Publications

Found 108 results
Filters: Author is J. Wang  [Clear All Filters]
2015
F. P. Wu, Wang, J., Wang, H., Liu, Y. J., Li, N., and Liu, Z. K., Chest wall recurrence location and the lower-bound target of preventive radiotherapy after transverse incision in modified radical mastectomy, vol. 14, pp. 1068-1075, 2015.
X. - G. Li, Wang, J., and Yu, Z. - Y., Cloning of an anthocyanidin synthase gene homolog from blackcurrant (Ribes nigrum L.) and its expression at different fruit stages, vol. 14, pp. 2726-2734, 2015.
Y. Zhao, Wang, J., Thammaratsuntorn, J., Wu, J. W., Wei, J. H., Wang, Y., Xu, J. W., and Zhao, J. L., Comparative transcriptome analysis of Nile tilapia (Oreochromis niloticus) in response to alkalinity stress, vol. 14, pp. 17916-17926, 2015.
J. Ge, Wang, J., Zhang, F., Diao, B., Song, Z. F., Shan, L. L., Wang, W., Cao, H. J., and Li, X. Q., Correlation between MTHFR gene methylation and pre-eclampsia, and its clinical significance, vol. 14, pp. 8021-8028, 2015.
X. W. Kang, Hu, J. L., Wang, S. K., and Wang, J., Effectiveness of muscle basal lamina carrying neural stem cells and olfactory ensheathing cells in spinal cord repair, vol. 14, pp. 13437-13455, 2015.
W. - C. Li, Zhang, L., Wang, J., Wang, D., Wang, T. - X., and Duan, C. - X., Effects of silicon on seed setting rate of rice intersubspecific hybrids, vol. 14, pp. 10359-10364, 2015.
J. Wang, Wang, T., Yin, G. - Y., Yang, L., Wang, Z. - G., and Bu, X. - B., Glutathione S-transferase polymorphisms influence chemotherapy response and treatment outcome in breast cancer, vol. 14, pp. 11126-11132, 2015.
H. W. Jiang, Wang, J., Li, H. J., Peng, J. K., Gao, X. P., and Chen, F., Influence of the DCC gene on proliferation and carcinoembryonic antigen expression in the human colorectal cancer cell line SW1116, vol. 14, pp. 10273-10280, 2015.
J. Wang, Zou, T., Yang, H. X., Gong, Y. Z., Xie, X. J., Liu, H. Y., and Liao, D. F., Insulin receptor binding motif tagged with IgG4 Fc (Yiminsu) works as an insulin sensitizer to activate Akt signaling in hepatocytes, vol. 14, pp. 8819-8828, 2015.
J. Y. Zhong, Zheng, X. W., Ye, H. D., Cui, H. B., Du, W. P., Zhang, Z. X., Fei, X. H., Lin, S. Y., Wang, J., Su, J., Chen, X. M., and Duan, S. W., Male-specific association of the APC rs383830 T allele with the risk of coronary heart disease, vol. 14, pp. 11745-11751, 2015.
J. Wang, Cao, H., Hong, X., Chen, G. H., Fan, H. M., Li, Q. C., Liu, Z. M., and Li, K. F., MicroRNA screening and functional study of obliterative bronchiolitis in a rat model simulating lung transplantation, vol. 14, pp. 19309-19316, 2015.
E. L. Wang, Wang, K. Y., Chen, D. F., Geng, Y., Huang, L. Y., Wang, J., and He, Y., Molecular cloning and bioinformatic analysis of the Streptococcus agalactiae neuA gene isolated from tilapia, vol. 14, pp. 6003-6017, 2015.
D. Hu, Song, X., Wang, N., Zhong, X., Wang, J., Liu, T., Jiang, Z., Dawa, T., Gu, X., Peng, X., and Yang, G., Molecular identification of Echinococcus granulosus on the Tibetan Plateau using mitochondrial DNA markers, vol. 14, pp. 13915-13923, 2015.
W. C. Li, Wang, J., Sun, Y. L., Ji, S. D., and Guo, S. W., Morphology and photosynthetic enzyme activity of maize phosphoenolpyruvate carboxylase transgenic rice, vol. 14, pp. 15572-15576, 2015.
X. B. Gao, Xiao, M., Wang, J., Liu, Y. J., Liu, Q. Z., and Qi, M. L., Optimization of candidate proteins for serological screening of Chlamydia trachomatis infection, vol. 14, pp. 12240-12246, 2015.
