure 1).

Figure 1
Figure 1 The role of T-box transcription factor 3 in cancers. A schema describing the regulation and function of T-box transcription factor 3 in cancers. P: Phosphorylation; EMT: Epithelial-mesenchymal transition; TGF-β: Transforming growth factor-β; TBX3: T-box transcription factor 3.
TBX3 IN BREAST CANCERS AND MELANOMAS

Overexpression of TBX3 has been reported in various cancers[35]. Among them, breast cancers and melanomas are the cancers in which the role of TBX3 has been intensively studied.

Sequencing of 100 primary breast cancers identified driver mutations in several genes, including TBX3[36], and another comprehensive study of 817 breast tumors identified that mutations in TBX3 were enriched in invasive breast cancers[37]. In addition, genomic sequencing of 1918 breast cancers also indicated that alterations in TBX3 were enriched in breast cancers[38]. These studies demonstrated that TBX3 is one of the key players in breast cancers. Recently, Kostecka et al[39] reported that sequencing of cancer-associated genes, including TBX3, in normal mammary glands of 52 patients with reportedly sporadic breast cancer revealed that subclonal somatic pathogenic variants of these genes were identified at considerable allelic frequencies. This suggests that TBX3 plays an important role in the initiation of breast cancers.

Functionally, TBX3 has been shown to promote the progression of breast cancers by suppressing cellular senescence and enhancing EMT, as described earlier in this review. However, although overexpression of TBX3 alone accelerated mammary epithelial cell proliferation and led to mammary gland hyperplasia, it did not lead to tumor development[40], which implies that overexpression of TBX3 alone is inadequate to initiate breast cancers.

Preceding studies have also reported the overexpression of TBX3 in melanomas[23,25,41], and interestingly, the constitutively active B-RAF mutation observed in melanomas was reported to induce TBX3[42]. Recently, a comprehensive study of 189 cohorts and 178 individual patients identified TBX3 as a marker of poorly differentiated melanomas[43]. Mechanistically, TBX3 was determined to promote tumor progression through inhibition of cellular senescence and promotion of EMT, similar to its role in breast cancers. However, in contrast to its role in breast cancers, the overexpression of TBX3 alone was sufficient to promote the formation and invasion of melanomas[44].

Taken together, the preceding studies suggested that TBX3 promotes tumor progression and invasion by suppressing senescence and enhancing EMT, but whether TBX3 can initiate cancers seems to depend on the type of cancer.

TBX3 IN COLORECTAL CANCERS, PANCREATIC CANCERS, AND LIVER CANCERS

The involvement of TBX3 has also been reported in colorectal cancers. A genome-wide meta-analysis revealed the association of polymorphisms in TBX3 with increased colorectal tumor risk[45]. Shan et al[46] reported that aberrant TBX3 expression was associated with a large tumor size, poor differentiation, invasion, lymph node metastasis, and advanced TNM stage in colorectal cancers, resulting in poor prognosis. They also showed through multivariate analysis that TBX3 can independently predict the outcome of colorectal cancer patients.

Similar to the findings in colorectal cancers, augmented TBX3 expression was associated with poor prognosis in pancreatic cancer patients and was reported to be an independent prognostic factor for overall survival[47]. Regarding the mechanism of TBX3, Perkhofer et al[48] demonstrated that TBX3 enhanced migration, invasion, and angiogenesis in pancreatic cancers through in vitro and in vivo studies.

TBX3 has also been reported to be associated with histological grade, tumor size, metastasis, and Ki-67 expression in HCC[49] and the expression of TBX3 in HCC was found to be induced by Wnt/β-catenin signaling[26,29,50]. Interestingly, Tbx3 in the hepatic microenvironment has been reported to play a crucial role in determining the fate of transformed hepatic cells and whether they develop HCC or intrahepatic cholangiocarcinoma[51].

Collectively, these studies demonstrated that TBX3 plays a major role in these cancers.

