Publications

Found 36 results
Filters: Author is H. Chen  [Clear All Filters]
2016
M. J. Li, Zhang, C. M., Lan, X. Y., Fang, X. T., Lei, C. Z., Chen, H., Li, M. J., Zhang, C. M., Lan, X. Y., Fang, X. T., Lei, C. Z., and Chen, H., Analysis of POU1F1 gene DdeI polymorphism in Chinese goats, vol. 15, p. -, 2016.
M. J. Li, Zhang, C. M., Lan, X. Y., Fang, X. T., Lei, C. Z., Chen, H., Li, M. J., Zhang, C. M., Lan, X. Y., Fang, X. T., Lei, C. Z., and Chen, H., Analysis of POU1F1 gene DdeI polymorphism in Chinese goats, vol. 15, p. -, 2016.
F. W. Wang, Wang, M. L., Guo, C., Wang, N., Li, X. W., Chen, H., Dong, Y. Y., Chen, X. F., Wang, Z. M., Li, H. Y., Wang, F. W., Wang, M. L., Guo, C., Wang, N., Li, X. W., Chen, H., Dong, Y. Y., Chen, X. F., Wang, Z. M., and Li, H. Y., Cloning and characterization of a novel betaine aldehyde dehydrogenase gene from Suaeda corniculata, vol. 15, p. -, 2016.
F. W. Wang, Wang, M. L., Guo, C., Wang, N., Li, X. W., Chen, H., Dong, Y. Y., Chen, X. F., Wang, Z. M., Li, H. Y., Wang, F. W., Wang, M. L., Guo, C., Wang, N., Li, X. W., Chen, H., Dong, Y. Y., Chen, X. F., Wang, Z. M., and Li, H. Y., Cloning and characterization of a novel betaine aldehyde dehydrogenase gene from Suaeda corniculata, vol. 15, p. -, 2016.
W. Teng, Chen, H., Guo, F., Du, X., Fu, X., Fang, Y., Zhang, H., Fang, M., Ding, M., Teng, W., Chen, H., Guo, F., Du, X., Fu, X., Fang, Y., Zhang, H., Fang, M., and Ding, M., Expression and distribution of SP and its NK1 receptor in the brain-gut axis in neonatal maternally separated rat model with visceral hypersensitivity, vol. 15, p. -, 2016.
W. Teng, Chen, H., Guo, F., Du, X., Fu, X., Fang, Y., Zhang, H., Fang, M., Ding, M., Teng, W., Chen, H., Guo, F., Du, X., Fu, X., Fang, Y., Zhang, H., Fang, M., and Ding, M., Expression and distribution of SP and its NK1 receptor in the brain-gut axis in neonatal maternally separated rat model with visceral hypersensitivity, vol. 15, p. -, 2016.
L. Chen, Chen, Y. Q., Ding, A. M., Chen, H., Xia, F., Wang, W. F., Sun, Y. H., Chen, L., Chen, Y. Q., Ding, A. M., Chen, H., Xia, F., Wang, W. F., and Sun, Y. H., Genome-wide analysis of TCP family in tobacco, vol. 15, p. -, 2016.
L. Chen, Chen, Y. Q., Ding, A. M., Chen, H., Xia, F., Wang, W. F., Sun, Y. H., Chen, L., Chen, Y. Q., Ding, A. M., Chen, H., Xia, F., Wang, W. F., and Sun, Y. H., Genome-wide analysis of TCP family in tobacco, vol. 15, p. -, 2016.
X. Huang, Jing, Y., Liu, D. J., Yang, B. Y., Chen, H., Li, M., Huang, X., Jing, Y., Liu, D. J., Yang, B. Y., Chen, H., and Li, M., Whole-transcriptome sequencing of Pinellia ternata using the Illumina platform, vol. 15, p. -, 2016.
X. Huang, Jing, Y., Liu, D. J., Yang, B. Y., Chen, H., Li, M., Huang, X., Jing, Y., Liu, D. J., Yang, B. Y., Chen, H., and Li, M., Whole-transcriptome sequencing of Pinellia ternata using the Illumina platform, vol. 15, p. -, 2016.
2015
Y. C. Fan, Min, L., Chen, H., and Liu, Y. L., Alternative splicing isoform of T cell factor 4K suppresses the proliferation and metastasis of non-small cell lung cancer cells, vol. 14, pp. 14009-14018, 2015.
W. P. Wu, Cao, J., Wu, J. Y., Chen, H., and Wang, D., Anticancer activity of Bombyx batryticatus ethanol extract against the human tumor cell line HeLa, vol. 14, pp. 79-88, 2015.
W. Zhang, Zhang, L. C., Chen, H., Tang, P. F., and Zhang, L. H., Association between polymorphisms in insulin-like growth factor-1 and risk of osteoporosis, vol. 14, pp. 7655-7660, 2015.
H. Chen, Lan, H., Huang, P., Zhang, Y., Yuan, X., Huang, X., Huang, J., and Zhang, H., Characterization of OsPM19L1 encoding an AWPM-19-like family protein that is dramatically induced by osmotic stress in rice, vol. 14, pp. 11994-12005, 2015.
H. B. Liu, Zhu, Y., Yang, Q. C., Shen, Y., Zhang, X. J., and Chen, H., Expression and clinical significance of ADAM17 protein in esophageal squamous cell carcinoma, vol. 14, pp. 4391-4398, 2015.
K. X. Gao, Chen, N. B., Liu, W. J., Li, R., Lan, X. Y., Chen, H., Lei, C. Z., and Dang, R. H., Frequency of gray coat color in native Chinese horse breeds, vol. 14, pp. 14144-14150, 2015.
H. Chen, He, R., Wang, Z. L., Wang, S. Y., Chen, Y., Zhu, Z. C., and Chen, X. M., Genetic diversity and variability in populations of the white wax insect Ericerus pela, assessed by AFLP analysis, vol. 14, pp. 17820-17827, 2015.
H. Chen, Ma, Y., Zhang, W. F., Ma, T., and Wu, H. X., Molecular phylogeny of Colletotrichum (Sordariomycetes: Glomerellaceae) inferred from multiple gene sequences, vol. 14, pp. 13649-13662, 2015.
P. Huang, Chen, H., Mu, R., Yuan, X., Zhang, H. S., and Huang, J., OsMYB511 encodes a MYB domain transcription activator early regulated by abiotic stress in rice, vol. 14, pp. 9506-9517, 2015.
2014
B. Wang, Yang, J., Xiao, J., Liang, B., Zhou, H. X., Su, Z., Xu, S., Chen, H., Ma, C., Deng, J., Li, D., Ou, X., and Feng, Y., Association of XRCC5 polymorphisms with COPD and COPD-related phenotypes in the Han Chinese population: a case-control cohort study, vol. 13, pp. 7070-7078, 2014.
X. X. Lu, Hu, J. J., Fang, Y., Wang, Z. T., Xie, J. J., Zhan, Q., Deng, X. X., Chen, H., Jin, J. B., Peng, C. H., Liu, J., Li, H. W., and Shen, B. Y., A case-control study indicates that the TRIB1 gene is associated with pancreatic cancer, vol. 13, pp. 6142-6147, 2014.
L. Gao, Zhang, J. P., Chen, H., Zhang, S. N., Chen, L. B., Tan, J. P., Liu, M. L., Meng, L. L., Wang, Y. H., Zhang, R., Liu, Y. L., and Cai, W. B., Characteristics of immune cell changes before and after immunotherapy and their clinical significance in patients with unexplained recurrent spontaneous abortion, vol. 13, pp. 1169-1178, 2014.