J. Wang, Yang, S., Guo, F. H., Mao, X., Zhou, H., Dong, Y. Q., Wang, Z. M., and Luo, F., Quantitative assessment of the association between the angiotensin-converting enzyme gene insertion/deletion polymorphism and digestive system cancer risk, vol. 14, pp. 14291-14299, 2015.
L. Yang, Wang, J., Li, F. - G., Han, M., Chang, X. - J., and Wang, Z. - T., Relationship between genetic polymorphisms in MCP-1, CCR-2, and non-small-cell lung cancer in the Han nationality of Northern China, vol. 14, pp. 3745-3752, 2015.
F. Fang, Wang, J., Pan, J., Su, G. H., Xu, L. X., and Li, G., Relationship between vitamin D (1,25-dihydroxyvitamin D3) receptor gene polymorphisms and primary biliary cirrhosis risk: a meta-analysis, vol. 14, pp. 981-988, 2015.
L. Zhang, Liu, R. X., Wang, J., Zhang, T., Li, J., Shi, J. H., Kang, B. Y., and Chen, S. Q., A SCAR marker for the analysis of chloroplast DNA from different cultivars of Cornus officinalis, vol. 14, pp. 17170-17181, 2015.
J. Wang, Sun, G. J., Ding, J., Zhang, J. X., Cui, Y., Li, H. R., and Wang, S. J., WY14643 combined with all-trans retinoic acid acts via p38 MAPK to induce “browning” of white adipocytes in mice, vol. 14, pp. 6978-6984, 2015.
2014
J. J. Zhu, Liu, X. F., Zhang, P. L., Yang, J. Z., Wang, J., Qin, Y., Zhang, G. L., Ren, D. Q., Cui, C. L., and Guo, X. G., Anatomical information for intercostobrachial nerve preservation in axillary lymph node dissection for breast cancer, vol. 13, pp. 9315-9323, 2014.
J. Wang, Simayi, M., Wushouer, Q., Xia, Y., He, Y., Yan, F., Zhang, J., Cui, S., and Wen, H., Association between polymorphisms in ADAM33, CD14, and TLR4 with asthma in the Uygur population in China, vol. 13, pp. 4680-4690, 2014.
Y. H. Pang, Zhao, J. X., Du, W. L., Li, Y. L., Wang, J., Wang, L. M., Wu, J., Cheng, X. N., Yang, Q. H., and Chen, X. H., Cytogenetic and molecular identification of a wheat-Leymus mollis alien multiple substitution line from octoploid Tritileymus x Triticum durum, vol. 13, pp. 3903-3913, 2014.
Y. H. Feng, Yang, Z. Q., Wang, J., Luo, Q. F., and Li, H. G., Development and characterization of SSR markers from Pinus massoniana and their transferability to P. elliottii, P. caribaea and P. yunnanensis, vol. 13, pp. 1508-1513, 2014.
J. Wang, Liu, Y., Dong, W. H., Huo, Y. J., Huang, X. G., Wu, S. L., and Bao, W. B., Dynamic changes in TAP1 expression levels in newborn to weaning piglets, and its association with Escherichia coli F18 resistance, vol. 13, pp. 3686-3692, 2014.
C. Wu, Wang, T., Wang, J., Qu, B., Wang, H., and Hu, Y., Effect of radiotherapy on the treatment of patients with extensive stage small cell lung cancer, vol. 13, pp. 8577-8585, 2014.
J. Wang, Xu, J., Wang, B., Shu, F. R., Chen, K., and Mi, M. T., Equol promotes rat osteoblast proliferation and differentiation through activating estrogen receptor, vol. 13, pp. 5055-5063, 2014.
J. L. Jia, Zhang, L. P., Wu, J. P., Wang, J., and Ding, Q., Establishment of the optimum two-dimensional electrophoresis system of ovine ovarian tissue, vol. 13, pp. 6528-6538, 2014.
Y. P. Feng, Chen, J. F., Huang, P., Wang, X., Wang, J., Peng, X. L., and Gong, Y. Z., Expression analysis of differentially expressed miRNAs in male and female chicken embryos, vol. 13, pp. 3060-3068, 2014.
X. L. Yang, Tian, J., Liang, Y., Ma, C. J., Yang, A. N., Wang, J., Ma, S. C., Cheng, Y., Hua, X., and Jiang, Y. D., Homocysteine induces blood vessel global hypomethylation mediated by LOX-1, vol. 13, pp. 3787-3799, 2014.
Y. Geng, Wang, J., Jing, H., Wang, H. W., and Bao, Y. X., Inhibitory effect of dexamethasone on Lewis mice lung cancer cells, vol. 13, pp. 6827-6836, 2014.