TBX3 IN GASTRIC CANCERS

Concerning gastric cancers, Miao et al[52] reported that TBX3 was overexpressed in 46 of 98 primary gastric cancer tissues, and its overexpression correlated with advanced TNM stage and with a higher relapse incidence. In vitro studies demonstrated that overexpression of TBX3 augmented cellular proliferation, whereas knockdown of TBX3 suppressed proliferation in gastric cancer cell lines. Regarding the mechanism involved in TBX3-induced accelerated proliferation, they showed that TBX3 overexpression led to a reduction in the percentage of cells in G1 phase and an increase in the percentage of cells in S and G2 phases in addition to augmented c-Myc and cyclin D1 expression, suggesting that TBX3 facilitated cell cycle progression. The in vitro studies also indicated that TBX3 downregulated E-cadherin and induced N-cadherin and vimentin expression, which suggested the enhancement of EMT. This enhancement of proliferation and EMT could be the reason why the expression of TBX3 is associated with advanced tumor stage in gastric cancers, similar to its correlation with poor prognosis in colorectal cancer patients[46].

Takeuchi et al[53] recently reported the essential role of Tbx3 in aging-related gastric carcinogenesis. Analysis of gastric organoids established from young and aged mice revealed that cellular proliferation was enhanced in aged gastric organoids due to Tbx3-induced repression of cellular senescence. Aged gastric organoids exhibited suppressed expression of Dickkopf3 (Dkk3), a Wnt antagonist, due to methylation of the Dkk3 gene, and consequently, the enhanced Wnt/β-catenin signaling induced Tbx3 expression. Epigenetic alterations, such as the methylation of the Dkk3 gene, are considered as one of the hallmarks of aging[54]. The stochastic process that involves alterations of the methylation state over time is referred to as epigenetic drift and is considered to track biological tissue aging[55]. Indeed, Takeuchi et al[53] showed that DKK3 expression in human gastric tissues decreased as the patient aged, whereas TBX3 expression in human gastric tissues exhibited a positive correlation with patient age. Furthermore, they showed that gastric cancer tissues exhibited lower DKK3 expression and higher TBX3 expression than normal oxyntic glands, suggesting the central role of TBX3 in aging-related gastric carcinogenesis.

Another study of gastric precancerous lesions in 449 patients identified TBX3, along with CDX2 and MYC, as one of the top 7 core genes that contributed to the progression from low-grade intraepithelial neoplasia to high-grade intraepithelial neoplasia[56], a finding that emphasizes the involvement of TBX3 in the early stage of gastric carcinogenesis.

Taken together, these studies imply that TBX3 plays a pivotal role in aging-related carcinogenesis and the progression of gastric cancers. Further studies are awaited to confirm the role of TBX3 in aging-related gastric carcinogenesis.

TBX3 AS A THERAPEUTIC TARGET

Since TBX3 is expressed in various cancers and possesses the ability to promote the progression of these tumors, it has been considered a therapeutic target in these cancers[57]. As TBX3 has been shown to promote cancer progression, its suppression will be required for therapies. Several microRNAs (miR) have been reported to inhibit TBX3. In adipocytes, miR-93 has been shown to inhibit TBX3 and negatively control adipogenesis[58]. On the other hand, miR-137 was reported to inhibit TBX3 in breast cancers[59] and melanomas[60]. In pancreatic cancers, members of the miR-17-92 cluster have been shown to inhibit TBX3 together with p21 and p57[61]. Furthermore, miR-183 was found to suppress TBX3 and enhanced sensitivity to chemotherapy in laryngeal cancers[62]. These miRNAs could be considered candidates for the treatment of TBX3-expressing cancers.

Concerning chemical reagents, an integrated computational approach indicated that two alkaloids, Jervine and Diflomotecan, can form stable complexes with TBX3 and suggested them as new effective drugs against breast cancers[63]. In another study, an aqueous extract of Fructus ligustri lucidi, a common Chinese herbal medicine, was reported to suppress TBX3 and enhance sensitivity to doxorubicin in colon cancer cells[64].