L. Gao, Zhang, J. P., Chen, H., Guo, Z. J., Chen, L. B., Tan, J. P., Wang, Y. H., Zhang, R., Liu, Y. L., and Cai, W. B., Fetal fibronectin detection for preterm birth prediction, vol. 13, pp. 1323-1328, 2014.
Z. L. Li, Guan, H. H., Xiao, X. M., Hui, Y., Jia, W. X., Yu, R. X., Chen, H., and Li, C. R., Germline mutation analysis in the CYLD gene in Chinese patients with multiple trichoepitheliomas, vol. 13, pp. 9650-9655, 2014.
L. H. Cao, Kuang, B. H., Chen, C., Hu, C., Sun, Z., Chen, H., Wang, S. S., and Luo, Y., Identification of a novel duplication mutation in the VHL gene in a large Chinese family with Von Hippel-Lindau (VHL) syndrome, vol. 13, pp. 10177-10183, 2014.
H. Chen, Duan, J., and Zuo, F., Mechanism of the reversal effect of mifepristone on drug resistance of the human cervical cancer cell line HeLa/MMC, vol. 13, pp. 1288-1295, 2014.
L. - Z. Zhang, Ren, G., Hua, L. - S., Lan, X. - Y., Lei, C. - Z., and Chen, H., Polymorphism in the 5ꞌ-UTR of the insulin-like growth factor I gene associated with production traits in Chinese cattle, vol. 13, pp. 6899-6905, 2014.
J. Zhang, Cui, H. H., Wang, Y., Zhang, Q. X., Deng, S. L., Chen, Y. D., and Chen, H., Study of Hgp44 from Porphyromonas gingivalis on inducing HUVECs to secrete IL-6 and IL-8, vol. 13, pp. 2208-2219, 2014.
H. B. Xu, Yang, H., Liu, G., and Chen, H., Systematic review of accuracy of prenatal diagnosis for abnormal chromosome diseases by microarray technology, vol. 13, pp. 9115-9121, 2014.
2013
Y. W. Sun, Xuan, Q., Shu, Q. A., Wu, S. S., Chen, H., Xiao, J., Xiang, P., Zhu, Y. P., Wang, F. L., and Zhao, S. T., Correlation of tumor relapse and elevated expression of survivin and vascular endothelial growth factor in superficial bladder transitional cell carcinoma, vol. 12, pp. 1045-1053, 2013.
Athanassiadou P, Grapsa D, Athanassiades P, Gonidi M, et al. (2008). The prognostic significance of COX-2 and survivin expression in ovarian cancer. Pathol. Res. Pract. 204: 241-249. http://dx.doi.org/10.1016/j.prp.2007.11.004 PMid:18171606   Celen O, Kahraman I, Yildirim E and Berberoglu U (2004). Correlation of vascular endothelial growth factor (VEGF) and CEA with clinicopathological variables in colorectal cancer patients. Neoplasma 51: 293-299. PMid:15254661   Deveraux QL and Reed JC (1999). IAP family proteins - suppressors of apoptosis. Genes Dev. 13: 239-252. http://dx.doi.org/10.1101/gad.13.3.239 PMid:9990849   Fleshner NE, Herr HW, Stewart AK, Murphy GP, et al. (1996). The National Cancer Data Base report on bladder carcinoma. The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer 78: 1505-1513. http://dx.doi.org/10.1002/(SICI)1097-0142(19961001)78:7<1505::AID-CNCR19>3.0.CO;2-3   Grabowski P, Kuhnel T, Muhr-Wilkenshoff F, Heine B, et al. (2003). Prognostic value of nuclear survivin expression in oesophageal squamous cell carcinoma. Br. J. Cancer 88: 115-119. http://dx.doi.org/10.1038/sj.bjc.6600696 PMid:12556969 PMCid:2376783   Ikeguchi M, Ueda T, Sakatani T, Hirooka Y, et al. (2002). Expression of survivin messenger RNA correlates with poor prognosis in patients with hepatocellular carcinoma. Diagn. Mol. Pathol. 11: 33-40. http://dx.doi.org/10.1097/00019606-200203000-00007 PMid:11854600   Islam A, Kageyama H, Takada N, Kawamoto T, et al. (2000). High expression of Survivin, mapped to 17q25, is significantly associated with poor prognostic factors and promotes cell survival in human neuroblastoma. Oncogene 19: 617-623. http://dx.doi.org/10.1038/sj.onc.1203358 PMid:10698506   Jin Q, Hemminki K, Enquist K, Lenner P, et al. (2005). Vascular endothelial growth factor polymorphisms in relation to breast cancer development and prognosis. Clin. Cancer Res. 11: 3647-3653. http://dx.doi.org/10.1158/1078-0432.CCR-04-1803 PMid:15897560   Kirkali Z, Chan T, Manoharan M, Algaba F, et al. (2005). Bladder cancer: epidemiology, staging and grading, and diagnosis. Urology 66: 4-34. http://dx.doi.org/10.1016/j.urology.2005.07.062 PMid:16399414   Koukourakis MI, Papazoglou D, Giatromanolaki A, Bougioukas G, et al. (2004). VEGF gene sequence variation defines VEGF gene expression status and angiogenic activity in non-small cell lung cancer. Lung Cancer 46: 293-298. http://dx.doi.org/10.1016/j.lungcan.2004.04.037 PMid:15541813   Monzo M, Rosell R, Felip E, Astudillo J, et al. (1999). A novel anti-apoptosis gene: Re-expression of survivin messenger RNA as a prognosis marker in non-small-cell lung cancers. J. Clin. Oncol. 17: 2100-2104. PMid:10561264   Mori A, Wada H, Nishimura Y, Okamoto T, et al. (2002). Expression of the antiapoptosis gene survivin in human leukemia. Int. J. Hematol. 75: 161-165. http://dx.doi.org/10.1007/BF02982021 PMid:11939262   Plate KH, Breier G, Weich HA and Risau W (1992). Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 359: 845-848. http://dx.doi.org/10.1038/359845a0 PMid:1279432   Riedel F, Gotte K, Bergler W and Hormann K (2001). Inverse correlation of apoptotic and angiogenic markers in squamous cell carcinoma of the head and neck. Oncol. Rep. 8: 471-476. PMid:11295065   Rohayem J, Diestelkoetter P, Weigle B, Oehmichen A, et al. (2000). Antibody response to the tumor-associated inhibitor of apoptosis protein survivin in cancer patients. Cancer Res. 60: 1815-1817. PMid:10766164   Sato K, Sasaki R, Ogura Y, Shimoda N, et al. (1998). Expression of vascular endothelial growth factor gene and its receptor (flt-1) gene in urinary bladder cancer. Tohoku J. Exp. Med. 185: 173-184. http://dx.doi.org/10.1620/tjem.185.173 PMid:9823778   Schultz IJ, Kiemeney LA, Witjes JA, Schalken JA, et al. (2003). Survivin mRNA expression is elevated in malignant urothelial cell carcinomas and predicts time to recurrence. Anticancer Res. 23: 3327-3331. PMid:12926071   Schultz IJ, Kiemeney LA, Karthaus HF, Witjes JA, et al. (2004). Survivin mRNA copy number in bladder washings predicts tumor recurrence in patients with superficial urothelial cell carcinomas. Clin. Chem. 50: 1425-1428. http://dx.doi.org/10.1373/clinchem.2004.032003 PMid:15277352   Smith BD, Smith GL, Carter D, Sasaki CT, et al. (2000). Prognostic significance of vascular endothelial growth factor protein levels in oral and oropharyngeal squamous cell carcinoma. J. Clin. Oncol. 18: 2046-2052. PMid:10811669   Suzuki A, Ito T, Kawano H, Hayashida M, et al. (2000). Survivin initiates procaspase 3/p21 complex formation as a result of interaction with Cdk4 to resist Fas-mediated cell death. Oncogene 19: 1346-1353. http://dx.doi.org/10.1038/sj.onc.1203429 PMid:10713676   Swana HS, Grossman D, Anthony JN, Weiss RM, et al. (1999). Tumor content of the antiapoptosis molecule survivin and recurrence of bladder cancer. N. Engl. J. Med. 341: 452-453. http://dx.doi.org/10.1056/NEJM199908053410614 PMid:10438269   Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, et al. (2006). Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur. Urol. 49: 466-477. http://dx.doi.org/10.1016/j.eururo.2005.12.031 PMid:16442208   Tran J, Rak J, Sheehan C, Saibil SD, et al. (1999). Marked induction of the IAP family antiapoptotic proteins survivin and XIAP by VEGF in vascular endothelial cells. Biochem. Biophys. Res. Commun. 264: 781-788. http://dx.doi.org/10.1006/bbrc.1999.1589 PMid:10544009   Waters WB (1996). Invasive bladder cancer - where do we go from here? J. Urol. 155: 1910-1911. http://dx.doi.org/10.1016/S0022-5347(01)66044-0   Zhu H, Chen XP, Zhang WG, Luo SF, et al. (2005). Expression and significance of new inhibitor of apoptosis protein survivin in hepatocellular carcinoma. World J. Gastroenterol. 11: 3855-3859. PMid:15991282
J. Wang, Wang, C., Gao, Y., Lan, X. - Y., Lei, C. - Z., Wang, J. - Q., and Chen, H., Impacts of single nucleotide polymorphisms and haplotypes in the bovine Dapper1 gene on body weight, vol. 12, pp. 1254-1268, 2013.
