Skip to main content
  • Research article
  • Open access
  • Published:

Association between TGFBR1*6A and osteosarcoma: A Chinese case-control study

Abstract

Background

TGFBR1*6A is a common hypomorphic variant of transforming growth factor β receptor 1 (TGFBR1). TGFBR1*6A is associated with an increased cancer risk, but the association of this polymorphism with osteosarcoma remains unknown. We have measured the frequency of TGFBR1*6A variants in osteosarcoma cases and controls.

Methods

Our case-control study is based on 168 osteosarcoma patients and 168 age- and gender-matched controls. Blood samples were obtained and the TGFBR1*6A variant determined by PCR amplification and DNA sequencing. The odds ratio (OR) and 95% confidence interval (95% CI) for the TGFBR1*6A polymorphism were calculated by unconditional logistic regression, adjusted for both age and gender. Three models - dominant, additive and recessive - were used to analyze the contribution of the TGFBR1*6A variant to osteosarcoma susceptibility.

Results

Heterozygotic and homozygotic TGFBR1*6A variants represented 50.4% and 6.0% of the 168 cases, whereas the controls had 18. 5% and 1.3%, respectively. ORs for homozygosity and heterozygosity of the TGFBR1*6A allele were 4.6 [95% CI, 2.33-7.97] and 2.9 [95% CI, 1.59-5.34] in the additive model. There were significant increases in the TGFBR1*6A variants in osteosarcoma cases compared to control in all 3 models. Further analysis showed that TGFBR1*6A genotypes were not associated with gender, age, or tumor location. However, TGFBR1*6A was significantly associated with less metastasis.

Conclusions

TGFBR1*6A, a dominant polymorphism of TGFBR1, is associated with increased susceptibility and metastasis spread of osteosarcoma.

Peer Review reports

Background

Osteosarcoma is the primary malignancy of bone that has peak occurrences in adolescence and after 50 years of age [1, 2]. Although there have been many studies on its genetics, biology, pathology and clinical aspects, the etiology of osteosarcoma is not well understood. Screening large series of children with osteosarcoma showed that ~4% carried a constitutional germline mutation in p53, suggesting a genetic predisposition of osteosarcoma [3].

TGF-β signaling plays an important role in the development of tumors because it is a potent inhibitor of cell proliferation in most cell types [4, 5]. Tumors frequently lose responsiveness to TGF-β-mediated growth inhibition due to disruption in the TGF-β signaling pathway [6]. TGF-β regulates gene expression and enzymes activity in osteosarcoma, and modulates the effects of hormones on osteosarcoma, suggesting that TGF-β signaling is involved in the development of osteosarcoma [79]. TGF-β binds to its receptor, TGF-β receptor 2 (TGFBR2), which forms a heteomeric complex with TGF-β receptor 1 (TGFBR1). This leads to the transduction of the TGF-β signal from the cell surface to the cytoplasm [10, 11]. Mutation of TGFBR1 may result in a high risk of certain cancers. For example, TGFBR1*6A is a common mutation of TGFBR1, and this variant is generated by deletion of 3 GCG triplets that coding for alanines within a 9 alanine (*9A) sequence at exon 1. TGFBR1*6A mutation can lead to the development of breast cancer, ovarian cancer, and renal cell carcinoma amongst others [1215]. However, the relationship between TGFBR1*6A mutation and osteosarcoma occurrence remains unknown. There have been no reports about the prevalence of TGFBR1*6A mutation in osteosarcoma. Clarification of the relationship between TGFBR1*6A and osteosarcoma may indicate a role of TGF-β signaling in the etiology of osteosarcoma and provide clues that might help to guide treatment of this diseases. To clarify this relationship, we have analyzed TGFBR1 variation in clinical cases of osteosarcoma and normal controls. TGFBR1*6A positively correlated with the occurrence of osteosarcoma.