Recently, Willmer et al[65] reported that the multifunctional phosphoprotein nucleolin is required for TBX3 to function and that the nucleolin-targeting aptamer AS1411 exhibited an anticancer effect against sarcomas. These reagents could contribute to anticancer therapy against TBX3-overexpressing cancers.

In addition to its role as a therapeutic target, TBX3 can also contribute to treatment by aiding in the selection of medication for chemotherapy. Freeman et al[43] proposed using TBX3 to predict the outcomes of immune checkpoint inhibitors against melanomas. They showed that patient stratification into risk groups regarding TBX3 and MAP4K1 expression was associated with overall survival; hence, evaluating the expression of these genes could enable individualized treatment for each patient. Similar findings may be found in other TBX3-expressing cancers, and further studies are warranted.

CONCLUSION

In this review, we discussed the role of TBX3 in cancers. TBX3 is expressed in various cancers and contributes to their progression mainly through the repression of senescence and the promotion of EMT. Given its crucial role in tumor progression, TBX3 could be a promising therapeutic target in malignant tumors, including gastric cancers.

ACKNOWLEDGEMENTS

The authors would like to thank the Biomedical Research Unit of Tohoku University Hospital for their technical support.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author's Membership in Professional Societies: The Japanese Society of Gastroenterology, No. 040658; American Gastroenterological Association, No. 307481.

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Japan

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Ji G, China; Zhang H, China S-Editor: Fan JR L-Editor: A P-Editor: Fan JR