J. H. Hu, Zhang, F. Y., Jiang, K. J., Fang, Y. B., Wang, J., Zhao, M., Qiao, Z. G., and Ma, L. B., Molecular characterization of thioredoxin-1 and thioredoxin reductase activity in mud crab Scylla paramamosain, vol. 13, pp. 10241-10255, 2014.
L. L. Zheng, Gao, Z., Wang, J., Zhang, H. R., and Wang, Y. C., Molecular cloning and functional characterization of a novel CBL-interacting protein kinase NtCIPK2 in the halophyte Nitraria tangutorum, vol. 13, pp. 4716-4728, 2014.
J. Wang, A new algorithm to construct phylogenetic networks from trees, vol. 13, pp. 1456-1464, 2014.
J. Wang, Feng, Y. - P., Zuo, B., Xiong, Y. - Z., and Deng, C. - Y., Transcriptional analysis of the porcine TTID gene and association of different TTID genotypes with carcass traits, vol. 13, pp. 1195-1202, 2014.
2013
Y. D. Li, Li, J. X., Tang, B. - P., Gan, T. Y., Xu, G. J., Zhou, X. H., Li, H., Guo, X., Mahemuti, A., Sun, Q., Zhang, Y. Y., and Wang, J., Expression of tyrosine hydroxylase and growth-associated protein 43 in aging atrial fibrillation patients of Xinjiang Uygur and Han nationality, vol. 12, pp. 5257-5266, 2013.
L. - F. Xu, Wang, J., Lv, F. B., and Song, Q., Functions of microRNA in response to cocaine stimulation, vol. 12, pp. 6160-6167, 2013.
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
J. Wang, Zhang, W., Zhao, H., Li, F. R., Wang, Z. G., Ji, J., Zhang, X. Q., Wang, D. W., and Li, J. M., Molecular cytogenetic characterization of the Aegilops biuncialis karyotype, vol. 12. pp. 683-692, 2013.
Badaeva ED (2002). Evaluation of phylogenetic relationships between five polyploid Aegilops L. species of the U-genome cluster by means of chromosomal analysis. Genetika 38: 799-811. PMid:12138779   Badaeva ED, Amosova AV, Samatadze TE, Zoshchuk SA, et al. (2004). Genome differentiation in Aegilops. 4. Evolution of the U-genome cluster. Plant Syst. Evol. 246: 45-76. http://dx.doi.org/10.1007/s00606-003-0072-4   Bedbrook JR, Jones J, O'Dell M, Thompson RD, et al. (1980). A molecular description of telometic heterochromatin in secale species. Cell 19: 545-560. http://dx.doi.org/10.1016/0092-8674(80)90529-2   Dhaliwal HS, Harjit-Singh and William M (2002). Transfer of rust resistance from Aegilops ovata into bread wheat (Triticum aestivum L.) and molecular characterisation of resistant derivatives. Euphytica 126: 153-159. http://dx.doi.org/10.1023/A:1016312723040   Friebe B and Heun M (1989). C-banding pattern and powdery mildew resistance of Triticum ovatum and four T. aestivum - T. ovatum chromosome addition lines. Theor. Appl. Genet. 78: 417-424. http://dx.doi.org/10.1007/BF00265306   Friebe B, Mukai Y and Gill BS (1992a). C-banding polymorphisms in several accessions of Triticum tauschii (Aegilops squarrosa). Genome 35: 192-199. http://dx.doi.org/10.1139/g92-030   Friebe B, Schubert V, Blüthner W and Hammer K (1992b). C-banding pattern and polymorphism of Aegilops caudata and chromosomal constitutions of the amphiploid T. aestivum - Ae. caudata and six derived chromosome addition lines. Theor. Appl. Genet. 83: 589-596. http://dx.doi.org/10.1007/BF00226902   Friebe B, Jiang J, Tuleen N and Gill BS (1995). Standard karyotype of Triticum umbellulatum and the characterization of derived chromosome addition and translocation lines in common wheat. Theor. Appl. Genet. 90: 150-156. http://dx.doi.org/10.1007/BF00221010   Friebe B, Badaeva ED, Kammer K and Gill BS (1996). Standard karyotypes of Aegilops uniaristata, Ae. mutica, Ae. comosa subspecies comosa and heldreichii (Poaceae). Plant Syst. Evol. 202: 199-210. http://dx.doi.org/10.1007/BF00983382   Friebe B, Qi LL, Nasuda S, Zhang P, et al. (2000). Development of a complete set of Triticum aestivum-Aegilops speltoides chromosome addition lines. Theor. Appl. Genet. 