Methods

Study Subjects

Patients with osteosarcoma admitted to the Department of Orthopedics at Tangdu Hospital from January 2001 to March 2009 were selected to participate in the study, making a total of 168 patients. The mean age in the cases was 30 years (range 10-71 years). All the cases were diagnosed without a familial cancer syndrome. As controls, DNA from 168 blood donors of mixed gender collected at Tangdu Hospital, Xi'an, China, were used. The controls were disease-free. Informed consent was obtained from all the subjects, and the study was approved by the ethical committee at Tangdu Hospital.

TGFBR1*6A genotyping

TGFBR1*6A genotyping invovled the protocol of Song et al [14]. PCR amplification of exon 1 of TGFBR1 was performed with the following primers: Forward 5'-GAG GCG AGG TTT GCT GGG GTG AGG-3' and Reverse 5'-CAT GTT TGA GAA AGA GCA GGA GCG-3'. The reactions were based in the protocol given by Invitrogen (Paisley, UK) in a total volume of 25 μl containing 50 ng DNA and 1.25 U Platinums Taq DNA polymerase. Fluorescently labeled PCR products were separated with an ABI 377 DNA Sequencer. Genotypes analysis was done with the GENESCANTM and GENOTYPERTM software. A product size of 247 bp represented the wild-type TGFBR1 (9A) allele, whereas a product size of 255 bp represented the TGFBR1*6A allele.

Statistical analysis

Genotype distribution in osteosarcoma cases and controls was tested for the Hardy-Weinberg equilibrium, and the 2 variants were shown to be in equilibrium in both the patients and the controls. Differences in TGFBR1*6A variant carrier frequencies between cases and controls for the dominant and recessive models were assessed by the χ2 test. The Armigate treand test was used to calculate P for trends in the additive model. Results are presented as odds ratios (ORs) with 95% test-based confidence intervals (CIs). All P values were 2-sided and P < 0.05 was considered significant.

Results

TGFBR1*6A genotypes and osteosarcoma susceptibility

The frequencies of 9A/6A and 6A/6A genotypes were significantly different between the cases and controls. In 168 cases, 9A/9A, 9A/6A and 6A/6A genotypes re[resented 107, 51 and 10, respectively. In 168 controls, the genotypes of 9A/9A, 9A/6A and 6A/6A were 134, 31 and 3, respectively. Heteozygotic and homozygotic TGFBR1*6A variants were 50.4% and 6.0% in total 168 cases, while they were 18.45% and 1.29% respectively in total 168 controls. When the data were analyzed with the dominant, additive and recessive models, there were significant increases in TGFBR1*6A variants in osteosarcoma cases compared to control in all of the (P < 0.01). The result are shown in Table 1.

Table 1 TGFBR1*6A genotypes in controls and patients with osteosarcoma

TGFBR1*6A genotypes and clinical parameters of osteosarcoma

No significant statistic differences were shown in the association of TGFBR1*6A genotypes with clinical parameters between males and females. Nor was any significant difference found between patirnts under 20 and those over 20. The location of osteosarcomas in long tubular bones and the axial skeleton was not significantly associated with a particular mutation of the TGFBR1*6A genotype. However, in cases with distant metastasis, the frequency of TGFBR1*6A genotypes was significantly decreased compared with those without distant metastasis (P < 0.01). The result are shown in Table 2.

Table 2 Associations of TGFBR1*6A genotypes with clinical parameters

Discussion

The 9A/6A and 6A/6A genotypes of TGFBR1 have a significantly higher frequency in osteosarcoma cases than in controls. TGFBR1*6A is a dominant susceptibility allele in the occurrence of osteosarcoma. The retrospective, non-randomized nature of most case-control studies limits the conclusion that can be reached from them. Because the subjects our study were from different regions of China for a long period from 2001 to 2009, the results have been more informative.

The association between TGFBR1*6A and tumorigenesis seems to be different in our case. Previous reports indicated that TGFBR1*6A increased susceptibility of tumorigenesis and development in renal cell carcinoma, ovarian cancer, and breast cancer [1214, 16], although others found no association of TGFBR1*6A with the occurrence of lung cancer, prostate cancer, colon cancer and bladder cancer [1721]. Thus TGFBR1*6A genotypes mya be specifically related to only certain kinds of cancer, in a population- and tissue-related manner.