References
1.  Naiche LA, Harrelson Z, Kelly RG, Papaioannou VE. T-box genes in vertebrate development. Annu Rev Genet. 2005;39:219-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 324]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
2.  Papaioannou VE. T-box genes in development: from hydra to humans. Int Rev Cytol. 2001;207:1-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 158]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
3.  Bamshad M, Lin RC, Law DJ, Watkins WC, Krakowiak PA, Moore ME, Franceschini P, Lala R, Holmes LB, Gebuhr TC, Bruneau BG, Schinzel A, Seidman JG, Seidman CE, Jorde LB. Mutations in human TBX3 alter limb, apocrine and genital development in ulnar-mammary syndrome. Nat Genet. 1997;16:311-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 395]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
4.  Davenport TG, Jerome-Majewska LA, Papaioannou VE. Mammary gland, limb and yolk sac defects in mice lacking Tbx3, the gene mutated in human ulnar mammary syndrome. Development. 2003;130:2263-2273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 223]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
5.  Singh R, Hoogaars WM, Barnett P, Grieskamp T, Rana MS, Buermans H, Farin HF, Petry M, Heallen T, Martin JF, Moorman AF, 't Hoen PA, Kispert A, Christoffels VM. Tbx2 and Tbx3 induce atrioventricular myocardial development and endocardial cushion formation. Cell Mol Life Sci. 2012;69:1377-1389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 96]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
6.  Motahari Z, Martinez-De Luna RI, Viczian AS, Zuber ME. Tbx3 represses bmp4 expression and, with Pax6, is required and sufficient for retina formation. Development. 2016;143:3560-3572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Aydoğdu N, Rudat C, Trowe MO, Kaiser M, Lüdtke TH, Taketo MM, Christoffels VM, Moon A, Kispert A. TBX2 and TBX3 act downstream of canonical WNT signaling in patterning and differentiation of the mouse ureteric mesenchyme. Development. 2018;145:dev171827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
8.  Kaiser M, Wojahn I, Rudat C, Lüdtke TH, Christoffels VM, Moon A, Kispert A, Trowe MO. Regulation of otocyst patterning by Tbx2 and Tbx3 is required for inner ear morphogenesis in the mouse. Development. 2021;148:dev195651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
9.  Fan W, Huang X, Chen C, Gray J, Huang T. TBX3 and its isoform TBX3+2a are functionally distinctive in inhibition of senescence and are overexpressed in a subset of breast cancer cell lines. Cancer Res. 2004;64:5132-5139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 115]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
10.  Müller CW, Herrmann BG. Crystallographic structure of the T domain-DNA complex of the Brachyury transcription factor. Nature. 1997;389:884-888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 272]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
11.  Carlson H, Ota S, Campbell CE, Hurlin PJ. A dominant repression domain in Tbx3 mediates transcriptional repression and cell immortalization: relevance to mutations in Tbx3 that cause ulnar-mammary syndrome. Hum Mol Genet. 2001;10:2403-2413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 88]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
12.  Hoogaars WM, Barnett P, Rodriguez M, Clout DE, Moorman AF, Goding CR, Christoffels VM. TBX3 and its splice variant TBX3 + exon 2a are functionally similar. Pigment Cell Melanoma Res. 2008;21:379-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
13.  Zhao D, Wu Y, Chen K. Tbx3 isoforms are involved in pluripotency maintaining through distinct regulation of Nanog transcriptional activity. Biochem Biophys Res Commun. 2014;444:411-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
14.  Brummelkamp TR, Kortlever RM, Lingbeek M, Trettel F, MacDonald ME, van Lohuizen M, Bernards R. TBX-3, the gene mutated in Ulnar-Mammary Syndrome, is a negative regulator of p19ARF and inhibits senescence. J Biol Chem. 2002;277:6567-6572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 121]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
