Editorial Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastrointest Oncol. Jan 15, 2012; 4(1): 1-8
Published online Jan 15, 2012. doi: 10.4251/wjgo.v4.i1.1
Debate about TGFBR1 and the susceptibility to colorectal cancer
Laura Valle, Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, 08908 Hospitalet de Llobregat, Spain
Author contributions: Valle L solely contributed to this paper.
Supported by The Spanish Ministry of Science and Innovation (Grant BFU2009-10281 and Ramón y Cajal contract) and the Scientific Foundation of Asociación Española Contra el Cáncer
Correspondence to: Laura Valle, PhD, Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, Av. Gran Vía 199-203, 08908 Hospitalet de Llobregat, Barcelona, Spain. lvalle@iconcologia.net
Telephone: +34-93-2607145 Fax: +34-93-2607466
Received: March 3, 2011
Revised: October 21, 2011
Accepted: October 28, 2011
Published online: January 15, 2012

Abstract

Recent years have witnessed enormous progress in our understanding of the genetic predisposition to colorectal cancer (CRC). Estimates suggest that all or most genetic susceptibility mechanisms proposed so far, ranging from high-penetrance genes to low-risk alleles, account for about 60% of the population-attributable fraction of CRC predisposition. In this context, there is increasing interest in the gene encoding the transforming growth factor β receptor 1 (TGFBR1); first when over a decade ago a common polymorphism in exon 1 (rs11466445, TGFBR1*6A/9A) was suggested to be a risk allele for CRC, then when linkage studies identified the chromosomal region where the gene is located as susceptibility locus for familial CRC, and more recently when the allele-specific expression (ASE) of the gene was proposed as a risk factor for CRC. Published data on the association of TGFBR1 with CRC, regarding polymorphisms and ASE and including sporadic and familial forms of the disease, are often contradictory. This review gives a general overview of the most relevant studies in order to clarify the role of TGFBR1 in the field of CRC genetic susceptibility.

Key Words: Transforming growth factorβreceptor 1, Transforming growth factorβreceptor 1*6A, 9q linkage peak, Allele-specific expression, Colorectal cancer risk



GENETICS OF COLORECTAL CANCER

The estimated annual worldwide incidence of colorectal cancer (CRC) is 1 235 108, with a mortality rate of 609 051[1]. Lynch syndrome, the most common CRC syndrome formerly also known as hereditary non-polyposis CRC, accounts for approximately 3% of all CRC cases, while Familial Adenomatous Polyposis syndrome occurs in about 0.01% of the population, as well as other rarer polyposis syndromes, such as MYH-adenomatous polyposis, hereditary mixed polyposis, juvenile polyposis or Peutz-Jeghers syndromes among others[2,3]. All the above mentioned syndromes show high penetrance with respect to CRC risk; however, collectively they account for at most 3%-6% of all CRCs. Based on crude estimates of familial CRC, defined by the presence of two or more first-degree relatives affected with CRC, it is thought to involve approximately 20% of all CRC[4,5]. In all, both case-control and twin studies indicate that hereditary factors contribute considerably to CRC[6].

Because of the complexity regarding the etiology of CRC that includes environmental as well as genetic factors, we now know that genetic susceptibility to CRC underlies an unknown proportion of both familial and sporadic cases. Therefore, the distinction between sporadic and familial cases of CRC is less dramatic than it has been classically considered. In fact, it has been thought for some time that a large fraction of familial and a majority of sporadic CRCs are likely to be due to low-penetrance alleles. Genome-wide association studies (GWAS) have identified a new repertoire of cancer susceptibility genes and loci characterized by high frequency of the risk allele and low relative risk, in line with the common disease-common variant paradigm[7-12]. There has been some enthusiasm in using combinations of low-risk alleles in individual risk assessment. However, even in combination, low-risk alleles tend to minimally improve the predictive power of the existing risk factors, such as family history. Recently, it was estimated that all or most genetic susceptibility mechanisms proposed so far account for about 60% of the population-attributable fraction of CRC predisposition[13], leaving approximately 40% of the genetic predisposition unexplained.

Moderate-penetrance genes are now thought to play a very important role in the already unexplained CRC susceptibility. However, until recently, important technical difficulties have prevented researchers from identifying them. These variants are rare, which may cause the inability of GWAS to detect them, and the risks conferred by them too low to be detected by linkage studies, the classical tool to identify high-penetrance disease genes. Hopefully, current whole-exome or -genome sequencing techniques will allow us to discover them.

Candidate gene approaches have sometimes been successful in identifying susceptibility variants. In this regard, considerable attention has been focused on the gene coding the transforming growth factor β receptor 1 (TGFBR1).

TRANSFORMING GROWTH FACTOR β PATHWAY IN CRC

The transforming growth factor β (TGF-β) pathway is an important modulator of several biological processes, including cell proliferation, differentiation, migration and apoptosis[14]. The signaling pathway of TGF-β1, the most abundant form of TGF-β, plays an important role in carcinogenesis, having both tumor-suppressing and promoting activities. In normal and premalignant cells, TGF-β enforces homeostasis and suppresses tumor progression directly through cell-autonomous tumor-suppressive effects (cytostasis, differentiation, apoptosis) or indirectly through effects on the stroma (suppression of inflammation and stroma-derived mitogens). However, when cancer cells lose TGF-β tumor-suppressive response, they can use TGF-β to their advantage to initiate immune evasion, growth factor production, differentiation into an invasive phenotype and metastatic dissemination, or to establish and expand metastatic colonies[15].