TGF-β signaling may not be the same in different tissues, which might explain the tissue-related manner of the association between TGFBR1*6A genotypes and cancer development. TGF-β signaling seems to be involved in tumorigenesis because it may behave aberrantly in its role as a potent inhibitor of cell proliferation in most epithelial and lymphoid cells [22]. Tumors frequently lose responsiveness to TGF-β-mediated growth inhibition due to disruption of its signaling pathway [23]. Loss of expression of TGF-β receptors may be as a prognostic factor in patients with renal cell carcinoma [24]. Receptor interactions at the level of the plasma membrane have been implicated in the regulation of TGF-β signaling pathway. In a mice model, constitutively reduced TGFBR1-mediated TGF-β signaling significantly enhances colorectal cancer development and more rapid tumor cell proliferation [25]. Tissue-specific expression of TGF-β receptors could explain the varying effects of TGF-β in different tissues. For example, targeted deletion of TGF-β receptors in mouse mammary epithelium results in excessive lobular-alveolar cell proliferation [26], but in contrast, no developmental changes were apparent upon deletion of TGF-β receptors in the epithelia of the oral cavity, esophagus, forestomach, pancreas and intestine of mice [2729]. The unresponsiveness of some tissues to TGF-β signaling may explain their insensitivity to TGFBR1*6A variant, and vice versa.

Our study firstly demonstrates the significant association between TGFBR1*6A and osteosarcoma, which may be due to the important role of TGF-β signaling in the control of osteosarcoma. Some data indicates that TGF-β can be an important factor in the occurrence of osteosarcoma. TGF-β receptor is expressed in osteosarcoma [30]. TGF-β increases alkaline phosphatase (AP) activity in the rat osteoblastic cell line, ROS 17/2.8, and may promote osteoblastic differentiation in rat osteosarcoma cells [31]. We found that TGFBR1*6A genotypes are significant associated with osteosarcoma, which lends support to the fact that TGF-β signaling plays a vital role in the occurrence of osteosarcoma. The TGFBR1*6A allele encodes a type I receptor of TGF-β with reduced growth-inhibitory signaling activity [32]. The genetic variant of TGFBR1*6A could represent a functional modification in the TGF-β signaling pathway in osteosarcoma. Although this may not be necessary in determining the occurrence of osteosarcoma, TGFBR1*6A variants appear to increase susceptibility to osteosarcoma.

Osteosarcoma has a high incidence in young children and is often located in long tubular bones [33]. Therefore, we analyzed the association of TGFBR1*6A with some clinical parameters including gender, age, location and distant metastasis. Groups are usually divided at the age of 20, We found no significant association between the two groups above and below this age, and therefore TGFBR1*6A variants are not associated with age. In addition, TGFBR1*6A variants is not associated with the location of osteosarcoma, i.e. when frequency in long tubular bones and axial skeleton were compared. Therefore, TGFBR1*6A variants increased the susceptibility of osteosarcoma universally but not specifically to some cases with different gender, age and location of the tumor. This indicates that TGFBR1*6A makes for some functional modification of TGF-β signaling which stimulates the occurrence of osteosarcoma without regard to gender, age and location.

Analysis of distant metastasis, however, showed that TGFBR1*6A is significantly related with metastasis in osteosarcoma. The decreased risk of distant metastasis of osteosarcoma in TGFBR1*6A variants suggests that TGF-β signaling is involved in the metastasis of osteosarcoma. Tumor metastasis is a complex process and many factors are involved in this process, TGF-β is only one of these factors. For example, TGF-β promotes tumor metastasis in cancer [3436]. Although the functional variants of TGFBR1*6A increases susceptibility to osteosarcoma, it decreases the probability of metastasis. TGFBR1*6A has been shown to enhance the migration and invasion of MCF-7 breast cancer cells [37]. Our finding of decreased metastasis in osteosarcoma cases with TGFBR1*6A contrasts with the result with in breast cancer cells. Since tumor cells may exhibit mutations, making their genotypes different from normal tissues, we examined the possibility of loss of homozygosity of TGFBR1 in osteosarcoma tissues in 10 cases with distant metastasis. TGFBR1*6A genotypes were the same as observed with blood samples (data not shown), with no loss of homozygosity of TGFBR1 in the tumor samples. Although there are differences between our results and those obtained with breast cancer cells, site specificity and tumor specificity for the role of TGFBR1*6A may be the reason for the difference since TGF-beta signaling differs within tissues of the body.