Barrett JC, Fry B, Maller J and Daly MJ (2005). Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics 21: 263-265. http://dx.doi.org/10.1093/bioinformatics/bth457 PMid:15297300   Cerpa W, Toledo EM, Varela-Nallar L and Inestrosa NC (2009). The role of Wnt signaling in neuroprotection. Drug News Perspect. 22: 579-591. http://dx.doi.org/10.1358/dnp.2009.22.10.1443391 PMid:20140278   Cheyette BN, Waxman JS, Miller JR, Takemaru K, et al. (2002). Dapper, a Dishevelled-associated antagonist of beta-catenin and JNK signaling, is required for notochord formation. Dev. Cell 2: 449-461. http://dx.doi.org/10.1016/S1534-5807(02)00140-5   Dale RM, Sisson BE and Topczewski J (2009). The emerging role of Wnt/PCP signaling in organ formation. Zebrafish 6: 9-14. http://dx.doi.org/10.1089/zeb.2008.0563 PMid:19250029 PMCid:2758485   Fisher DA, Kivimae S, Hoshino J, Suriben R, et al. (2006). Three Dact gene family members are expressed during embryonic development and in the adult brains of mice. Dev. Dyn. 235: 2620-2630. http://dx.doi.org/10.1002/dvdy.20917 PMid:16881060   Fukuda T, Kokabu S, Ohte S, Sasanuma H, et al. (2010). Canonical Wnts and BMPs cooperatively induce osteoblastic differentiation through a GSK3beta-dependent and beta-catenin-independent mechanism. Differentiation 80: 46-52. http://dx.doi.org/10.1016/j.diff.2010.05.002 PMid:20546990   Gao X, Wen J, Zhang L, Li X, et al. (2008). Dapper1 is a nucleocytoplasmic shuttling protein that negatively modulates Wnt signaling in the nucleus. J. Biol. Chem. 283: 35679-35688. http://dx.doi.org/10.1074/jbc.M804088200 PMid:18936100   Gloy J, Hikasa H and Sokol SY (2002). Frodo interacts with Dishevelled to transduce Wnt signals. Nat. Cell Biol. 4: 351-357. PMid:11941372   Katoh M and Katoh M (2003). Identification and characterization of human DAPPER1 and DAPPER2 genes in silico. Int. J. Oncol. 22: 907-913. PMid:12632086   Kawai M, Mushiake S, Bessho K, Murakami M, et al. (2007). Wnt/Lrp/beta-catenin signaling suppresses adipogenesis by inhibiting mutual activation of PPARgamma and C/EBPalpha. Biochem. Biophys. Res. Commun. 363: 276-282. http://dx.doi.org/10.1016/j.bbrc.2007.08.088 PMid:17888405   Kimchi-Sarfaty C, Oh JM, Kim IW, Sauna ZE, et al. (2007). A "silent" polymorphism in the MDR1 gene changes substrate specificity. Science 315: 525-528. http://dx.doi.org/10.1126/science.1135308 PMid:17185560   Komar AA (2007). Silent SNPs: impact on gene function and phenotype. Pharmacogenomics 8: 1075-1080. http://dx.doi.org/10.2217/14622416.8.8.1075 PMid:17716239   Kweekel DM, Antonini NF, Nortier JW, Punt CJ, et al. (2009). Explorative study to identify novel candidate genes related to oxaliplatin efficacy and toxicity using a DNA repair array. Br. J. Cancer 101: 357-362. http://dx.doi.org/10.1038/sj.bjc.6605134 PMid:19536092 PMCid:2720215   Lango H, Palmer CN, Morris AD, Zeggini E, et al. (2008). Assessing the combined impact of 18 common genetic variants of modest effect sizes on type 2 diabetes risk. Diabetes 57: 3129-3135. http://dx.doi.org/10.2337/db08-0504 PMid:18591388 PMCid:2570411   Marty A, Amigues Y, Servin B, Renand G, et al. (2010). Genetic variability and linkage disequilibrium patterns in the bovine DNAJA1 gene. Mol. Biotechnol. 44: 190-197. http://dx.doi.org/10.1007/s12033-009-9228-y PMid:20012712   Mullenbach R, Lagoda PJ and Welter C (1989). An efficient salt-chloroform extraction of DNA from blood and tissues. Trends Genet. 5: 391. PMid:2623762   Nei M and Roychoudhury AK (1974). Sampling variances of heterozygosity and genetic distance. Genetics 76: 379-390. PMid:4822472 PMCid:1213072   Sham P, Bader JS, Craig I, O'Donovan M, et al. (2002). DNA Pooling: a tool for large-scale association studies. Nat. Rev. Genet. 3: 862-871. http://dx.doi.org/10.1038/nrg930 PMid:12415316   Stephens M, Smith NJ and Donnelly P (2001). A new statistical method for haplotype reconstruction from population data. Am. J. Hum. Genet. 68: 978-989. http://dx.doi.org/10.1086/319501 PMid:11254454 PMCid:1275651   Su Y, Zhang L, Gao X, Meng F, et al. (2007). The evolutionally conserved activity of Dapper2 in antagonizing TGF-beta signaling. FASEB J. 21: 682-690. http://dx.doi.org/10.1096/fj.06-6246com PMid:17197390   Tee JM, van Rooijen C, Boonen R and Zivkovic D (2009). Regulation of slow and fast muscle myofibrillogenesis by Wnt/ beta-catenin and myostatin signaling. PLoS One 4: e5880. http://dx.doi.org/10.1371/journal.pone.0005880 PMid:19517013 PMCid:2690692   Wang J, Li ZJ, Lan XY, Hua LS, et al. (2010). Two novel SNPs in the coding region of the bovine PRDM16 gene and its associations with growth traits. Mol. Biol. Rep. 37: 571-577. http://dx.doi.org/10.1007/s11033-009-9816-8 PMid:19760096   Waxman JS, Hocking AM, Stoick CL and Moon RT (2004). Zebrafish Dapper1 and Dapper2 play distinct roles in Wnt-mediated developmental processes. Development 131: 5909-5921. http://dx.doi.org/10.1242/dev.01520 PMid:15539487   Xu N, Chen CY and Shyu AB (1997). Modulation of the fate of cytoplasmic mRNA by AU-rich elements: key sequence features controlling mRNA deadenylation and decay. Mol. Cell Biol. 17: 4611-4621. PMid:9234718 PMCid:232314   Xu N, Loflin P, Chen CY and Shyu AB (1998). A broader role for AU-rich element-mediated mRNA turnover revealed by a new transcriptional pulse strategy. Nucleic Acids Res. 26: 558-565. http://dx.doi.org/10.1093/nar/26.2.558 PMid:9421516 PMCid:147286   Xu Y, Liu J, Lan X, Zhang Y, et al. (2011). Consistent effects of single and combined SNP(s) within bovine paired box 7 gene (Pax7) on growth traits. J. Genet. 90: e53-e57. PMid:21873775   Zhang L, Gao X, Wen J, Ning Y, et al. (2006). Dapper 1 antagonizes Wnt signaling by promoting dishevelled degradation. J. Biol. Chem. 281: 8607-8612. http://dx.doi.org/10.1074/jbc.M600274200 PMid:16446366   Zhao H, Nettleton D and Dekkers JCM (2007). Evaluation of linkage disequilibrium measures between multi-allelic markers as predictors of linkage disequilibrium between single nucleotide polymorphisms. Genet. Res. 89: 1-6. http://dx.doi.org/10.1017/S0016672307008634 PMid:17517154