15.  Lingbeek ME, Jacobs JJ, van Lohuizen M. The T-box repressors TBX2 and TBX3 specifically regulate the tumor suppressor gene p14ARF via a variant T-site in the initiator. J Biol Chem. 2002;277:26120-26127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 134]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
16.  Yarosh W, Barrientos T, Esmailpour T, Lin L, Carpenter PM, Osann K, Anton-Culver H, Huang T. TBX3 is overexpressed in breast cancer and represses p14 ARF by interacting with histone deacetylases. Cancer Res. 2008;68:693-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 82]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
17.  Carlson H, Ota S, Song Y, Chen Y, Hurlin PJ. Tbx3 impinges on the p53 pathway to suppress apoptosis, facilitate cell transformation and block myogenic differentiation. Oncogene. 2002;21:3827-3835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 78]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
18.  Platonova N, Scotti M, Babich P, Bertoli G, Mento E, Meneghini V, Egeo A, Zucchi I, Merlo GR. TBX3, the gene mutated in ulnar-mammary syndrome, promotes growth of mammary epithelial cells via repression of p19ARF, independently of p53. Cell Tissue Res. 2007;328:301-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
19.  Burgucu D, Guney K, Sahinturk D, Ozbudak IH, Ozel D, Ozbilim G, Yavuzer U. Tbx3 represses PTEN and is over-expressed in head and neck squamous cell carcinoma. BMC Cancer. 2012;12:481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
20.  Huang Y, Zhu H, Ji X, Chen Y, Zhang Y, Huang R, Xie J, Dong P. TBX3 knockdown suppresses the proliferation of hypopharyngeal carcinoma FaDu cells by inducing G1/S cell cycle arrest and apoptosis. Oncol Lett. 2020;19:113-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
21.  Ito A, Asamoto M, Hokaiwado N, Takahashi S, Shirai T. Tbx3 expression is related to apoptosis and cell proliferation in rat bladder both hyperplastic epithelial cells and carcinoma cells. Cancer Lett. 2005;219:105-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
22.  Wensing LA, Campos AH. TBX3, a downstream target of TGF-β1, inhibits mesangial cell apoptosis. Exp Cell Res. 2014;328:340-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
23.  Rodriguez M, Aladowicz E, Lanfrancone L, Goding CR. Tbx3 represses E-cadherin expression and enhances melanoma invasiveness. Cancer Res. 2008;68:7872-7881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 120]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
24.  Dong L, Lyu X, Faleti OD, He ML. The special stemness functions of Tbx3 in stem cells and cancer development. Semin Cancer Biol. 2019;57:105-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
25.  Peres J, Mowla S, Prince S. The T-box transcription factor, TBX3, is a key substrate of AKT3 in melanomagenesis. Oncotarget. 2015;6:1821-1833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
26.  Liang B, Zhou Y, Qian M, Xu M, Wang J, Zhang Y, Song X, Wang H, Lin S, Ren C, Monga SP, Wang B, Evert M, Chen Y, Chen X, Huang Z, Calvisi DF. TBX3 functions as a tumor suppressor downstream of activated CTNNB1 mutants during hepatocarcinogenesis. J Hepatol. 2021;75:120-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
27.  Jin Y, Anbarchian T, Wu P, Sarkar A, Fish M, Peng WC, Nusse R. Wnt signaling regulates hepatocyte cell division by a transcriptional repressor cascade. Proc Natl Acad Sci U S A. 2022;119:e2203849119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
28.  Asano N, Takeuchi A, Imatani A, Saito M, Jin X, Hatta W, Uno K, Koike T, Masamune A. Wnt Signaling and Aging of the Gastrointestinal Tract. Int J Mol Sci. 2022;23:12210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
29.  Renard CA, Labalette C, Armengol C, Cougot D, Wei Y, Cairo S, Pineau P, Neuveut C, de Reyniès A, Dejean A, Perret C, Buendia MA. Tbx3 is a downstream target of the Wnt/beta-catenin pathway and a critical mediator of beta-catenin survival functions in liver cancer. Cancer Res. 2007;67:901-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 124]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
30.  Li J, Weinberg MS, Zerbini L, Prince S. The oncogenic TBX3 is a downstream target and mediator of the TGF-β1 signaling pathway. Mol Biol Cell. 2013;24:3569-3576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