Conclusions

This case-control study shows a significant statistical association between TGFBR1*6A variant and osteosarcoma in a Chinese population. The TGFBR1*6A variant is also significantly associated with the distant metastasis of osteosarcoma in the Chinese population studied.

References

  1. Dorfman HD, Czerniak B: Bone cancers. Cancer. 1995, 75 (1 Suppl): 203-210. 10.1002/1097-0142(19950101)75:1+<203::AID-CNCR2820751308>3.0.CO;2-V.

    Article  CAS  PubMed  Google Scholar 

  2. Messerschmitt PJ, Garcia RM, Abdul-Karim FW, Greenfield EM, Getty PJ: Osteosarcoma. J Am Acad Orthop Surg. 2009, 17 (8): 515-527.

    Article  PubMed  Google Scholar 

  3. McIntyre JF, Smith-Sorensen B, Friend SH, Kassell J, Borresen AL, Yan YX, Russo C, Sato J, Barbier N, Miser J, et al: Germline mutations of the p53 tumor suppressor gene in children with osteosarcoma. J Clin Oncol. 1994, 12 (5): 925-930.

    CAS  PubMed  Google Scholar 

  4. Massague J: TGFbeta in Cancer. Cell. 2008, 134 (2): 215-230. 10.1016/j.cell.2008.07.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Neil JR, Galliher AJ, Schiemann WP: TGF-beta in cancer and other diseases. Future Oncol. 2006, 2 (2): 185-189. 10.2217/14796694.2.2.185.

    Article  CAS  PubMed  Google Scholar 

  6. Podar K, Raje N, Anderson KC: Inhibition of the TGF-beta signaling pathway in tumor cells. Recent Results Cancer Res. 2007, 172: 77-97. full_text.

    Article  CAS  PubMed  Google Scholar 

  7. Mioh H, Chen JK: Differential inhibitory effects of TGF-beta on EGF-, PDGF-, and HBGF-1-stimulated MG63 human osteosarcoma cell growth: possible involvement of growth factor interactions at the receptor and postreceptor levels. J Cell Physiol. 1989, 139 (3): 509-516. 10.1002/jcp.1041390309.

    Article  CAS  PubMed  Google Scholar 

  8. Seitz PK, Zhu BT, Cooper CW: Effect of transforming growth factor beta on parathyroid hormone receptor binding and cAMP formation in rat osteosarcoma cells. J Bone Miner Res. 1992, 7 (5): 541-546. 10.1002/jbmr.5650070510.

    Article  CAS  PubMed  Google Scholar 

  9. Pontbriant CM, Chen JK, Orlando JA: TGF-beta inhibits the platelet-derived growth factor-induced formation of inositol trisphosphate in MG-63 human osteosarcoma cells. J Cell Physiol. 1990, 145 (3): 488-495. 10.1002/jcp.1041450314.

    Article  CAS  PubMed  Google Scholar 

  10. Derynck R, Feng XH: TGF-beta receptor signaling. Biochim Biophys Acta. 1997, 1333 (2): F105-150.

    CAS  PubMed  Google Scholar 

  11. Roberts AB: TGF-beta signaling from receptors to the nucleus. Microbes Infect. 1999, 1 (15): 1265-1273. 10.1016/S1286-4579(99)00258-0.

    Article  CAS  PubMed  Google Scholar 

  12. 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 (2): 392-397. 10.1158/1078-0432.CCR-05-1518.