2012
J. Wang, Wang, C., Tian, R., Huang, Y. - Z., Lai, X. - S., Lan, X. - Y., Wang, J. - Q., and Chen, H., Sequence variants in the bovine PRDM16 gene associated with body weight in Chinese cattle breeds, vol. 11, pp. 746-755, 2012.
Chen H and Leibenguth F (1995). Studies on multilocus fingerprints, RAPD markers, and mitochondrial DNA of a gynogenetic fish (Carassius auratus gibelio). Biochem. Genet. 33: 297-306. http://dx.doi.org/10.1007/BF02399929 PMid:8748455 Chen DX, Jin QJ, Fang XT, Zhang CL, et al. (2010). Analysis of the polymorphisms in the caprine PRDM16, SHH and SF-1 genes and their association with production traits in goats. Small Ruminant Res. 93: 193-197. http://dx.doi.org/10.1016/j.smallrumres.2010.04.022 Cousin B, Cinti S, Morroni M, Raimbault S, et al. (1992). Occurrence of brown adipocytes in rat white adipose tissue: molecular and morphological characterization. J. Cell Sci. 10: 931-942. Farmer SR (2008). Molecular determinants of brown adipocyte formation and function. Genes Dev. 22: 1269-1275. http://dx.doi.org/10.1101/gad.1681308 Jenuwein T (2001). Re-SET-ting heterochromatin by histone methyltransferases. Trends Cell Biol. 11: 266-273. http://dx.doi.org/10.1016/S0962-8924(01)02001-3 Kajimura S, Seale P, Tomaru T, Erdjument-Bromage H, et al. (2008). Regulation of the brown and white fat gene programs through a PRDM16/CtBP transcriptional complex. Genes Dev. 22: 1397-1409. http://dx.doi.org/10.1101/gad.1666108 Kimchi-Sarfaty C, Oh JM, Kim IW, Sauna ZE, et al. (2007). A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science 315: 525-528. http://dx.doi.org/10.1126/science.1135308 PMid:17185560 Kinameri E, Inoue T, Aruga J, Imayoshi I, et al. (2008). Prdm proto-oncogene transcription factor family expression and interaction with the Notch-Hes pathway in mouse neurogenesis. PLoS One 3: e3859. http://dx.doi.org/10.1371/journal.pone.0003859 PMid:19050759    PMCid:2585159 Komar AA (2007). Silent SNPs: impact on gene function and phenotype. Pharmacogenomics 8: 1075-1080. http://dx.doi.org/10.2217/14622416.8.8.1075 PMid:17716239 Lai X, Lan X, Chen H, Wang X, et al. (2009). A novel SNP of the Hesx1 gene in bovine and its associations with average daily gain. Mol. Biol. Rep. 36: 1677-1681. http://dx.doi.org/10.1007/s11033-008-9368-3 PMid:18853282 Lan XY, Pan CY, Chen H and Zhang CL (2007). An AluI PCR-RFLP detecting a silent allele at the goat POU1F1 locus and its association with production traits. Small Ruminant Res. 73: 8-12. http://dx.doi.org/10.1016/j.smallrumres.2006.10.009 Nedergaard J, Bengtsson T and Cannon B (2007). Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 293: E444-E452. http://dx.doi.org/10.1152/ajpendo.00691.2006 PMid:17473055 Nei M and Roychoudhury AK (1974). Sampling variances of heterozygosity and genetic distance. Genetics 76: 379-390. PMid:4822472    PMCid:1213072 Oh I, Shimizu H, Satoh T, Okada S, et al. (2006). Identification of nesfatin-1 as a satiety molecule in the hypothalamus. Nature 443: 709-712. http://dx.doi.org/10.1038/nature05162 PMid:17036007 Ren G, Chen H, Zhang LZ, Lan XY, et al. (2010). A coding SNP of LHX4 gene is associated with body weight and body length in bovine. Mol. Biol. Rep. 37: 417-422. http://dx.doi.org/10.1007/s11033-009-9486-6 PMid:19283511 Rhee EJ, Oh KW, Lee WY, Kim SY, et al. (2006). Effects of two common polymorphisms of peroxisome proliferator-activated receptor-gamma gene on metabolic syndrome. Arch. Med. Res. 37: 86-94. http://dx.doi.org/10.1016/j.arcmed.2005.04.008 PMid:16314192 Rosado EL, Bressan J, Martins MF, Cecon PR, et al. (2007). Polymorphism in the PPARgamma2 and beta2-adrenergic genes and diet lipid effects on body composition, energy expenditure and eating behavior of obese women. Appetite 49: 635-643. http://dx.doi.org/10.1016/j.appet.2007.04.003 PMid:17658197 Sauna ZE, Kimchi-Sarfaty C, Ambudkar SV and Gottesman MM (2007). Silent polymorphisms speak: how they affect pharmacogenomics and the treatment of cancer. Cancer Res. 67: 9609-9612. http://dx.doi.org/10.1158/0008-5472.CAN-07-2377 PMid:17942888 Seale P, Kajimura S, Yang W, Chin S, et al. (2007). Transcriptional control of brown fat determination by PRDM16. Cell Metab. 6: 38-54. http://dx.doi.org/10.1016/j.cmet.2007.06.001 PMid:17618855    PMCid:2564846 Seale P, Bjork B, Yang W, Kajimura S, et al. (2008). PRDM16 controls a brown fat/skeletal muscle switch. Nature 454: 961-967. http://dx.doi.org/10.1038/nature07182 PMid:18719582    PMCid:2583329 Walczak R, Tontonoz P and Edward AD (2003). PPAR[gamma] Signaling in Adipose Tissue Development. In: Handbook of Cell Signaling, Academic Press, Burlington, 39-46. Wang YH, Bower NI, Reverter A, Tan SH, et al. (2009). Gene expression patterns during intramuscular fat development in cattle. J. Anim. Sci. 87: 119-130. http://dx.doi.org/10.2527/jas.2008-1082 PMid:18820161 Warner DR, Horn KH, Mudd L, Webb CL, et al. (2007). PRDM16/MEL1: a novel Smad binding protein expressed in murine embryonic orofacial tissue. Biochim. Biophys. Acta 1773: 814-820. http://dx.doi.org/10.1016/j.bbamcr.2007.03.016 PMid:17467076 Yang LL, Hua Q, Liu RK and Yang Z (2009). Association between two common polymorphisms of PPARgamma gene and metabolic syndrome families in a Chinese population. Arch. Med. Res. 40: 89-96. http://dx.doi.org/10.1016/j.arcmed.2008.11.005 PMid:19237017 Zhang C, Wang Y, Chen H, Lan X, et al. (2007). Enhance the efficiency of single-strand conformation polymorphism analysis by short polyacrylamide gel and modified silver staining. Anal. Biochem. 365: 286-287. http://dx.doi.org/10.1016/j.ab.2007.03.023 PMid:17449006
2011
C. He, Wang, C., Chang, Z. H., Guo, B. L., Li, R., Yue, X. P., Lan, X. Y., Chen, H., and Lei, C. Z., AGPAT6 polymorphism and its association with milk traits of dairy goats, vol. 10, pp. 2747-2756, 2011.