31.  Lee YJ, Park JH, Oh SM. TOPK promotes epithelial-mesenchymal transition and invasion of breast cancer cells through upregulation of TBX3 in TGF-β1/Smad signaling. Biochem Biophys Res Commun. 2020;522:270-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
32.  Asano N, Watanabe T, Kitani A, Fuss IJ, Strober W. Notch1 signaling and regulatory T cell function. J Immunol. 2008;180:2796-2804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 75]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
33.  Luo K. Signaling Cross Talk between TGF-β/Smad and Other Signaling Pathways. Cold Spring Harb Perspect Biol. 2017;9:a022137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 376]  [Cited by in F6Publishing: 363]  [Article Influence: 51.9]  [Reference Citation Analysis (0)]
34.  Rentschler S, Yen AH, Lu J, Petrenko NB, Lu MM, Manderfield LJ, Patel VV, Fishman GI, Epstein JA. Myocardial Notch signaling reprograms cardiomyocytes to a conduction-like phenotype. Circulation. 2012;126:1058-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
35.  Wansleben S, Peres J, Hare S, Goding CR, Prince S. T-box transcription factors in cancer biology. Biochim Biophys Acta. 2014;1846:380-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
36.  Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, Easton D, Langerød A; Oslo Breast Cancer Consortium (OSBREAC), Lee MT, Shen CY, Tee BT, Huimin BW, Broeks A, Vargas AC, Turashvili G, Martens J, Fatima A, Miron P, Chin SF, Thomas G, Boyault S, Mariani O, Lakhani SR, van de Vijver M, van 't Veer L, Foekens J, Desmedt C, Sotiriou C, Tutt A, Caldas C, Reis-Filho JS, Aparicio SA, Salomon AV, Børresen-Dale AL, Richardson AL, Campbell PJ, Futreal PA, Stratton MR. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486:400-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1340]  [Cited by in F6Publishing: 1280]  [Article Influence: 106.7]  [Reference Citation Analysis (0)]
37.  Ciriello G, Gatza ML, Beck AH, Wilkerson MD, Rhie SK, Pastore A, Zhang H, McLellan M, Yau C, Kandoth C, Bowlby R, Shen H, Hayat S, Fieldhouse R, Lester SC, Tse GM, Factor RE, Collins LC, Allison KH, Chen YY, Jensen K, Johnson NB, Oesterreich S, Mills GB, Cherniack AD, Robertson G, Benz C, Sander C, Laird PW, Hoadley KA, King TA; TCGA Research Network, Perou CM. Comprehensive Molecular Portraits of Invasive Lobular Breast Cancer. Cell. 2015;163:506-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1083]  [Cited by in F6Publishing: 1282]  [Article Influence: 160.3]  [Reference Citation Analysis (0)]
38.  Razavi P, Chang MT, Xu G, Bandlamudi C, Ross DS, Vasan N, Cai Y, Bielski CM, Donoghue MTA, Jonsson P, Penson A, Shen R, Pareja F, Kundra R, Middha S, Cheng ML, Zehir A, Kandoth C, Patel R, Huberman K, Smyth LM, Jhaveri K, Modi S, Traina TA, Dang C, Zhang W, Weigelt B, Li BT, Ladanyi M, Hyman DM, Schultz N, Robson ME, Hudis C, Brogi E, Viale A, Norton L, Dickler MN, Berger MF, Iacobuzio-Donahue CA, Chandarlapaty S, Scaltriti M, Reis-Filho JS, Solit DB, Taylor BS, Baselga J. The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell. 2018;34:427-438.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 590]  [Article Influence: 98.3]  [Reference Citation Analysis (0)]
39.  Kostecka A, Nowikiewicz T, Olszewski P, Koczkowska M, Horbacz M, Heinzl M, Andreou M, Salazar R, Mair T, Madanecki P, Gucwa M, Davies H, Skokowski J, Buckley PG, Pęksa R, Śrutek E, Szylberg Ł, Hartman J, Jankowski M, Zegarski W, Tiemann-Boege I, Dumanski JP, Piotrowski A. High prevalence of somatic PIK3CA and TP53 pathogenic variants in the normal mammary gland tissue of sporadic breast cancer patients revealed by duplex sequencing. NPJ Breast Cancer. 2022;8:76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Reference Citation Analysis (0)]
40.  Liu J, Esmailpour T, Shang X, Gulsen G, Liu A, Huang T. TBX3 over-expression causes mammary gland hyperplasia and increases mammary stem-like cells in an inducible transgenic mouse model. BMC Dev Biol. 2011;11:65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