101: 51-58. http://dx.doi.org/10.1007/s001220051448   Gerlach WL and Bedbrook JR (1979). Cloning and characterization of ribosomal RNA genes from wheat and barley. Nucleic Acids Res. 7: 1869-1885. http://dx.doi.org/10.1093/nar/7.7.1869 PMid:537913 PMCid:342353   Gerlach WL and Dyer TA (1980). Sequence organization of the repeating units in the nucleus of wheat which contain 5S rRNA genes. Nucleic Acids Res. 8: 4851-4865. http://dx.doi.org/10.1093/nar/8.21.4851 PMid:7443527 PMCid:324264   Gill BS and Kimber G (1974). Giemsa C-banding and the evolution of wheat. Proc. Natl. Acad. Sci. U. S. A. 71: 4086- 4090. http://dx.doi.org/10.1073/pnas.71.10.4086 PMid:16592188 PMCid:434333   Makkouk K, Ghulam W and Comeau A (1994). Resistance to barley yellow dwarf luteovirus in Aegilops species. Can. J. Plant Sci. 74: 631-634. http://dx.doi.org/10.4141/cjps94-113   McIntyre CL, Pereira S, Moran LB and Appels R (1990). New secale cereale (rye) DNA derivatives for the detection of rye chromosome segments in wheat. Genome 33: 635-640. http://dx.doi.org/10.1139/g90-094 PMid:2262137   Molnár I, Gáspár L, Sárvári É, Dulai S, et al. (2004). Physiological and morphological responses to water stress in Aegilops biuncialis and Triticum aestivum genotypes with differing tolerance to drought. Funct. Plant Biol. 31: 1149-1159. http://dx.doi.org/10.1071/FP03143   Mukai Y, Nakahara Y and Yamamoto M (1993). Simultaneous discrimination of the three genomes in hexaploid wheat by multicolor fluorescence in situ hybridization using total genomic and highly repeated DNA probes. Genome 36: 489-494. http://dx.doi.org/10.1139/g93-067 PMid:18470003   Nagy ED, Molnar-Lang M, Linc G and Lang L (2002). Identification of wheat-barley translocations by sequential GISH and two-colour FISH in combination with the use of genetically mapped barley SSR markers. Genome 45: 1238- 1247. http://dx.doi.org/10.1139/g02-068 PMid:12502270   Rayburn AL and Gill BS (1986). Isolation of a D-genome specific repeated DNA sequence from Aegilops squarrosa. Plant Mol. Biol. Rep. 4: 102-109. http://dx.doi.org/10.1007/BF02732107   Resta P, Zhang HB, Dubcovsky J and Dvorak J (1996). The origins of the genomes of Triticum biunciale, T. ovatum, T. neglectum, T. columnare, and T. rectum (Poaceae) based on variation in repeated nucleotide sequences. Am. J. Bot. 83: 1556-1565. http://dx.doi.org/10.2307/2445829   Riley R, Chapman V and Johnson R (1968). Introduction of yellow rust resistance of Aegilops comosa into wheat by genetically induced homoeologous recombination. Nature 217: 383-384. http://dx.doi.org/10.1038/217383a0   Schneider A, Linc G, Molnar I and Molnar-Lang M (2005). Molecular cytogenetic characterization of Aegilops biuncialis and its use for the identification of 5 derived wheat - Aegilops biuncialis disomic addition lines. Genome 48: 1070- 1082. http://dx.doi.org/10.1139/g05-062 PMid:16391676   van Slageren MWSJ (1994). Wild Wheats: A Monograph of Aegilops L. and Amblyopyrum (Jaub. & Spach) Eig (Poaceae): A Revision of All Taxa Closely Related to Wheat, Excluding Wild Triticum Species, with Notes on Other Genera in the Tribe Triticcae, Especially Triticum: Wageningen Agricultural University, Wageningen.   Wang ZG, An TG, Li JM, Marta ML, et al. (2004). Fluorescent in situ hybridization analysis of rye chromatin in the background of "Xiaoyan No. 6". Acta Bot. Sin. 46: 436-442.
W. L. Du, Wang, J., Wang, L. M., Pang, Y. H., Wu, J., Zhao, J. X., Yang, Q. H., and Chen, X. H., A novel SCAR marker for detecting Psathyrostachys huashanica Keng chromatin introduced in wheat, vol. 12, pp. 4797-4806, 2013.
W. He, Zhao, S., Liu, X., Dong, S., Lv, J., Liu, D., Wang, J., and Meng, Z., ReSeqTools: an integrated toolkit for large-scale next-generation sequencing based resequencing analysis, vol. 12, pp. 6275-6283, 2013.