Agarwal AK, Barnes RI and Garg A (2006). Functional characterization of human 1-acylglycerol-3-phosphate acyltransferase isoform 8: cloning, tissue distribution, gene structure, and enzymatic activity. Arch. Biochem. Biophys. 449: 64-76. http://dx.doi.org/10.1016/j.abb.2006.03.014 PMid:16620771 Agarwal AK, Sukumaran S, Bartz R, Barnes RI, et al. (2007). Functional characterization of human 1-acylglycerol- 3-phosphate-O-acyltransferase isoform 9: cloning, tissue distribution, gene structure, and enzymatic activity. J. Endocrinol. 193: 445-457. http://dx.doi.org/10.1677/JOE-07-0027 PMid:17535882 Aguado B and Campbell RD (1998). Characterization of a human lysophosphatidic acid acyltransferase that is encoded by a gene located in the class III region of the human major histocompatibility complex. J. Biol. Chem. 273: 4096-4105. http://dx.doi.org/10.1074/jbc.273.7.4096 PMid:9461603 Beigneux AP, Vergnes L, Qiao X, Quatela S, et al. (2006). Agpat6 - a novel lipid biosynthetic gene required for triacylglycerol production in mammary epithelium. J. Lipid Res. 47: 734-744. http://dx.doi.org/10.1194/jlr.M500556-JLR200 PMid:16449762    PMCid:3196597 Bionaz M and Loor JJ (2008). ACSL1, AGPAT6, FABP3, LPIN1, and SLC27A6 are the most abundant isoforms in bovine mammary tissue and their expression is affected by stage of lactation. J. Nutr. 138: 1019-1024. PMid:18492828 Chen YQ, Kuo MS, Li S, Bui HH, et al. (2008). AGPAT6 is a novel microsomal glycerol-3-phosphate acyltransferase. J. Biol. Chem. 283: 10048-10057. http://dx.doi.org/10.1074/jbc.M708151200 PMid:18238778    PMCid:2442282 Coleman RA and Lee DP (2004). Enzymes of triacylglycerol synthesis and their regulation. Prog. Lipid Res. 43: 134-176. http://dx.doi.org/10.1016/S0163-7827(03)00051-1 Kimchi-Sarfaty C, Oh JM, Kim IW, Sauna ZE, et al. (2007). A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science 315: 525-528. http://dx.doi.org/10.1126/science.1135308 PMid:17185560 Komar AA (2007). Silent SNPs: impact on gene function and phenotype. Pharmacogenomics. 8: 1075-1080. http://dx.doi.org/10.2217/14622416.8.8.1075 PMid:17716239 Lan XY, Pan CY, Chen H and Zhang CL (2007). An AluI PCR-RFLP detecting a silent allele at the goat POU1F1 locus and its association with production traits. Small Rumin. Res. 73: 8-12. http://dx.doi.org/10.1016/j.smallrumres.2006.10.009 Nagle CA, Vergnes L, Dejong H, Wang S, et al. (2008). Identification of a novel sn-glycerol-3-phosphate acyltransferase isoform, GPAT4, as the enzyme deficient in Agpat6-/- mice. J. Lipid Res. 49: 823-831. http://dx.doi.org/10.1194/jlr.M700592-JLR200 PMid:18192653    PMCid:2819352 Nei M and Roychoudhury AK (1974). Sampling variances of heterozygosity and genetic distance. Genetics 76: 379-390. PMid:4822472    PMCid:1213072 Sambrook J and Russell DW (2001). Molecular Cloning: A Laboratory Manual. 3rd edn. Cold Spring Harbor Laboratory Press, New York. Sham P, Bader JS, Craig I, O’Donovan M, et al. (2002). DNA Pooling: a tool for large-scale association studies. Nat. Rev. Genet. 3: 862-871. http://dx.doi.org/10.1038/nrg930 PMid:12415316 Sukumaran S, Barnes RI, Garg A and Agarwal AK (2009). Functional characterization of the human 1-acylglycerol- 3-phosphate-O-acyltransferase isoform 10/glycerol-3-phosphate acyltransferase isoform 3. J. Mol. Endocrinol. 42: 469-478. http://dx.doi.org/10.1677/JME-09-0010 PMid:19318427 Takeuchi K and Reue K (2009). Biochemistry, physiology, and genetics of GPAT, AGPAT, and lipin enzymes in triglyceride synthesis. Am. J. Physiol. Endocrinol. Metab. 296: E1195-E1209. http://dx.doi.org/10.1152/ajpendo.90958.2008 PMid:19336658    PMCid:2692402 Vergnes L, Beigneux AP, Davis R, Watkins SM, et al. (2006). Agpat6 deficiency causes subdermal lipodystrophy and resistance to obesity. J. Lipid Res. 47: 745-754. http://dx.doi.org/10.1194/jlr.M500553-JLR200 PMid:16436371    PMCid:2901549 Ye GM, Chen C, Huang S, Han DD, et al. (2005). Cloning and characterization a novel human 1-acyl-sn-glycerol-3- phosphate acyltransferase gene AGPAT7. DNA Seq. 16: 386-390. http://dx.doi.org/10.1080/10425170500213712 PMid:16243729
Y. H. Pang, Lei, C. Z., Zhang, C. L., Lan, X. Y., Shao, S., Gao, X. M., Wang, J. Q., and Chen, H., Lack of association of single nucleotide polymorphisms of the bovine Flt-1 gene with growth traits in Chinese cattle breeds, vol. 10, pp. 359-367, 2011.