41.  Peres J, Damerell V, Chauhan J, Popovic A, Desprez PY, Galibert MD, Goding CR, Prince S. TBX3 Promotes Melanoma Migration by Transcriptional Activation of ID1, which Prevents Activation of E-Cadherin by MITF. J Invest Dermatol. 2021;141:2250-2260.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
42.  Boyd SC, Mijatov B, Pupo GM, Tran SL, Gowrishankar K, Shaw HM, Goding CR, Scolyer RA, Mann GJ, Kefford RF, Rizos H, Becker TM. Oncogenic B-RAF(V600E) signaling induces the T-Box3 transcriptional repressor to repress E-cadherin and enhance melanoma cell invasion. J Invest Dermatol. 2013;133:1269-1277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
43.  Freeman SS, Sade-Feldman M, Kim J, Stewart C, Gonye ALK, Ravi A, Arniella MB, Gushterova I, LaSalle TJ, Blaum EM, Yizhak K, Frederick DT, Sharova T, Leshchiner I, Elagina L, Spiro OG, Livitz D, Rosebrock D, Aguet F, Carrot-Zhang J, Ha G, Lin Z, Chen JH, Barzily-Rokni M, Hammond MR, Vitzthum von Eckstaedt HC, Blackmon SM, Jiao YJ, Gabriel S, Lawrence DP, Duncan LM, Stemmer-Rachamimov AO, Wargo JA, Flaherty KT, Sullivan RJ, Boland GM, Meyerson M, Getz G, Hacohen N. Combined tumor and immune signals from genomes or transcriptomes predict outcomes of checkpoint inhibition in melanoma. Cell Rep Med. 2022;3:100500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
44.  Peres J, Prince S. The T-box transcription factor, TBX3, is sufficient to promote melanoma formation and invasion. Mol Cancer. 2013;12:117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
45.  Peters U, Jiao S, Schumacher FR, Hutter CM, Aragaki AK, Baron JA, Berndt SI, Bézieau S, Brenner H, Butterbach K, Caan BJ, Campbell PT, Carlson CS, Casey G, Chan AT, Chang-Claude J, Chanock SJ, Chen LS, Coetzee GA, Coetzee SG, Conti DV, Curtis KR, Duggan D, Edwards T, Fuchs CS, Gallinger S, Giovannucci EL, Gogarten SM, Gruber SB, Haile RW, Harrison TA, Hayes RB, Henderson BE, Hoffmeister M, Hopper JL, Hudson TJ, Hunter DJ, Jackson RD, Jee SH, Jenkins MA, Jia WH, Kolonel LN, Kooperberg C, Küry S, Lacroix AZ, Laurie CC, Laurie CA, Le Marchand L, Lemire M, Levine D, Lindor NM, Liu Y, Ma J, Makar KW, Matsuo K, Newcomb PA, Potter JD, Prentice RL, Qu C, Rohan T, Rosse SA, Schoen RE, Seminara D, Shrubsole M, Shu XO, Slattery ML, Taverna D, Thibodeau SN, Ulrich CM, White E, Xiang Y, Zanke BW, Zeng YX, Zhang B, Zheng W, Hsu L; Colon Cancer Family Registry and the Genetics and Epidemiology of Colorectal Cancer Consortium. Identification of Genetic Susceptibility Loci for Colorectal Tumors in a Genome-Wide Meta-analysis. Gastroenterology. 2013;144:799-807.e24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 265]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
46.  Shan ZZ, Yan XB, Yan LL, Tian Y, Meng QC, Qiu WW, Zhang Z, Jin ZM. Overexpression of Tbx3 is correlated with Epithelial-Mesenchymal Transition phenotype and predicts poor prognosis of colorectal cancer. Am J Cancer Res. 2015;5:344-353.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Wang HC, Meng QC, Shan ZZ, Yuan Z, Huang XY. Overexpression of Tbx3 predicts poor prognosis of patients with resectable pancreatic carcinoma. Asian Pac J Cancer Prev. 2015;16:1397-1401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
48.  Perkhofer L, Walter K, Costa IG, Carrasco MC, Eiseler T, Hafner S, Genze F, Zenke M, Bergmann W, Illing A, Hohwieler M, Köhntop R, Lin Q, Holzmann KH, Seufferlein T, Wagner M, Liebau S, Hermann PC, Kleger A, Müller M. Tbx3 fosters pancreatic cancer growth by increased angiogenesis and activin/nodal-dependent induction of stemness. Stem Cell Res. 2016;17:367-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
49.  Li Z, Wang Y, Duan S, Shi Y, Li S, Zhang X, Ren J. Expression of TBX3 in Hepatocellular Carcinoma and Its Clinical Implication. Med Sci Monit. 2018;24:9324-9333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
50.  Lachenmayer A, Alsinet C, Savic R, Cabellos L, Toffanin S, Hoshida Y, Villanueva A, Minguez B, Newell P, Tsai HW, Barretina J, Thung S, Ward SC, Bruix J, Mazzaferro V, Schwartz M, Friedman SL, Llovet JM. Wnt-pathway activation in two molecular classes of hepatocellular carcinoma and experimental modulation by sorafenib. Clin Cancer Res. 2012;18:4997-5007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 230]  [Article Influence: 19.2]  [Reference Citation Analysis (1)]