Briefly, TGF-β binds to the cell surface receptor transforming growth factor β receptor 2 (TGFBR2), which results in their binding to and phosphorylation of TGFBR1. Subsequently, SMADs are phosphorylated by activated TGFBR1 and translocated into the nucleus, where they regulate transcription of their target genes[14,16,17].

The TGF-β and bone morphogenetic protein (BMP) pathways play an important role in the pathogenesis of CRC and other intestinal tumors. Inactivating somatic mutations in TGFBR2 occur in CRCs with microsatellite instability[18,19]. Whether TGFBR2 mutations have a causative role in colorectal carcinogenesis or whether they arise as a consequence of the hypermutable phenotype observed in cells with defective mismatch repair machinery is still a topic of debate. Mutations in TGFBR1 have been identified in CRC cell lines but are uncommon[20]. TGFBR1*6A/9A (rs11466445) is a common polymorphism in exon 1 of the gene that results in the deletion of three alanines from a stretch of nine alanines. Functional studies have suggested that TGFBR1*6A responds less well than the TGFBR1*9A allele to growth inhibitory signals of TGF-β. Moreover, it has been shown that TGFBR1*6A is somatically acquired in CRC and further analyses suggested that this somatic acquisition is a critical event in the early stages of cancer development, occurring both in epithelial and stromal cells during colorectal carcinogenesis[21,22]. SMAD2 and SMAD4 both map to chromosome 18q, a region commonly deleted in colon adenocarcinomas[19]. SMAD4 is mutated in 10%-38% of CRCs[23-27] and SMAD2 in 6%-8%[27,28]. SMAD3 mutations seem to be infrequent in tumors. BMP members belong to the TGF-β superfamily of proteins and the BMP pathway is inactivated in up to 70% of CRCs[29].

From the germline point of view, mutations in SMAD4 and BMPR1A cause juvenile polyposis, a CRC susceptibility syndrome[30,31], and GWAS have identified low penetrance susceptibility alleles in the BMP pathway and SMAD[9,12]. TGFBR1 risk alleles will be discussed in the following section.

TGFBR1 POLYMORPHIC VARIANTS AND CRC RISK

TGFBR1*6A/9A (rs11466445) was identified in 1998 by Pasche et al[32]. From that moment on, it was considered a potential tumor susceptibility allele that has been associated with an increased incidence of several types of tumors, including CRC. Overall, however, for a long time the results were inconclusive and mixed, partially because small cohorts had been studied[33-55]. In order to overcome this problem, meta-analyses considering increasing number of studies have been published in the last years[56-60]. One of the most recent meta-analysis included 32 studies (9 for CRC) from different countries and types of tumors and comprised a total of 13 662 cancer cases and 14 147 controls, 2833 and 4255 respectively for CRC[59]. The results showed significantly higher overall cancer risk associated with TGFBR1*6A in all genetic models (for allelic effect: OR = 1.11, 95% CI: 1.03-1.21). However, when the analysis was subdivided by cancer type, significant associations were found in breast (for allelic effect: OR = 1.16, 95% CI: 1.01-1.34) and ovarian (for allelic effect: OR = 1.24, 95% CI: 1.00-1.54) cancers, but not in colorectal, bladder and prostate tumors. While for bladder and prostate cancers results were clearly non-significant, for CRC slightly borderline non-significance was found (for allelic effect: OR = 1.16, 95% CI: 0.94-1.42). A subsequent meta-analysis based on 14 subgroup CRC case-control studies found that the heterozygote form 6A/9A showed a 12% increase of CRC risk compared to 9A/9A (OR = 1.12, 95% CI: 1.02-1.23), although no association was found for 6A/6A homozygotes[60].

In addition to TGFBR1*6A, another polymorphic variant, Int7G24A (rs334354), has also been implicated in cancer susceptibility, associations with kidney, bladder, invasive breast and non-small cell lung carcinomas, and osteosarcoma being reported[45,46,61-63]. When analyzed in CRC case-control cohorts, contradictory results have been obtained[64,65].

Due to the previous conflicting results published on TGFBR1 variants, especially TGFBR1*6A, Carvajal-Carmona et al[66] carried out a thorough assessment of TGFBR1 polymorphisms in relation to CRC risk in three series of CRC cases (n = 3101) and controls (n = 3334) of northern European ancestry. They found no association between CRC and TGBR1*6A, not even when they considered interaction with other candidate variants in CRC genes that map close to the TGF-β/BMP pathway genes GREM1, BMP2, BMP4 and SMAD7. They also performed a comprehensive evaluation of common and rarer variants (n = 102) within the 75 kb haplotype block containing TGFBR1 and concluded that common variation at the TGFBR1 locus is unlikely to be associated with CRC risk. The lack of association persisted when long-range regulation was assessed by extending the analysis 500 kb on each side of the TGFBR1 haplotype block or by analyzing haplotypes instead of alleles.

Abulí et al[67] recently screened 7 polymorphic TGFBR1 variants with potential pathogenic effect, including TGFBR1*6A, in 515 CRC cases and 515 controls. Their results showed borderline significant association for TGFBR1*6A (unadjusted P = 0.049, dominant inheritance), but did not reach significance after multiple testing correction. No evidence of association with CRC risk was found for the other six TGFBR1 variants analyzed.