Alitalo K, Tammela T and Petrova TV (2005). Lymphangiogenesis in development and human disease. Nature 438: 946- 953. http://dx.doi.org/10.1038/nature04480 PMid:16355212   Autiero M, Luttun A, Tjwa M and Carmeliet P (2003). Placental growth factor and its receptor, vascular endothelial growth factor receptor-1: novel targets for stimulation of ischemic tissue revascularization and inhibition of angiogenic and inflammatory disorders. J. Thromb. Haemost. 1: 1356-1370. http://dx.doi.org/10.1046/j.1538-7836.2003.00263.x PMid:12871269   Cao H, Urban JF Jr and Anderson RA (2008). Insulin increases tristetraprolin and decreases VEGF gene expression in mouse 3T3-L1 adipocytes. Obesity 16: 1208-1218. http://dx.doi.org/10.1038/oby.2008.65 PMid:18388887   De Vries C, Escobedo JA, Ueno H, Houck K, et al. (1992). The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science 255: 989-991. http://dx.doi.org/10.1126/science.1312256 PMid:1312256   Ferrara N (2004). Vascular endothelial growth factor: basic science and clinical progress. Endocrinol. Rev. 25: 581-611. http://dx.doi.org/10.1210/er.2003-0027 PMid:15294883   Ferrara N, Gerber HP and LeCouter J (2003). The biology of VEGF and its receptors. Nat. Med. 9: 669-676. http://dx.doi.org/10.1038/nm0603-669 PMid:12778165   Giantonio BJ, Catalano PJ, Meropol NJ, O'Dwyer PJ, et al. (2007). Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J. Clin. Oncol. 25: 1539-1544. http://dx.doi.org/10.1200/JCO.2006.09.6305 PMid:17442997   MacNeil MD and Grosz MD (2002). Genome-wide scans for QTL affecting carcass traits in Hereford x composite double backcross populations. J. Anim. Sci. 80: 2316-2324. PMid:12350008   Pang Y, Lei C, Zhang C, Lan X, et al. (2010). The polymorphisms of bovine VEGF gene and their associations with growth traits in Chinese cattle. Mol. Biol. Rep. DOI: 10.1007/s11033-010-0163-6. http://dx.doi.org/10.1007/s11033-010-0163-6   Peng H, Usas A, Olshanski A, Ho AM, et al. (2005). VEGF improves, whereas sFlt1 inhibits, BMP2-induced bone formation and bone healing through modulation of angiogenesis. J. Bone Miner. Res. 20: 2017-2027. http://dx.doi.org/10.1359/JBMR.050708 PMid:16234975   Quinn TP, Peters KG, de Vries C, Ferrara N, et al. (1993). Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium. Proc. Natl. Acad. Sci. U. S. A. 90: 7533-7537. http://dx.doi.org/10.1073/pnas.90.16.7533 PMid:8356051 PMCid:47176   Sambrook J and Russell DW (2001). Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, New York.   Tang K, Breen EC, Gerber HP, Ferrara NM, et al. (2004). Capillary regression in vascular endothelial growth factor-deficient skeletal muscle. Physiol. Genomics 18: 63-69. http://dx.doi.org/10.1152/physiolgenomics.00023.2004 PMid:15084712   Tebbe CC, Schmalenberger A, Peters S and Schwieger F (2001). Single Strand Conformation Polymorphism (SSCP) for Microbial Community Analysis. In: Environmental Molecular Microbiology: Protocols and Applications (Rochelle PA, ed.). Horizon Scientific Press, Wymondham, 161-175.   Yang D, Chen H, Wang X, Tian Z, et al. (2007). Association of polymorphisms of leptin gene with body weight and body sizes indexes in Chinese indigenous cattle. J. Genet. Genomics 34: 400-405. http://dx.doi.org/10.1016/S1673-8527(07)60043-5   Yang JC, Haworth L, Sherry RM, Hwu P, et al. (2003). A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N. Engl. J. Med. 349: 427-434. http://dx.doi.org/10.1056/NEJMoa021491 PMid:12890841 PMCid:2275324   Zhang C, Chen H, Zhang L, Zhao M, et al. (2008). Association of polymorphisms of the GHRHR gene with growth traits in cattle. Arch. Tierz. 51: 300-301.   Zhang Q, Chen H, Zhao S, Zhang L, et al. (2009). Single nucleotide polymorphisms and haplotypic diversity in the bovine PRKAB1 gene. Mol. Biotechnol. 43: 193-199. http://dx.doi.org/10.1007/s12033-009-9194-4 PMid:19578998
C. Y. Pan, Lan, X. Y., Zhao, H. Y., Hu, S. R., Huai, Y. T., Lei, C. Z., and Chen, H., A novel genetic variant of the goat Six6 gene and its association with production traits in Chinese goat breeds, vol. 10, pp. 3888-3900, 2011.
Betty P, Nathalie I and Jean B (2004). Efficient screening for expressed sequence tag polymorphisms (ESTPs) by DNA pool sequencing and denaturing gradient gel electrophoresis (DGGE) in spruces. Mol. Breed. 13: 263-279. http://dx.doi.org/10.1023/B:MOLB.0000022528.01656.c8   Bhattacharya TK, Chatterjee RN, Sharma RP, Niranjan M, et al. (2011). Associations between novel polymorphisms at the 5'-UTR region of the prolactin gene and egg production and quality in chickens. Theriogenology 75: 655-661. http://dx.doi.org/10.1016/j.theriogenology.2010.10.005 PMid:21111467   Cheyette BN, Green PJ, Martin K, Garren H, et al. (1994). The Drosophila sine oculis locus encodes a homeodomain-containing protein required for the development of the entire visual system. Neuron 12: 977-996. http://dx.doi.org/10.1016/0896-6273(94)90308-5   Conte I, Morcillo J and Bovolenta P (2005). Comparative analysis of Six 3 and Six 6 distribution in the developing and adult mouse brain. Dev. Dyn. 234: 718-725. http://dx.doi.org/10.1002/dvdy.20463 PMid:15973738   Diaczok D, Romero C, Zunich J, Marshall I, et al. (2008). A novel dominant negative mutation of OTX2 associated with combined pituitary hormone deficiency. J. Clin. Endocr. Metab. 93: 4351-4359. http://dx.doi.org/10.1210/jc.2008-1189 PMid:18728160 PMCid:2582563   Eller A, Branch DW, Nelson L and Silver R (2008). 247: The -634GC polymorphism in the regulatory 5ꞌ untranslated region (5' UTR) of the vascular endothelial growth factor (VEGF) gene is associated with unexplained recurrent pregnancy loss (RPL). Am. J. Obstet. Gynecol. 199 (Suppl 1): S81. http://dx.doi.org/10.1016/j.ajog.2008.09.275   Greenwood TA and Kelsoe JR (2003). Promoter and intronic variants affect the transcriptional regulation of the human dopamine transporter gene. Genomics 82: 511-520. http://dx.doi.org/10.1016/S0888-7543(03)00142-3   Gupta N, Ahlawat SPS, Kumar D, Gupta SC, et al. (2007). Single nucleotide polymorphism in growth hormone gene exon-4 and exon-5 using PCR-SSCP in Black Bengal goats - a prolific meat breed of India. Meat Sci. 76: 658-665. http://dx.doi.org/10.1016/j.meatsci.2007.02.005 PMid:22061242   Hu S, Mamedova A and Hegde RS (2008). DNA-binding and regulation mechanisms of the SIX family of retinal determination proteins. Biochemistry 47: 3586-3594. http://dx.doi.org/10.1021/bi702186s PMid:18293925   Huai YT, Lan XY, Ma L, Wang J, et al. (2011). Novel mutation in TGA stop-codon of bovine SIX6 gene. Mol. Biol. 45: 218-224. http://dx.doi.org/10.1134/S0026893310061093   Jean D, Bernier G and Gruss P (1999). Six6 (Optx2) is a novel murine Six3-related homeobox gene that demarcates the presumptive pituitary/hypothalamic axis and the ventral optic stalk. Mech. Dev. 84: 31-40. http://dx.doi.org/10.1016/S0925-4773(99)00068-4   Kim DJ, Park BL, Yoon S, Lee HK, et al. (2007). 