    Article  CAS  PubMed  Google Scholar 

  13. Rosman DS, Kaklamani V, Pasche B: New insights into breast cancer genetics and impact on patient management. Curr Treat Options Oncol. 2007, 8 (1): 61-73. 10.1007/s11864-007-0021-5.

    Article  PubMed  PubMed Central  Google Scholar 

  14. 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 (8): 1175-1179. 10.1038/sj.bjc.6603961.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 (3): 420-425. 10.1002/ijc.20419.

    Article  CAS  PubMed  Google Scholar 

  16. 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 (17): 3236-3243. 10.1200/JCO.2003.11.524.

    Article  CAS  PubMed  Google Scholar 

  17. Castillejo A, Rothman N, Murta-Nascimento C, Malats N, Garcia-Closas M, Gomez-Martinez A, Lloreta J, Tardon A, Serra C, Garcia-Closas R, et al: TGFB1 and TGFBR1 polymorphic variants in relationship to bladder cancer risk and prognosis. Int J Cancer. 2009, 124 (3): 608-613. 10.1002/ijc.24013.

    Article  CAS  PubMed  Google Scholar 

  18. Castillejo A, Mata-Balaguer T, Montenegro P, Ochoa E, Lazaro R, Martinez-Canto A, Castillejo MI, Guarinos C, Barbera VM, Guillen-Ponce C, et al: The TGFBR1*6A allele is not associated with susceptibility to colorectal cancer in a Spanish population: a case-control study. BMC Cancer. 2009, 9: 193-10.1186/1471-2407-9-193.

    Article  PubMed  PubMed Central  Google Scholar 

  19. 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: 175-10.1186/1471-2407-7-175.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kaklamani V, Baddi L, Rosman D, Liu J, Ellis N, Oddoux C, Ostrer H, Chen Y, Ahsan H, Offit K, et al: No major association between TGFBR1*6A and prostate cancer. BMC Genet. 2004, 5: 28-10.1186/1471-2156-5-28.

    Article  PubMed  PubMed Central  Google Scholar 

  21. 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 (7): 657-659. 10.1097/JTO.0b013e318070ccd7.

    Article  PubMed  Google Scholar 

  22. Roberts AB, Thompson NL, Heine U, Flanders C, Sporn MB: Transforming growth factor-beta: possible roles in carcinogenesis. Br J Cancer. 1988, 57 (6): 594-600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fynan TM, Reiss M: Resistance to inhibition of cell growth by transforming growth factor-beta and its role in oncogenesis. Crit Rev Oncog. 1993, 4 (5): 493-540.

    CAS  PubMed  Google Scholar 

  24. Miyajima A, Asano T, Seta K, Asano T, Kakoi N, Hayakawa M: Loss of expression of transforming growth factor-beta receptor as a prognostic factor in patients with renal cell carcinoma. Urology. 2003, 61 (5): 1072-1077. 10.1016/S0090-4295(02)02553-0.

    Article  PubMed  Google Scholar 

  25. Zeng Q, Phukan S, Xu Y, Sadim M, Rosman DS, Pennison M, Liao J, Yang GY, Huang CC, Valle L, et al: Tgfbr1 haploinsufficiency is a potent modifier of colorectal cancer development. Cancer Res. 2009, 69 (2): 678-686. 10.1158/0008-5472.CAN-08-3980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Forrester E, Chytil A, Bierie B, Aakre M, Gorska AE, Sharif-Afshar AR, Muller WJ, Moses HL: Effect of conditional knockout of the type II TGF-beta receptor gene in mammary epithelia on mammary gland development and polyomavirus middle T antigen induced tumor formation and metastasis. Cancer Res. 2005, 65 (6): 2296-2302. 10.1158/0008-5472.CAN-04-3272.