ALLELE-SPECIFIC EXPRESSION OF TGFBR1

Allele-specific expression (ASE), meaning that one allele is less or more expressed than the other, is now considered a mutational mechanism with phenotypic consequences and has been associated with increased cancer risk in some instances[68-71].

Studies in mice point to the relevance of haploinsufficiency of TGFBR1 in colorectal tumorigenesis. While the homozygous loss of Tgfbr1 in mice (Tgfbr1-/-) is lethal, the heterozygous loss (Tgfbr1+/-) causes no obvious phenotypic traits. However, when Tgfbr1+/- mice were bred into mice heterozygous for the ApcMin mutation, the double mutants acquired approximately a 2-fold increase in the number of intestinal adenomas in comparison with the ApcMin/+ mice, as well as colonic carcinomas, suggesting that haploinsufficiency for Tgfbr1 predisposes to CRC[72].

Given the previous existing evidence, we studied ASE of TGFBR1 in unaffected tissue (blood) of CRC patients and controls using the SNaPshot technology and found that the reduced expression of one allele was a quantitative trait that was more common in patients (10%-20%) than in controls (1%-3%), conferring a substantially increased risk of CRC (OR = 8.7, 95% CI: 2.6-29.1). We also assessed the effect of ASE on the TGF-β pathway observing a subtle reduction of the SMAD-mediated signaling. Two major TGFBR1 haplotypes were predominant among the ASE cases; however, the causative genetic cause was not identified[73]. Given the potential use of ASE of TGFBR1 in the clinical evaluation of CRC risk, additional studies were consequently published[66,74-78]. Table 1 shows a summary of the studies published to date.

Table 1 Main characteristics of the studies published on allele-specific expression of transforming growth factor β receptor 1 and colorectal cancer risk.
StudyMajority populationSampleMethodAllelic markersInformative cases/controlsASE (binary) cases/controlsASE higher in CRC cases
BinaryContinuous
Valle et al[73] 2008CaucasianBloodSNaPshotrs334348138/105221.0%/2.9%YesYes
rs7871490
rs334349
rs1590
Guda et al[74] 2009CaucasianLymph. cell linePyroseqrs868Familial: 46/1734.3%/0%No
Normal colonrs334348Sporadic: 44/030%/-
rs334349
rs420549
rs1590
Carvajal-Carmona et al[66] 2010CaucasianLymph. cell lineGenescan*6A/9AFamilial: 24/45329.2%/26.7%NoNo
SNaPshotrs1590
Pasche et al[75] 2010CaucasianLymph. cell lineSNaPshotrs33434874/0314.9%/-Yes
rs7871490
rs334349
rs1590
Tomsic et al[76] 2010CaucasianBloodPyroseqrs8681109/12531.8%/1.6%NoYes
rs334348246.8%/31.2%
rs334349
rs420549
rs1590
Seguí et al[77] 2011CaucasianNormal colonPyroseqrs334349171/9030%/2.2%NoNo
AshkenaziLymphocytesrs785089522.3%/2.2%
rs420549
rs1590
Abadie et al[78] 2011CaucasianBloodSNaPshotrs33434869/9830%/0%NoNo
rs7871490
rs334349
rs1590

Although the balance is level regarding the number of studies that found more ASE in cases and controls, or no differences between both groups, several characteristics that may tip the balance should be considered: On the one hand, when trying to assess the robustness and reproducibility of the two standard methodologies to measure ASE, SNaPshot and pyrosequencing, it was found that, in contrast to pyrosequencing, SNaPshot yields high variability among different SNP markers, being highly dependent on RNA quality to obtain reliable and consistent results[74,76,77,78]. Recently Abadie et al[78] reported a study where exactly the same methodological approach as the original study[73] had been used, finding no differences between cases and controls. In that instance, high quality RNA was ensured by the careful and standardized procedure of blood collection and sample processing carried out, thus guaranteeing consistent results even when SNaPshot was used to measure ASE[78]. On the other hand, it seems that ASE might be more common among individuals who carry minor alleles for specific TGFBR1 SNPs. Therefore ASE could result more or less frequently, depending on the SNP markers used to define informative individuals. Another source of variability among studies might be the different unaffected tissues from which nucleic acids for ASE determination were extracted. Although we observed no differences in ASE frequencies when studying two different groups of CRC patients with different sources (uncultured) of nucleic acids[77], the fact that different types of tissues from the same individuals have never been analyzed still leaves a certain degree of uncertainty.

In all, the most recent results suggest that ASE differences between cases and controls are too subtle, if not nonexistent, to be used to assess CRC risk[66,74,76-78]. As clearly pointed out by several authors, the real extent of ASE of TGFBR1 will probably only be known when technological and conceptual advances allow greater precision and circumvent the need of naturally occurring transcribed SNPs to differentiate the two alleles. With the current technologies and depending on the population studied, ASE can only be assessed in 25%-60% of all individuals, leaving open the possibility that ASE occurs, or does not occur, preferentially in those individuals uninformative for the allelic markers analyzed.