5' UTR polymorphism of dopamine receptor D1 (DRD1) associated with severity and temperament of alcoholism. Biochem. Biophys. Res. Commun. 357: 1135-1141. http://dx.doi.org/10.1016/j.bbrc.2007.04.074 PMid:17466946   Kumar JP (2009). The sine oculis homeobox (SIX) family of transcription factors as regulators of development and disease. Cell Mol. Life Sci. 66: 565-583. http://dx.doi.org/10.1007/s00018-008-8335-4 PMid:18989625 PMCid:2716997   Lai X, Lan X, Chen H, Wang X, et al. (2009). A novel SNP of the Hesx1 gene in bovine and its associations with average daily gain. Mol. Biol. Rep. 36: 1677-1681. http://dx.doi.org/10.1007/s11033-008-9368-3 PMid:18853282   Lan XY, Pan CY, Chen H, Zhang CL, et al. (2007). An AluI PCR-RFLP detecting a silent allele at the goat POU1F1 locus and its association with production traits. Small Rumin. Res. 73: 12. http://dx.doi.org/10.1016/j.smallrumres.2006.10.009   Lan XY, Pan CY, Li JY, Guo YW, et al. (2009a). Twelve novel SNPs of the goat POU1F1 gene and their associations with cashmere traits. Small Rumin. Res. 85: 116-121. http://dx.doi.org/10.1016/j.smallrumres.2009.08.002   Lan X, Pan C, Zhang L, Zhao M, et al. (2009b). A novel missense (A79V) mutation of goat PROP1 gene and its association with production traits. Mol. Biol. Rep. 36: 2069-2073. http://dx.doi.org/10.1007/s11033-008-9418-x PMid:19031010   Lan XY, Lai X, Li ZJ, Wang J, et al. (2010) Effects of genetic variability of the caprine homeobox transcription factor HESX1 gene on performance traits. Mol. Biol. Rep. 37: 441-449. http://dx.doi.org/10.1007/s11033-009-9625-0 PMid:19629745   Lasky-Su J and Lange C (2010). Statistical challenges for genome-wide association studies of suicidality using family data. Eur. Psychiatry 25: 307-309. http://dx.doi.org/10.1016/j.eurpsy.2009.12.019 PMid:20447807 PMCid:2925169   Lawson MJ and Zhang L (2008). Housekeeping and tissue-specific genes differ in simple sequence repeats in the 5'-UTR region. Gene 407: 54-62. http://dx.doi.org/10.1016/j.gene.2007.09.017 PMid:17964742   Li MJ, Lan XY, Chen H, Zhang LZ, et al. (2008). The novel missense mutation of goat LHX4 gene. Small Rumin. Res. 79: 109-112. http://dx.doi.org/10.1016/j.smallrumres.2008.06.005   Li S, Crenshaw EB, Rawson EJ, Simmons DM, et al. (1990). Dwarf locus mutants lacking three pituitary cell types result from mutations in the POU-domain gene Pit-1. Nature 347: 528-533. http://dx.doi.org/10.1038/347528a0 PMid:1977085   Liang Y, Cui J, Yang G, Leung FC, et al. (2006). Polymorphisms of 5' flanking region of chicken prolactin gene. Domest. Anim. Endocrinol. 30: 1-16. http://dx.doi.org/10.1016/j.domaniend.2005.05.006 PMid:15970423   Medeiros-Neto G, de Lacerda L and Wondisford FE (1997). Familial Congenital Hypothyroidism Caused by Abnormal and Bioinactive TSH due to Mutations in the beta-Subunit Gene. Trends Endocrinol. Metab. 8: 15-20. http://dx.doi.org/10.1016/S1043-2760(96)00203-2   Nei M and Li WH (1979). Mathematical model for studying genetic variation in terms of restriction endonucleases. Proc. Natl. Acad. Sci. U. S. A. 76: 5269-5273. http://dx.doi.org/10.1073/pnas.76.10.5269 PMid:291943 PMCid:413122   Pedersen LD, Sorensen AC and Berg P (2009). Marker-assisted selection can reduce true as well as pedigree-estimated inbreeding. J. Dairy Sci. 92: 2214-2223. http://dx.doi.org/10.3168/jds.2008-1616 PMid:19389980   Petrie JR, Pearson ER and Sutherland C (2011). Implications of genome wide association studies for the understanding of type 2 diabetes pathophysiology. Biochem. Pharmacol. 81: 471-477. http://dx.doi.org/10.1016/j.bcp.2010.11.010 PMid:21111713   Pfäffle R and Klammt J (2011). Pituitary transcription factors in the aetiology of combined pituitary hormone deficiency. Best. Pract. Res. Clin. Endocrinol. Metab. 25: 43-60. http://dx.doi.org/10.1016/j.beem.2010.10.014 PMid:21396574   Rachel L, Daniel DC, Nichol LGM and Mellon PL (2011). Hypothalamic dysregulation and infertility in mice lacking the homeodomain protein Six6. J. Neurosci. 31: 426-438. http://dx.doi.org/10.1523/JNEUROSCI.1688-10.2011 PMid:21228153 PMCid:3103738   Sahana G, Guldbrandtsen B and Lund MS (2011). Genome-wide association study for calving traits in Danish and Swedish Holstein cattle. J. Dairy Sci. 94: 479-486. http://dx.doi.org/10.3168/jds.2010-3381 PMid:21183059   Sambrook J and Russell DW (2001). Molecular Cloning: A Laboratory Manual. 3rd edn. Cold Spring Harbor Laboratory Press, New York.   Sham P, Bader JS, Craig I, O'Donovan M, et al. (2002). DNA Pooling: a tool for large-scale association studies. Nat. Rev. Genet. 3: 862-871. http://dx.doi.org/10.1038/nrg930 PMid:12415316   Sobrier ML, Maghnie M, Vie-Luton MP, Secco A, et al. (2006). Novel HESX1 mutations associated with a life-threatening neonatal phenotype, pituitary aplasia, but normally located posterior pituitary and no optic nerve abnormalities. Clin. Endocrinol. Metab. 91: 4528-4536. http://dx.doi.org/10.1210/jc.2006-0426 PMid:16940453   Tétreault N, Champagne MP and Bernier G (2009). The LIM homeobox transcription factor Lhx2 is required to specify the retina field and synergistically cooperates with Pax6 for Six6 trans-activation. Dev. Biol. 327: 541-550. http://dx.doi.org/10.1016/j.ydbio.2008.12.022 PMid:19146846   Thomas MG, Enns RM, Shirley KL, Garcia MD, et al. (2007). Associations of DNA polymorphisms in growth hormone and its transcriptional regulators with growth and carcass traits in two populations of Brangus bulls. Genet. Mol. Res. 6: 222-237. PMid:17469072   Wang DY, Zhang YJ, Liu YQ and Yang YZ (2009). Karyotype and single nucleotide polymorphism of the prolactin gene in milking bucks. Small Rumin. Res. 87: 96-101. http://dx.doi.org/10.1016/j.smallrumres.2009.09.032   Wang GB, Li CR, Yang J, Wen PQ, et al. (2011). A regulatory polymorphism in promoter region of TNFR1 gene is associated with Kawasaki disease in Chinese individuals. Hum. Immunol. 72: 451-457. http://dx.doi.org/10.1016/j.humimm.2011.02.004 PMid:21315128   Weasner BP and Kumar JP (2009). The non-conserved C-terminal segments of Sine Oculis Homeobox (SIX) proteins confer functional specificity. Genesis 47: 514-523. http://dx.doi.org/10.1002/dvg.20517 PMid:19422020 PMCid:2738589   Weller JI and Ron M (2011). Invited review: quantitative trait nucleotide determination in the era of genomic selection. J. Dairy Sci. 94: 1082-1090. http://dx.doi.org/10.3168/jds.2010-3793 PMid:21338774
2010
B. L. Guo, Jiao, Y., He, C., Wei, L. X., Chang, Z. H., Yue, X. P., Lan, X. Y., Chen, H., and Lei, C. Z., A novel polymorphism of the lactoferrin gene and its association with milk composition and body traits in dairy goats, vol. 9, pp. 2199-2206, 2010.