J. Wang, Zhou, X., Zhao, J., Li, Z., and Li, X., Screening for feature genes associated with hereditary hemochromatosis and functional analysis with DNA microarrays, vol. 12, pp. 6240-6248, 2013.
J. Wang, Pan, Z. Y., Zheng, X. R., Wu, Z. C., Su, X. M., Zhu, G. Q., Huang, X. G., Wu, S. L., and Bao, W. B., TLR4 gene expression in pig populations and its association with resistance to Escherichia coli F18, vol. 12, pp. 2625-2632, 2013.
2012
J. Wang, Guo, M. - Z., and Xing, L. L., FastJoin, an improved neighbor-joining algorithm, vol. 11. pp. 1909-1922, 2012.
Elias I and Lagergren J (2005). Fast Neighbor Joining. In: Proceedings of the 32nd International Colloquium: 11-15 July 2005 (L Caires, GF Italiano, L Monteiro, C Palamidessi, et al., eds.). Springer Berlin Heidelberg, Lisbon, 1263-1274.   Elias I and Lagergren J (2009). Fast neighbor joining. Theor. Comput. 410: 1993-2000. http://dx.doi.org/10.1016/j.tcs.2008.12.040   Evans J, Sheneman L and Foster J (2006). Relaxed neighbor joining: a fast distance-based phylogenetic tree construction method. J. Mol. Evol. 62: 785-792. http://dx.doi.org/10.1007/s00239-005-0176-2 PMid:16752216   Finn RD, Mistry J, Schuster-Bockler B, Griffiths-Jones S, et al. (2006). Pfam: clans, web tools and services. Nucleic Acids Res. 34: D247-D251. http://dx.doi.org/10.1093/nar/gkj149 PMid:16381856 PMCid:1347511   Howe K, Bateman A and Durbin R (2002). QuickTree: building huge neighbor-joining trees of protein sequences. Bioinformatics 18: 1546-1547. http://dx.doi.org/10.1093/bioinformatics/18.11.1546 PMid:12424131   Mailund T and Pedersen CNS (2004). QuickJoin - fast neighbor-joining tree reconstruction. Bioinformatics 20: 3261- 3262. http://dx.doi.org/10.1093/bioinformatics/bth359 PMid:15201185   Mailund T, Brodal GS, Fagerberg R, Pedersen CNS, et al. (2006). Recrafting the neighbor-joining method. BMC Bioinformatics 7: 29. http://dx.doi.org/10.1186/1471-2105-7-29 PMid:16423304 PMCid:3271233   Nakhleh L, Moret BME, Roshan U and John KS (2002). The Accuracy of Fast Phylogenetic Methods for Large Datasets. In: Proceedings of the Seventh Pacific Symposium on Biocomputing: 2001 (Altman RB, Dunker AK, Hunter L, Lauderdale K, et al., eds.). World Scientific, Singapore, 211-222. PMid:11928477   Price MN, Dehal PS and Arkin AP (2009). FastTree: computing large minimum evolution trees with profiles instead of a distance matrix. Mol. Biol. Evol. 26: 1641-1650. http://dx.doi.org/10.1093/molbev/msp077 PMid:19377059 PMCid:2693737   Price MN, Dehal PS and Arkin AP (2010). FastTree 2-approximately maximum-likelihood trees for large alignments. PLoS One 5: e9490. http://dx.doi.org/10.1371/journal.pone.0009490 PMid:20224823 PMCid:2835736   Saitou N and Nei M (1987). The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4: 406-425. PMid:3447015   Sheneman L, Evans J and Foster JA (2006). Clearcut: a fast implementation of relaxed neighbor joining. Bioinformatics 22: 2823-2824. http://dx.doi.org/10.1093/bioinformatics/btl478 PMid:16982706   Simonsen M, Mailund T and Pedersen CNS (2008). Rapid Neighbor-Joining. In: Proceedings of the Eighth International Workshop on Algorithms in Bioinformatics: 15-19 September 2008 (Crandall KA and Lagergren J, eds.). Springer Berlin Heidelberg, Karlsruhe, 113-122. http://dx.doi.org/10.1007/978-3-540-87361-7_10   Simonsen M, Mailund T and Pedersen CNS (2011). Inference of Large Phylogenies Using Neighbor-Joining. In: Proceedings of the Third International Joint Conference: 20-23 January 2010 (Fred A, Filipe J and Gamboa H, eds.). Springer Berlin Heidelberg, Valencia, 334-344. PMCid:3224425   Studier JA and Keppler KJ (1988). A note on the neighbor-joining algorithm of Saitou and Nei. Mol. Biol. Evol. 5: 729- 731. PMid:3221794   Wheeler TJ (2009). Large-Scale Neighbor-Joining with NINJA. In: Proceedings of the Ninth International Workshop on Algorithms in Bioinformatics: 12-13 September 2009 (Salzberg SL and Warnow T, eds.). Springer Berlin Heidelberg, Philadelphia, 375-389.   Zaslavsky L and Tatusova T (2008). Accelerating the Neighbor-Joining Algorithm Using the Adaptive Bucket Data Structure. In: Proceedings of the Fourth International Symposium: 6-9 May 2008 (Mandoiu I, Sunderraman R and Zelikovsky A, eds.). Springer Berlin Heidelberg, Atlanta, 122-133.