TGFBR1 IN FAMILIAL CRC
Linkage to 9q22 in familial CRC

The TGFBR1 gene co-localizes to the chromosomal region 9q22.2-31.2, first identified in 2003 as a putative susceptibility locus for colorectal neoplasia by Wiesner and colleagues using data from both discordant and concordant sibling pairs from 53 families[79,80]. This was later validated in studies from Sweden and the United Kingdom[81,82] and the locus designated as Colorectal Cancer Susceptibility 1 (CRCS1; MIM608812). It was estimated that it accounted for approximately 35% of the inherited susceptibility to CRC. Very recently, Wiesner and co-workers validated the original results in an independent sample (256 sibling pairs belonging to 110 families, 179 and 50 of them, respectively, from the original study) where the evidence of linkage to this region increased and the linkage on 9q22-31 was narrowed from 13.5 to 7.7 cm[83].

Other genome-wide linkage studies have failed to detect the 9q locus and it seems the underlying complexity of the 9q region and the differences in study design could explain the contradictory results[84]. Evidence suggests that the disease locus housed on 9q is specific to a familial syndrome with a phenotype of young age of onset and/or severity of the colorectal neoplasia[80,83].

TGFBR1*6A in familial CRC

Given the previous reports suggesting that TGFBR1*6A was a CRC susceptibility allele in the general population, in 2005 Pasche and co-workers hypothesized that this allele might explain a proportion of CRC patients with family histories meeting the Amsterdam criteria but without an identifiable mutation in a MMR gene, the so called familial CRC of type X (fCRC-X). In their series, TGFBR1*6A homozygotes were 13-fold times more frequent among fCRC-X patients (n = 64) than in the general population[85]. Other studies unsuccessfully tried to replicate the original results in larger series of fCRC-X patients[51], or of familial CRC selected based on more relaxed criteria to define heritability, such as the CORGI cohort[66]. Similarly, the TGFBR1*6A allele was excluded as a disease-causing variant in the CRC families that showed linkage at 9q22[51,82,86].

ASE of TGFBR1 in familial CRC

When ASE of TGFBR1 was first described as a putative CRC susceptibility genetic trait, increasing interest was generated about its role in familial CRC. Already in the original study, familial cases were over-represented. Although the proportion of ASE was slightly higher among familial (25%) than non-familial cases (17%), the difference was not statistically significant[73]. Guda et al[74] studied ASE in 46 informative familial cases, 31 of which (derived from 22 families) had previously shown linkage to 9q22. They detected ASE in two individuals, both from different families belonging to the 9q22 kinked cohort. Carvajal-Carmona et al[66] assessed ASE in 46 informative familial CRC patients from the CORGI cohort and did not find higher ASE in cases compared with controls. Likewise, Abadie et al[78], who included familial history and early-onset diagnosis of CRC as criteria for patients’ selection, did not find increased ASE in cases than in controls.

CONCLUSION

Researchers were very enthusiastic when TGFBR1*6A was first proposed as a putative CRC susceptibility allele, both for CRC in the general population and for familial CRC. However, the information obtained from larger series, meta-analyses and comprehensive studies including genetic variation in the whole TGFBR1 gene and large flanking regions suggest that the role of this allele in CRC predisposition is, at best, very subtle. A similar scenario is found regarding ASE of TGFBR1 related to CRC susceptibility. In this case, methodological improvements are key to perform an accurate assessment of ASE. The development of new technological advances that allow the measurement of ASE in a more precise and informative manner will provide the definitive answer to what the real extent of ASE of TGFBR1 in CRC patients is.

ACKNOWLEDGMENTS

Valle L thanks Gabriel Capellá and Víctor Moreno for helpful discussions and critical review.

Footnotes

Peer reviewers: Runjan Chetty, Professor, Department of Pathology and Gene Regulation, University of Glasgow, Western Infirmary (Pathology), Dumbarton Road, Glasgow, G11 6NT, Scotland, United Kingdom; Ke-Bin Liu, Assistant Professor, Department of Biochemistry and Molecular Biology, School of Medicine, Medical College of Georgia, Augusta, GA 30912, United States