    Article  CAS  PubMed  Google Scholar 

  27. Ijichi H, Chytil A, Gorska AE, Aakre ME, Fujitani Y, Fujitani S, Wright CV, Moses HL: Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes Dev. 2006, 20 (22): 3147-3160. 10.1101/gad.1475506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lu SL, Herrington H, Reh D, Weber S, Bornstein S, Wang D, Li AG, Tang CF, Siddiqui Y, Nord J, et al: Loss of transforming growth factor-beta type II receptor promotes metastatic head-and-neck squamous cell carcinoma. Genes Dev. 2006, 20 (10): 1331-1342. 10.1101/gad.1413306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Munoz NM, Upton M, Rojas A, Washington MK, Lin L, Chytil A, Sozmen EG, Madison BB, Pozzi A, Moon RT, et al: Transforming growth factor beta receptor type II inactivation induces the malignant transformation of intestinal neoplasms initiated by Apc mutation. Cancer Res. 2006, 66 (20): 9837-9844. 10.1158/0008-5472.CAN-06-0890.

    Article  CAS  PubMed  Google Scholar 

  30. Kloen P, Jennings CL, Gebhardt MC, Springfield DS, Mankin HJ: Expression of transforming growth factor-beta (TGF-beta) receptors, TGF-beta 1 and TGF-beta 2 production and autocrine growth control in osteosarcoma cells. Int J Cancer. 1994, 58 (3): 440-445. 10.1002/ijc.2910580323.

    Article  CAS  PubMed  Google Scholar 

  31. Noda M, Rodan GA: Type beta transforming growth factor (TGF beta) regulation of alkaline phosphatase expression and other phenotype-related mRNAs in osteoblastic rat osteosarcoma cells. J Cell Physiol. 1987, 133 (3): 426-437. 10.1002/jcp.1041330303.

    Article  CAS  PubMed  Google Scholar 

  32. Pasche B, Kolachana P, Nafa K, Satagopan J, Chen YG, Lo RS, Brener D, Yang D, Kirstein L, Oddoux C, et al: TbetaR-I(6A) is a candidate tumor susceptibility allele. Cancer Res. 1999, 59 (22): 5678-5682.

    CAS  PubMed  Google Scholar 

  33. Picci P: Osteosarcoma (osteogenic sarcoma). Orphanet J Rare Dis. 2007, 2: 6-10.1186/1750-1172-2-6.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Cui W, Fowlis DJ, Bryson S, Duffie E, Ireland H, Balmain A, Akhurst RJ: TGFbeta1 inhibits the formation of benign skin tumors, but enhances progression to invasive spindle carcinomas in transgenic mice. Cell. 1996, 86 (4): 531-542. 10.1016/S0092-8674(00)80127-0.

    Article  CAS  PubMed  Google Scholar 

  35. Muraoka RS, Dumont N, Ritter CA, Dugger TC, Brantley DM, Chen J, Easterly E, Roebuck LR, Ryan S, Gotwals PJ, et al: Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases. J Clin Invest. 2002, 109 (12): 1551-1559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Padua D, Massague J: Roles of TGFbeta in metastasis. Cell Res. 2009, 19 (1): 89-102. 10.1038/cr.2008.316.

    Article  CAS  PubMed  Google Scholar 

  37. Rosman DS, Phukan S, Huang CC, Pasche B: TGFBR1*6A enhances the migration and invasion of MCF-7 breast cancer cells through RhoA activation. Cancer Res. 2008, 68 (5): 1319-1328. 10.1158/0008-5472.CAN-07-5424.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Pre-publication history

Download references

Acknowledgements

This work was supported by grants from national natural science foundation of China (No. 30672143).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bao-An Ma.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

YSH and YP originally conceptualized the analysis, prepared the samples, and wrote the manuscript. WHL contributed to PCR and gene sequencing studies. YZ help with data analysis. BAM coordinated the study and participated in its design. All authors read and approved the final manuscript.

Yun-Sheng Hu, Yong Pan contributed equally to this work.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Hu, YS., Pan, Y., Li, WH. et al. Association between TGFBR1*6A and osteosarcoma: A Chinese case-control study. BMC Cancer 10, 169 (2010). https://doi.org/10.1186/1471-2407-10-169

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2407-10-169

Keywords