Brandl N, Zemann A, Kaupe I, Marlovits S, et al. (2010). Signal transduction and metabolism in chondrocytes is modulated by lactoferrin. Osteoarthritis Cartilage 18: 117-125. http://dx.doi.org/10.1016/j.joca.2009.08.012 PMid:19747587   Bullen JJ (1972). Iron-binding proteins in milk and resistance to Escherichia coli infection in infants. Proc. R. Soc. Med. 65: 1086. PMid:4568537 PMCid:1644425   Cohen MS, Britigan BE, French M and Bean K (1987). Preliminary observations on lactoferrin secretion in human vaginal mucus: variation during the menstrual cycle, evidence of hormonal regulation, and implications for infection with Neisseria gonorrhoeae. Am. J. Obstet. Gynecol. 157: 1122-1125. PMid:3120589   Cornish J (2004). Lactoferrin promotes bone growth. Biometals 17: 331-335. http://dx.doi.org/10.1023/B:BIOM.0000027713.18694.91 PMid:15222486   Cornish J, Grey AB, Naot D and Palmano KP (2005). Lactoferrin and bone: an overview of recent progress. Aust. J. Dairy Technol. 60: 53-57.   Gutteridge JM, Paterson SK, Segal AW and Halliwell B (1981). Inhibition of lipid peroxidation by the iron-binding protein lactoferrin. Biochem. J. 199: 259-261. PMid:7337708 PMCid:1163360   Jenssen H and Hancock RE (2009). Antimicrobial properties of lactoferrin. Biochimie 91: 19-29. http://dx.doi.org/10.1016/j.biochi.2008.05.015 PMid:18573312   Jeremy B (1995). Lactoferrin: a multifunctional immunoregulatory protein? Immunol. Today 16: 417-419. http://dx.doi.org/10.1016/0167-5699(95)80016-6   Kim SJ, Sohn BH, Jeong S, Pak KW, et al. (1999). High-level expression of human lactoferrin in milk of transgenic mice using genomic lactoferrin sequence. J. Biochem. 126: 320-325. http://dx.doi.org/10.1093/oxfordjournals.jbchem.a022452 PMid:10423524   Kinsella JE and Whitehead DM (1989). Proteins in whey: chemical, physical, and functional properties. Adv. Food Nutr. Res. 33: 343-438. http://dx.doi.org/10.1016/S1043-4526(08)60130-8   Lan XY, Pan CY, Chen H and Zhang CL (2007). An AluI PCR-RFLP detecting a silent allele at the goat POU1F1 locus and its association with production traits. Small Ruminant Res. 73: 8-12. http://dx.doi.org/10.1016/j.smallrumres.2006.10.009   Leon-Sicairos N, Canizalez-Roman A, de la Garza M, Reyes-Lopez M, et al. (2009). Bactericidal effect of lactoferrin and lactoferrin chimera against halophilic Vibrio parahaemolyticus. Biochimie 91: 133-140. http://dx.doi.org/10.1016/j.biochi.2008.06.009 PMid:18625283   Li GH, Zhang Y, Sun DX and Li N (2004). Study on the polymorphism of bovine lactoferrin gene and its relationship with mastitis. Anim. Biotechnol. 15: 67-76. http://dx.doi.org/10.1081/ABIO-120037899 PMid:15248601   Liu LH, Gladwell W and Teng CT (2002). Detection of exon polymorphisms in the human lactoferrin gene. Biochem. Cell Biol. 80: 17-22. http://dx.doi.org/10.1139/o01-207 PMid:11908638   Livney YD (2010). Milk proteins as vehicles for bioactives. Curr. Opin. Colloid Interface Sci. 15: 73-83. http://dx.doi.org/10.1016/j.cocis.2009.11.002   Masson PL, Heremans JF and Dive CH (1966). An iron-binding protein common to many external secretions. Clin. Chim. Acta 14: 735-739. http://dx.doi.org/10.1016/0009-8981(66)90004-0   Mohamed JA, DuPont HL, Jiang ZD, Belkind-Gerson J, et al. (2007). A novel single-nucleotide polymorphism in the lactoferrin gene is associated with susceptibility to diarrhea in North American travelers to Mexico. Clin. Infect. Dis. 44: 945-952. http://dx.doi.org/10.1086/512199 PMid:17342646   Nei M and Roychoudhury AK (1974). Sampling variances of heterozygosity and genetic distance. Genetics 76: 379-390. PMid:4822472 PMCid:1213072   Nei M and Li WH (1979). Mathematical model for studying genetic variation in terms of restriction endonucleases. Proc. Natl. Acad. Sci. U. S. A. 76: 5269-5273. http://dx.doi.org/10.1073/pnas.76.10.5269 PMid:291943 PMCid:413122   Nichols BL, McKee KS, Henry JF and Putman M (1987). Human lactoferrin stimulates thymidine incorporation into DNA of rat crypt cells. Pediatr. Res. 21: 563-567. http://dx.doi.org/10.1203/00006450-198706000-00011 PMid:3496579   Park I, Schaeffer E, Sidoli A, Baralle FE, et al. (1985). Organization of the human transferrin gene: direct evidence that it originated by gene duplication. Proc. Natl. Acad. Sci. U. S. A. 82: 3149-3153. http://dx.doi.org/10.1073/pnas.82.10.3149 PMid:3858812 PMCid:397732   Teng CT, Pentecost BT, Marshall A, Solomon A, et al. (1987). Assignment of the lactotransferrin gene to human chromosome 3 and to mouse chromosome 9. Somat. Cell Mol. Genet. 13: 689-693. http://dx.doi.org/10.1007/BF01534490 PMid:3478818   Teng CT, Pentecost BT, Chen YH, Newbold RR, et al. (1989). Lactotransferrin gene expression in the mouse uterus and mammary gland. Endocrinology 124: 992-999. http://dx.doi.org/10.1210/endo-124-2-992 PMid:2463910   Williams J (1982). The evolution of transferrin. Trends Biochem. Sci. 7: 394-397. http://dx.doi.org/10.1016/0968-0004(82)90183-9   Yamauchi K, Tomita M, Giehl TJ and Ellison RT III (1993). Antibacterial activity of lactoferrin and a pepsin-derived lactoferrin peptide fragment. Infect. Immun. 61: 719-728. PMid:8423097 PMCid:302785   Yamauchi K, Wakabayashi H, Shin K and Takase M (2006). Bovine lactoferrin: benefits and mechanism of action against infections. Biochem. Cell Biol. 84: 291-296. http://dx.doi.org/10.1139/o06-054 PMid:16936799