S- Editor Wang JL L- Editor Roemmele A E- Editor Zheng XM

References
1.  Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM.  GLOBOCAN 2008, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 [Internet]. Lyon, France: International Agency for Research on Cancer 2010;  Available from: http://globocan.iarc.fr.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch JF, Boland CR. Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet. 2009;76:1-18.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Sweet K, Willis J, Zhou XP, Gallione C, Sawada T, Alhopuro P, Khoo SK, Patocs A, Martin C, Bridgeman S. Molecular classification of patients with unexplained hamartomatous and hyperplastic polyposis. JAMA. 2005;294:2465-2473.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Aaltonen L, Johns L, Järvinen H, Mecklin JP, Houlston R. Explaining the familial colorectal cancer risk associated with mismatch repair (MMR)-deficient and MMR-stable tumors. Clin Cancer Res. 2007;13:356-361.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Abdel-Rahman WM, Peltomäki P. Lynch syndrome and related familial colorectal cancers. Crit Rev Oncog. 2008;14:1-22; discussion 23-31.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  de la Chapelle A. Genetic predisposition to colorectal cancer. Nat Rev Cancer. 2004;4:769-780.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Zanke BW, Greenwood CM, Rangrej J, Kustra R, Tenesa A, Farrington SM, Prendergast J, Olschwang S, Chiang T, Crowdy E. Genome-wide association scan identifies a colorectal cancer susceptibility locus on chromosome 8q24. Nat Genet. 2007;39:989-994.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Tomlinson I, Webb E, Carvajal-Carmona L, Broderick P, Kemp Z, Spain S, Penegar S, Chandler I, Gorman M, Wood W. A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat Genet. 2007;39:984-988.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Broderick P, Carvajal-Carmona L, Pittman AM, Webb E, Howarth K, Rowan A, Lubbe S, Spain S, Sullivan K, Fielding S. A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk. Nat Genet. 2007;39:1315-1317.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Tomlinson IP, Webb E, Carvajal-Carmona L, Broderick P, Howarth K, Pittman AM, Spain S, Lubbe S, Walther A, Sullivan K. A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat Genet. 2008;40:623-630.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Tenesa A, Farrington SM, Prendergast JG, Porteous ME, Walker M, Haq N, Barnetson RA, Theodoratou E, Cetnarskyj R, Cartwright N. Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat Genet. 2008;40:631-637.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Houlston RS, Webb E, Broderick P, Pittman AM, Di Bernardo MC, Lubbe S, Chandler I, Vijayakrishnan J, Sullivan K, Penegar S. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet. 2008;40:1426-1435.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hemminki K, Försti A, Lorenzo Bermejo J. Surveying the genomic landscape of colorectal cancer. Am J Gastroenterol. 2009;104:789-790.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Massagué J, Blain SW, Lo RS. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell. 2000;103:295-309.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Massagué J. TGFbeta in Cancer. Cell. 2008;134:215-230.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Akhurst RJ, Derynck R. TGF-beta signaling in cancer--a double-edged sword. Trends Cell Biol. 2001;11:S44-S51.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Xu Y, Pasche B. TGF-beta signaling alterations and susceptibility to colorectal cancer. Hum Mol Genet. 2007;16 Spec No 1:R14-R20.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, Fan RS, Zborowska E, Kinzler KW, Vogelstein B. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 1995;268:1336-1338.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Grady WM, Markowitz SD. Genetic and epigenetic alterations in colon cancer. Annu Rev Genomics Hum Genet. 2002;3:101-128.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Ku JL, Park SH, Yoon KA, Shin YK, Kim KH, Choi JS, Kang HC, Kim IJ, Han IO, Park JG. Genetic alterations of the TGF-beta signaling pathway in colorectal cancer cell lines: a novel mutation in Smad3 associated with the inactivation of TGF-beta-induced transcriptional activation. Cancer Lett. 2007;247:283-292.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Pasche B, Knobloch TJ, Bian Y, Liu J, Phukan S, Rosman D, Kaklamani V, Baddi L, Siddiqui FS, Frankel W. Somatic acquisition and signaling of TGFBR1*6A in cancer. JAMA. 2005;294:1634-1646.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Bian Y, Knobloch TJ, Sadim M, Kaklamani V, Raji A, Yang GY, Weghorst CM, Pasche B. Somatic acquisition of TGFBR1*6A by epithelial and stromal cells during head and neck and colon cancer development. Hum Mol Genet. 2007;16:3128-3135.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Thiagalingam S, Lengauer C, Leach FS, Schutte M, Hahn SA, Overhauser J, Willson JK, Markowitz S, Hamilton SR, Kern SE. Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nat Genet. 1996;13:343-346.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Takagi Y, Kohmura H, Futamura M, Kida H, Tanemura H, Shimokawa K, Saji S. Somatic alterations of the DPC4 gene in human colorectal cancers in vivo. Gastroenterology. 1996;111:1369-1372.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Salovaara R, Roth S, Loukola A, Launonen V, Sistonen P, Avizienyte E, Kristo P, Järvinen H, Souchelnytskyi S, Sarlomo-Rikala M. Frequent loss of SMAD4/DPC4 protein in colorectal cancers. Gut. 2002;51:56-59.