X. Chen, Su, Y. Q., Wang, J., Liu, M., Niu, S. F., Zhong, S. P., and Qiu, F., Isolation and identification of the immune-relevant ribosomal protein L10 (RPL10/QM-like gene) from the large yellow croaker Pseudosciaena crocea (Pisces: Sciaenidae), vol. 11, pp. 3755-3765, 2012.
Chavez-Rios R and Vargas-Mejia M (2000). Isolation and identification of L10 gene from Entamoeba histolytica homologous to Wilms' tumor suppressor. Arch. Med. Res. 31: S305-S306. http://dx.doi.org/10.1016/S0188-4409(00)00118-1   Chen X, Wu CW, Zhong SP, Zeng FR, et al. (2011). Molecular characterization and structure analysis of RPL10/QM-like protein from the red drum Sciaenops ocellatus (Sciaenidae). Genet. Mol. Res. 10: 576-587. http://dx.doi.org/10.4238/vol10-2gmr1134 PMid:21491368   Chou MY, Hsiao CD, Chen SC, Chen IW, et al. (2008). Effects of hypothermia on gene expression in zebrafish gills: upregulation in differentiation and function of ionocytes as compensatory responses. J. Exp. Biol. 211: 3077-3084. http://dx.doi.org/10.1242/jeb.019950 PMid:18805806   dos Santos NM, Taverne-Thiele JJ, Barnes AC, van Muiswinkel WB, et al. (2001). The gill is a major organ for antibody secreting cell production following direct immersion of sea bass (Dicentrarchus labrax, L.) in a Photobacterium damselae ssp. piscicida bacterin: an ontogenetic study. Fish Shellfish Immunol. 11: 65-74. http://dx.doi.org/10.1006/fsim.2000.0295 PMid:11271603   Dowdy SF, Lai KM, Weissman BE, Matsui Y, et al. (1991). The isolation and characterization of a novel cDNA demonstrating an altered mRNA level in nontumorigenic Wilms' microcell hybrid cells. Nucleic Acids Res. 19: 5763-5769. http://dx.doi.org/10.1093/nar/19.20.5763 PMid:1658743 PMCid:328988   Eisinger DP, Dick FA and Trumpower BL (1997). Qsr1p, a 60S ribosomal subunit protein, is required for joining of 40S and 60S subunits. Mol. Cell Biol. 17: 5136-5145. PMid:9271391 PMCid:232364   Green H, Canfield AE, Hillarby MC, Grant ME, et al. (2000). The ribosomal protein QM is expressed differentially during vertebrate endochondral bone development. J. Bone Miner. Res. 15: 1066-1075. http://dx.doi.org/10.1359/jbmr.2000.15.6.1066 PMid:10841175   Haugarvoll E, Bjerkas I, Nowak BF, Hordvik I, et al. (2008). Identification and characterization of a novel intraepithelial lymphoid tissue in the gills of Atlantic salmon. J. Anat. 213: 202-209. http://dx.doi.org/10.1111/j.1469-7580.2008.00943.x PMid:19172734 PMCid:2526113   Hwang JS, Goo TW, Yun EY, Lee JH, et al. (2000). Tissue-/stage-dependent expression of a cloned Bombyx mandarina QM homologue. Biomol. Eng. 16: 211-215. http://dx.doi.org/10.1016/S1389-0344(00)00056-3   Jin S, Cai WQ and Wang GL (2002). Studies on the pathogenic bacteria diseases of cultured Pseudosciaena crocea. J. Zhejiang Ocean Univ. 21: 225-230.   Kaneko K, Kobayashi H, Onodera O, Miyatake T, et al. (1992). Genomic organization of a cDNA (QM) demonstrating an altered mRNA level in nontumorigenic Wilms' microcell hybrid cells and its localization to Xq28. Hum. Mol. Genet. 1: 529-533. http://dx.doi.org/10.1093/hmg/1.7.529 PMid:1339145   Koppang EO, Hordvik I, Bjerkas I, Torvund J, et al. (2003). Production of rabbit antisera against recombinant MHC class II β chain and identification of immunoreactive cells in Atlantic salmon (Salmo salar). Fish Shellfish Immunol. 14: 115-132. http://dx.doi.org/10.1006/fsim.2002.0424 PMid:12526876   Li N, Yang Z, Bai J, Fu X, et al. (2010). A shared antigen among Vibrio species: outer membrane protein-OmpK as a versatile Vibriosis vaccine candidate in Orange-spotted grouper (Epinephelus coioides). Fish Shellfish Immunol. 