51.  Seehawer M, Heinzmann F, D'Artista L, Harbig J, Roux PF, Hoenicke L, Dang H, Klotz S, Robinson L, Doré G, Rozenblum N, Kang TW, Chawla R, Buch T, Vucur M, Roth M, Zuber J, Luedde T, Sipos B, Longerich T, Heikenwälder M, Wang XW, Bischof O, Zender L. Necroptosis microenvironment directs lineage commitment in liver cancer. Nature. 2018;562:69-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 261]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
52.  Miao ZF, Liu XY, Xu HM, Wang ZN, Zhao TT, Song YX, Xing YN, Huang JY, Zhang JY, Xu H, Xu YY. Tbx3 overexpression in human gastric cancer is correlated with advanced tumor stage and nodal status and promotes cancer cell growth and invasion. Virchows Arch. 2016;469:505-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
53.  Takeuchi A, Asano N, Imatani A, Saito M, Jin X, Kanno T, Hatta W, Uno K, Koike T, Masamune A. Suppressed cellular senescence mediated by T-box3 in aged gastric epithelial cells may contribute to aging-related carcinogenesis. Cancer Res Commun. 2022;2:772-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
54.  López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10570]  [Cited by in F6Publishing: 9098]  [Article Influence: 827.1]  [Reference Citation Analysis (0)]
55.  Yu M, Hazelton WD, Luebeck GE, Grady WM. Epigenetic Aging: More Than Just a Clock When It Comes to Cancer. Cancer Res. 2020;80:367-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 63]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
56.  Jin XX, Xie XL, Niu F, Yin KG, Ji CG, Cui JF, Liu L, Feng ZJ. A Single-Center Follow-Up Study of Low-Grade Gastric Intraepithelial Neoplasia and the Screening of Key Genes of Precancerous Lesions. Front Oncol. 2022;12:899055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
57.  Lu J, Li XP, Dong Q, Kung HF, He ML. TBX2 and TBX3: the special value for anticancer drug targets. Biochim Biophys Acta. 2010;1806:268-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
58.  Cioffi M, Vallespinos-Serrano M, Trabulo SM, Fernandez-Marcos PJ, Firment AN, Vazquez BN, Vieira CR, Mulero F, Camara JA, Cronin UP, Perez M, Soriano J, G Galvez B, Castells-Garcia A, Haage V, Raj D, Megias D, Hahn S, Serrano L, Moon A, Aicher A, Heeschen C. MiR-93 Controls Adiposity via Inhibition of Sirt7 and Tbx3. Cell Rep. 2015;12:1594-1605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
59.  Lee JM, Cho KW, Kim EJ, Tang Q, Kim KS, Tickle C, Jung HS. A contrasting function for miR-137 in embryonic mammogenesis and adult breast carcinogenesis. Oncotarget. 2015;6:22048-22059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
60.  Peres J, Kwesi-Maliepaard EM, Rambow F, Larue L, Prince S. The tumour suppressor, miR-137, inhibits malignant melanoma migration by targetting the TBX3 transcription factor. Cancer Lett. 2017;405:111-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
61.  Cioffi M, Trabulo SM, Sanchez-Ripoll Y, Miranda-Lorenzo I, Lonardo E, Dorado J, Reis Vieira C, Ramirez JC, Hidalgo M, Aicher A, Hahn S, Sainz B Jr, Heeschen C. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem cells. Gut. 2015;64:1936-1948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 106]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
62.  Lou G, Huang JCLWYYY. Biological functions of miR-183 on chemosensitivity of laryngeal cancer cells. J BUON. 2021;26:785-791.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Paul A, Limon BH, Hossain M, Raza T. An integrated computational approach to screening of alkaloids inhibitors of TBX3 in breast cancer cell lines. J Biomol Struct Dyn. 2022;1-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
64.  Zhang JF, He ML, Qi Dong, Xie WD, Chen YC, Lin MC, Leung PC, Zhang YO, Kung HF. Aqueous extracts of Fructus Ligustri Lucidi enhance the sensitivity of human colorectal carcinoma DLD-1 cells to doxorubicin-induced apoptosis via Tbx3 suppression. Integr Cancer Ther. 2011;10:85-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
65.  Willmer T, Damerell V, Smyly S, Sims D, Du Toit M, Ncube S, Sinkala M, Govender D, Sturrock E, Blackburn JM, Prince S. Targeting the oncogenic TBX3:nucleolin complex to treat multiple sarcoma subtypes. Am J Cancer Res. 2021;11:5680-5700.  [PubMed]  [DOI]  [Cited in This Article: ]