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Ando T, Sugai T, Habano W, Jiao YF, Suzuki K. Analysis of SMAD4/DPC4 gene alterations in multiploid colorectal carcinomas. J Gastroenterol. 2005;40:708-715.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Forbes SA, Bhamra G, Bamford S, Dawson E, Kok C, Clements J, Menzies A, Teague JW, Futreal PA, Stratton MR. The Catalogue of Somatic Mutations in Cancer (COSMIC). Curr Protoc Hum Genet. 2008;Chapter 10:Unit 10.11.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Eppert K, Scherer SW, Ozcelik H, Pirone R, Hoodless P, Kim H, Tsui LC, Bapat B, Gallinger S, Andrulis IL. MADR2 maps to 18q21 and encodes a TGFbeta-regulated MAD-related protein that is functionally mutated in colorectal carcinoma. Cell. 1996;86:543-552.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kodach LL, Wiercinska E, de Miranda NF, Bleuming SA, Musler AR, Peppelenbosch MP, Dekker E, van den Brink GR, van Noesel CJ, Morreau H. The bone morphogenetic protein pathway is inactivated in the majority of sporadic colorectal cancers. Gastroenterology. 2008;134:1332-1341.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Howe JR, Roth S, Ringold JC, Summers RW, Järvinen HJ, Sistonen P, Tomlinson IP, Houlston RS, Bevan S, Mitros FA. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science. 1998;280:1086-1088.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Howe JR, Bair JL, Sayed MG, Anderson ME, Mitros FA, Petersen GM, Velculescu VE, Traverso G, Vogelstein B. Germline mutations of the gene encoding bone morphogenetic protein receptor 1A in juvenile polyposis. Nat Genet. 2001;28:184-187.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Pasche B, Luo Y, Rao PH, Nimer SD, Dmitrovsky E, Caron P, Luzzatto L, Offit K, Cordon-Cardo C, Renault B. Type I transforming growth factor beta receptor maps to 9q22 and exhibits a polymorphism and a rare variant within a polyalanine tract. Cancer Res. 1998;58:2727-2732.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Pasche B, Kolachana P, Nafa K, Satagopan J, Chen YG, Lo RS, Brener D, Yang D, Kirstein L, Oddoux C. TbetaR-I(6A) is a candidate tumor susceptibility allele. Cancer Res. 1999;59:5678-5682.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Chen T, de Vries EG, Hollema H, Yegen HA, Vellucci VF, Strickler HD, Hildesheim A, Reiss M. Structural alterations of transforming growth factor-beta receptor genes in human cervical carcinoma. Int J Cancer. 1999;82:43-51.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  van Tilborg AA, de Vries A, Zwarthoff EC. The chromosome 9q genes TGFBR1, TSC1, and ZNF189 are rarely mutated in bladder cancer. J Pathol. 2001;194:76-80.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Stefanovska AM, Efremov GD, Dimovski AJ, Jasar D, Zografski G, Josifovski T, Panovski M, Jankova R, Spiroski M. TbetaR-I(6A) polymorphism is not a tumor susceptibility allele in Macedonian colorectal cancer patients. Correspondence re: B. Pasche et al. Type I TbetaR-I(6A) Is a Candidate Tumor Susceptibility Allele. Cancer Res., 58: 2727-2732, 1998. Cancer Res. 2001;61:8351-8352.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Samowitz WS, Curtin K, Leppert MF, Slattery ML. Uncommon TGFBRI allele is not associated with increased susceptibility to colon cancer. Genes Chromosomes Cancer. 2001;32:381-383.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Baxter SW, Choong DY, Eccles DM, Campbell IG. Transforming growth factor beta receptor 1 polyalanine polymorphism and exon 5 mutation analysis in breast and ovarian cancer. Cancer Epidemiol Biomarkers Prev. 2002;11:211-214.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Kaklamani V, Baddi L, Rosman D, Liu J, Ellis N, Oddoux C, Ostrer H, Chen Y, Ahsan H, Offit K. No major association between TGFBR1*6A and prostate cancer. BMC Genet. 2004;5:28.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Jin Q, Hemminki K, Grzybowska E, Klaes R, Söderberg M, Zientek H, Rogozinska-Szczepka J, Utracka-Hutka B, Pamula J, Pekala W. Polymorphisms and haplotype structures in genes for transforming growth factor beta1 and its receptors in familial and unselected breast cancers. Int J Cancer. 2004;112:94-99.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Suarez BK, Pal P, Jin CH, Kaushal R, Sun G, Jin L, Pasche B, Deka R, Catalona WJ. TGFBR1*6A is not associated with prostate cancer in men of European ancestry. Prostate Cancer Prostatic Dis. 2005;8:50-53.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Spillman MA, Schildkraut JM, Halabi S, Moorman P, Calingaert B, Bentley RC, Marks JR, Murphy S, Berchuck A. Transforming growth factor beta receptor I polyalanine repeat polymorphism does not increase ovarian cancer risk. Gynecol Oncol. 2005;97:543-549.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Kaklamani VG, Baddi L, Liu J, Rosman D, Phukan S, Bradley C, Hegarty C, McDaniel B, Rademaker A, Oddoux C. Combined genetic assessment of transforming growth factor-beta signaling pathway variants may predict breast cancer risk. Cancer Res. 2005;65:3454-3461.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Kaklamani V, Pasche B. Transforming Growth Factor Beta and breast cancer. Cancer Treat Res. 2005;126:129-156.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Chen T, Jackson CR, Link A, Markey MP, Colligan BM, Douglass LE, Pemberton JO, Deddens JA, Graff JR, Carter JH. Int7G24A variant of transforming growth factor-beta receptor type I is associated with invasive breast cancer. Clin Cancer Res. 2006;12:392-397.