28: 952-956. http://dx.doi.org/10.1016/j.fsi.2010.02.010 PMid:20170736   Liu M and Sadovy de Mitcheson Y (2008). Profile of a fishery collapse: why mariculture failed to save the large yellow croaker. Fish Fish. 9: 219-242. http://dx.doi.org/10.1111/j.1467-2979.2008.00278.x   Livak KJ and Schmittgen TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402-408.   Mao Y, Xu B, Su YQ, Zhang ZW, et al. (2010). Cloning and mRNA expression of macrophage migration inhibitory factor (MIF) gene of large yellow croaker (Pseudosciaena crocea). Acta Oceanol. Sin. 29: 63-73. http://dx.doi.org/10.1007/s13131-010-0037-8   Oh C, De Zoysa M, Nikapitiya C, Whang I, et al. (2010). Tumor suppressor QM-like gene from disk abalone (Haliotis discus discus): molecular characterization and transcriptional analysis upon immune challenge. Fish Shellfish Immunol. 29: 494-500. http://dx.doi.org/10.1016/j.fsi.2010.05.007 PMid:20580829   Ohta Y, Landis E, Boulay T, Phillips RB, et al. (2004). Homologs of CD83 from elasmobranch and teleost fish. J. Immunol. 173: 4553-4560. PMid:15383588   Rocha CS, Santos AA, Machado JP and Fontes EP (2008). The ribosomal protein L10/QM-like protein is a component of the NIK-mediated antiviral signaling. Virology 380: 165-169. http://dx.doi.org/10.1016/j.virol.2008.08.005 PMid:18789471   Sambrook J and David WR (2001). Molecular Cloning 3. Cold Spring Harbor Laboratory Press, Cold Spring Harbor.   Scotto-Lavino E, Du G and Frohman MA (2006). 5' end cDNA amplification using classic RACE. Nat. Protoc. 1: 2555-2562. http://dx.doi.org/10.1038/nprot.2006.480 PMid:17406509   Tanguy A, Guo X and Ford SE (2004). Discovery of genes expressed in response to Perkinsus marinus challenge in Eastern (Crassostrea virginica) and Pacific (C. gigas) oysters. Gene 338: 121-131. http://dx.doi.org/10.1016/j.gene.2004.05.019 PMid:15302413   Tobback E, Hermans K, Decostere A, Van den Broeck W, et al. (2010). Interactions of virulent and avirulent Yersinia ruckeri strains with isolated gill arches and intestinal explants of rainbow trout Oncorhynchus mykiss. Dis. Aquat. Organ. 90: 175-179. http://dx.doi.org/10.3354/dao02230 PMid:20815325   Wang J, Su YQ, Zhang ZX, Li M, et al. (2001). Bacterial pathogenetic biology of cultured Pseudosciaena crocea in southern Fujian. J. Xiamen Univ. 40: 85-91.   Wen Y, Shao JZ, Pan XX and Xiang LX (2005). Molecular cloning, characterization and expression analysis of QM gene from grass carp (Ctenopharyngodon idellus) homologous to Wilms' tumor suppressor. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 141: 356-365. http://dx.doi.org/10.1016/j.cbpc.2005.04.007 PMid:15936234   Wool IG (1996). Extraribosomal functions of ribosomal proteins. Trends Biochem. Sci. 21: 164-165. PMid:8871397   Workenhe ST, Rise ML, Kibenge MJ and Kibenge FS (2010). The fight between the teleost fish immune response and aquatic viruses. Mol. Immunol. 47: 2525-2536. http://dx.doi.org/10.1016/j.molimm.2010.06.009 PMid:20797792   Xu J, Wu S and Zhang X (2008). Novel function of QM protein of shrimp (Penaeus japonicus) in regulation of phenol oxidase activity by interaction with hemocyanin. Cell Physiol. Biochem. 21: 473-480. http://dx.doi.org/10.1159/000129640 PMid:18453755   Zhou F, Jiang S, Huang J, Qiu L, et al. (2011). Molecular analysis of the QM gene from Penaeus monodon and its expression on the different ovarian stages of development. Mol. Biol. Rep. 38: 1921-1927. http://dx.doi.org/10.1007/s11033-010-0312-y PMid:20872074
C. Huang, Yang, Y. - F., Zhang, H., Xie, L., Chen, J. - L., Wang, J., Tan, Z. - P., and Luo, H., Microdeletion on 17p11.2 in a Smith-Magenis syndrome patient with mental retardation and congenital heart defect: first report from China, vol. 11, pp. 2321-2327, 2012.