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Feigelson HS, Patel AV, Diver WR, Stevens VL, Thun MJ, Calle EE. Transforming growth factor beta receptor type I and transforming growth factor beta1 polymorphisms are not associated with postmenopausal breast cancer. Cancer Epidemiol Biomarkers Prev. 2006;15:1236-1237.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  You W, Liu Z, Zhao J, Zheng M, Zheng SY, Liu X, Zhang HT. No association between TGFBR1*6A and lung cancer. J Thorac Oncol. 2007;2:657-659.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Cox DG, Penney K, Guo Q, Hankinson SE, Hunter DJ. TGFB1 and TGFBR1 polymorphisms and breast cancer risk in the Nurses' Health Study. BMC Cancer. 2007;7:175.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Song B, Margolin S, Skoglund J, Zhou X, Rantala J, Picelli S, Werelius B, Lindblom A. TGFBR1(*)6A and Int7G24A variants of transforming growth factor-beta receptor 1 in Swedish familial and sporadic breast cancer. Br J Cancer. 2007;97:1175-1179.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Skoglund J, Song B, Dalén J, Dedorson S, Edler D, Hjern F, Holm J, Lenander C, Lindforss U, Lundqvist N. Lack of an association between the TGFBR1*6A variant and colorectal cancer risk. Clin Cancer Res. 2007;13:3748-3752.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Skoglund Lundin J, Vandrovcova J, Song B, Zhou X, Zelada-Hedman M, Werelius B, Houlston RS, Lindblom A. TGFBR1 variants TGFBR1(*)6A and Int7G24A are not associated with an increased familial colorectal cancer risk. Br J Cancer. 2009;100:1674-1679.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Castillejo A, Mata-Balaguer T, Montenegro P, Ochoa E, Lázaro R, Martínez-Cantó A, Castillejo MI, Guarinos C, Barberá VM, Guillén-Ponce C. The TGFBR1*6A allele is not associated with susceptibility to colorectal cancer in a Spanish population: a case-control study. BMC Cancer. 2009;9:193.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Jakubowska A, Gronwald J, Menkiszak J, Górski B, Huzarski T, Byrski T, Tołoczko-Grabarek A, Gilbert M, Edler L, Zapatka M. BRCA1-associated breast and ovarian cancer risks in Poland: no association with commonly studied polymorphisms. Breast Cancer Res Treat. 2010;119:201-211.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Colleran G, McInerney N, Rowan A, Barclay E, Jones AM, Curran C, Miller N, Kerin M, Tomlinson I, Sawyer E. The TGFBR1*6A/9A polymorphism is not associated with differential risk of breast cancer. Breast Cancer Res Treat. 2010;119:437-442.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hu YS, Pan Y, Li WH, Zhang Y, Li J, Ma BA. Association between TGFBR1*6A and osteosarcoma: a Chinese case-control study. BMC Cancer. 2010;10:169.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Kaklamani VG, Hou N, Bian Y, Reich J, Offit K, Michel LS, Rubinstein WS, Rademaker A, Pasche B. TGFBR1*6A and cancer risk: a meta-analysis of seven case-control studies. J Clin Oncol. 2003;21:3236-3243.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Pasche B, Kaklamani V, Hou N, Young T, Rademaker A, Peterlongo P, Ellis N, Offit K, Caldes T, Reiss M. TGFBR1*6A and cancer: a meta-analysis of 12 case-control studies. J Clin Oncol. 2004;22:756-758.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Zhang HT, Zhao J, Zheng SY, Chen XF. Is TGFBR1*6A really associated with increased risk of cancer? J Clin Oncol. 2005;23:7743-774; author reply 7743-774;.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Liao RY, Mao C, Qiu LX, Ding H, Chen Q, Pan HF. TGFBR1*6A/9A polymorphism and cancer risk: a meta-analysis of 13,662 cases and 14,147 controls. Mol Biol Rep. 2010;37:3227-3232.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Zhang X, Wu L, Sheng Y, Zhou W, Huang Z, Qu J, Gao G, Cai D, Zhang M. The association of polymorphisms on TGFBR1 and colorectal cancer risk: a meta-analysis. Mol Biol Rep. 2011;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Chen T, Jackson C, Costello B, Singer N, Colligan B, Douglass L, Pemberton J, Deddens J, Graff JR, Carter JH. An intronic variant of the TGFBR1 gene is associated with carcinomas of the kidney and bladder. Int J Cancer. 2004;112:420-425.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Zhang HT. Int7G24A variant of the TGFBR1 gene and cancer risk: a meta-analysis of three case-control studies. Lung Cancer. 2005;49:419-420.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Hu YS, Pan Y, Li WH, Zhang Y, Li J, Ma BA. Int7G24A variant of transforming growth factor-beta receptor 1 is associated with osteosarcoma susceptibility in a Chinese population. Med Oncol. 2011;28:622-625.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Försti A, Li X, Wagner K, Tavelin B, Enquist K, Palmqvist R, Altieri A, Hallmans G, Hemminki K, Lenner P. Polymorphisms in the transforming growth factor beta 1 pathway in relation to colorectal cancer progression. Genes Chromosomes Cancer. 2010;49:270-281.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Castillejo A, Mata-Balaguer T, Guarinos C, Castillejo MI, Martínez-Cantó A, Barberá VM, Montenegro P, Ochoa E, Lázaro R, Guillén-Ponce C. The Int7G24A variant of transforming growth factor-beta receptor type I is a risk factor for colorectal cancer in the male Spanish population: a case-control study. BMC Cancer. 2009;9:406.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Carvajal-Carmona LG, Churchman M, Bonilla C, Walther A, Lefèvre JH, Kerr D, Dunlop M, Houlston R, Bodmer WF, Tomlinson I. Comprehensive assessment of variation at the transforming growth factor beta type 1 receptor locus and colorectal cancer predisposition. Proc Natl Acad Sci U S A. 2010;107:7858-7862.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Abulí A, Fernández-Rozadilla C, Giráldez MD, Muñoz J, Gonzalo V, Bessa X, Bujanda L, Reñé JM, Lanas A, García AM. A two-phase case-control study for colorectal cancer genetic susceptibility: candidate genes from chromosomal regions 9q22 and 3q22. Br J Cancer. 2011;105:870-875.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Yan H, Dobbie Z, Gruber SB, Markowitz S, Romans K, Giardiello FM, Kinzler KW, Vogelstein B. Small changes in expression affect predisposition to tumorigenesis. Nat Genet. 2002;30:25-26.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Yan H, Yuan W, Velculescu VE, Vogelstein B, Kinzler KW. Allelic variation in human gene expression. Science. 2002;297:1143.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Raval A, Tanner SM, Byrd JC, Angerman EB, Perko JD, Chen SS, Hackanson B, Grever MR, Lucas DM, Matkovic JJ. Downregulation of death-associated protein kinase 1 (DAPK1) in chronic lymphocytic leukemia. Cell. 2007;129:879-890.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Chen X, Weaver J, Bove BA, Vanderveer LA, Weil SC, Miron A, Daly MB, Godwin AK. Allelic imbalance in BRCA1 and BRCA2 gene expression is associated with an increased breast cancer risk. Hum Mol Genet. 2008;17:1336-1348.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Zeng Q, Phukan S, Xu Y, Sadim M, Rosman DS, Pennison M, Liao J, Yang GY, Huang CC, Valle L. Tgfbr1 haploinsufficiency is a potent modifier of colorectal cancer development. Cancer Res. 2009;69:678-686.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Valle L, Serena-Acedo T, Liyanarachchi S, Hampel H, Comeras I, Li Z, Zeng Q, Zhang HT, Pennison MJ, Sadim M. Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer. Science. 2008;321:1361-1365.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Guda K, Natale L, Lutterbaugh J, Wiesner GL, Lewis S, Tanner SM, Tomsic J, Valle L, de la Chapelle A, Elston RC. Infrequent detection of germline allele-specific expression of TGFBR1 in lymphoblasts and tissues of colon cancer patients. Cancer Res. 2009;69:4959-4961.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Pasche B, Wisinski KB, Sadim M, Kaklamani V, Pennison MJ, Zeng Q, Bellam N, Zimmerman J, Yi N, Zhang K. Constitutively decreased TGFBR1 allelic expression is a common finding in colorectal cancer and is associated with three TGFBR1 SNPs. J Exp Clin Cancer Res. 2010;29:57.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Tomsic J, Guda K, Liyanarachchi S, Hampel H, Natale L, Markowitz SD, Tanner SM, de la Chapelle A. Allele-specific expression of TGFBR1 in colon cancer patients. Carcinogenesis. 2010;31:1800-1804.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Seguí N, Stevens KN, Guinó E, Rozek LS, Moreno VR, Capellá G, Gruber SB, Valle L. No association between germline allele-specific expression of TGFBR1 and colorectal cancer risk in Caucasian and Ashkenazi populations. Br J Cancer. 2011;104:735-740.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Abadie C, Killian A, Tinat J, Bougeard M, Medhaoui D, Cailleux AF, Baert-Desurmont S, Frebourg T. Allelic imbalance of the TGFβR1 is not a major contributor to the genetic predisposition to colorectal cancer. Br J Cancer. 2011;104:1517-158; author reply 1517-158;.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Wiesner GL, Daley D, Lewis S, Ticknor C, Platzer P, Lutterbaugh J, MacMillen M, Baliner B, Willis J, Elston RC. A subset of familial colorectal neoplasia kindreds linked to chromosome 9q22.2-31.2. Proc Natl Acad Sci U S A. 2003;100:12961-12965.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Daley D, Lewis S, Platzer P, MacMillen M, Willis J, Elston RC, Markowitz SD, Wiesner GL. Identification of susceptibility genes for cancer in a genome-wide scan: results from the colon neoplasia sibling study. Am J Hum Genet. 2008;82:723-736.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Skoglund J, Djureinovic T, Zhou XL, Vandrovcova J, Renkonen E, Iselius L, Bisgaard ML, Peltomäki P, Lindblom A. Linkage analysis in a large Swedish family supports the presence of a susceptibility locus for adenoma and colorectal cancer on chromosome 9q22.32-31.1. J Med Genet. 2006;43:e7.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Kemp ZE, Carvajal-Carmona LG, Barclay E, Gorman M, Martin L, Wood W, Rowan A, Donohue C, Spain S, Jaeger E. Evidence of linkage to chromosome 9q22.33 in colorectal cancer kindreds from the United Kingdom. Cancer Res. 2006;66:5003-5006.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Gray-McGuire C, Guda K, Adrianto I, Lin CP, Natale L, Potter JD, Newcomb P, Poole EM, Ulrich CM, Lindor N. Confirmation of linkage to and localization of familial colon cancer risk haplotype on chromosome 9q22. Cancer Res. 2010;70:5409-5418.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Papaemmanuil E, Carvajal-Carmona L, Sellick GS, Kemp Z, Webb E, Spain S, Sullivan K, Barclay E, Lubbe S, Jaeger E. Deciphering the genetics of hereditary non-syndromic colorectal cancer. Eur J Hum Genet. 2008;16:1477-1486.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Bian Y, Caldes T, Wijnen J, Franken P, Vasen H, Kaklamani V, Nafa K, Peterlongo P, Ellis N, Baron JA. TGFBR1*6A may contribute to hereditary colorectal cancer. J Clin Oncol. 2005;23:3074-3078.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Daley D, Morgan W, Lewis S, Willis J, Elston RC, Markowitz SD, Wiesner GL. Is TGFBR1*6A a susceptibility allele for nonsyndromic familial colorectal neoplasia? Cancer Epidemiol Biomarkers Prev. 2007;16:892-894.  [PubMed]  [DOI]  [Cited in This Article: ]