Cassidy SB and Driscoll DJ (2009). Prader-Willi syndrome. Eur. J. Hum. Genet. 17: 3-13. http://dx.doi.org/10.1038/ejhg.2008.165 PMid:18781185 PMCid:2985966   Chen JL, Yang YF, Huang C, Wang J, et al. (2012). Clinical and molecular delineation of 16p13.3 duplication in a patient with congenital heart defect and multiple congenital anomalies. Am. J. Med. Genet. A 158A: 685-688. http://dx.doi.org/10.1002/ajmg.a.34434 PMid:22307725   Edelman EA, Girirajan S, Finucane B, Patel PI, et al. (2007). Gender, genotype, and phenotype differences in Smith- Magenis syndrome: a meta-analysis of 105 cases. Clin. Genet. 71: 540-550. http://dx.doi.org/10.1111/j.1399-0004.2007.00815.x PMid:17539903   Elsea SH and Girirajan S (2008). Smith-Magenis syndrome. Eur. J. Hum. Genet. 16: 412-421. http://dx.doi.org/10.1038/sj.ejhg.5202009 PMid:18231123   Engelstad H, Carney G, S'aulis D, Rise J, et al. (2011). Large contiguous gene deletions in Sjogren-Larsson syndrome. Mol. Genet. Metab. 104: 356-361. http://dx.doi.org/10.1016/j.ymgme.2011.05.015 PMid:21684788 PMCid:3196763   Gamba BF, Vieira GH, Souza DH, Monteiro FF, et al. (2011). Smith-Magenis syndrome: clinical evaluation in seven Brazilian patients. Genet. Mol. Res. 10: 2664-2670. http://dx.doi.org/10.4238/2011.October.31.17 PMid:22057962   Girirajan S, Elsas LJ, Devriendt K and Elsea SH (2005). RAI1 variations in Smith-Magenis syndrome patients without 17p11.2 deletions. J. Med. Genet. 42: 820-828. http://dx.doi.org/10.1136/jmg.2005.031211 PMid:15788730 PMCid:1735950   Girirajan S, Vlangos CN, Szomju BB, Edelman E, et al. (2006). Genotype-phenotype correlation in Smith-Magenis syndrome: evidence that multiple genes in 17p11.2 contribute to the clinical spectrum. Genet. Med. 8: 417-427. http://dx.doi.org/10.1097/01.gim.0000228215.32110.89 PMid:16845274   Greenberg F, Guzzetta V, Montes dO-L, Magenis RE, et al. (1991). Molecular analysis of the Smith-Magenis syndrome: a possible contiguous-gene syndrome associated with del(17)(p11.2). Am. J. Hum. Genet. 49: 1207-1218. PMid:1746552 PMCid:1686451   Greenberg F, Lewis RA, Potocki L, Glaze D, et al. (1996). Multi-disciplinary clinical study of Smith-Magenis syndrome (deletion 17p11.2). Am. J. Med. Genet. 62: 247-254. http://dx.doi.org/10.1002/(SICI)1096-8628(19960329)62:3<247::AID-AJMG9>3.0.CO;2-Q   Gropman AL, Duncan WC and Smith AC (2006). Neurologic and developmental features of the Smith-Magenis syndrome (del 17p11.2). Pediatr. Neurol. 34: 337-350. http://dx.doi.org/10.1016/j.pediatrneurol.2005.08.018 PMid:16647992   Huang C, Yang YF, Yin N, Chen JL, et al. (2012). Congenital heart defect and mental retardation in a patient with a 13q33.1-34 deletion. Gene 498: 308-310. http://dx.doi.org/10.1016/j.gene.2012.01.083 PMid:22366306   Kalay E, Uzumcu A, Krieger E, Caylan R, et al. (2007). MYO15A (DFNB3) mutations in Turkish hearing loss families and functional modeling of a novel motor domain mutation. Am. J. Med. Genet. A 143A: 2382-2389. http://dx.doi.org/10.1002/ajmg.a.31937 PMid:17853461   Kleefstra T, van Zelst-Stams WA, Nillesen WM, Cormier-Daire V, et al. (2009). Further clinical and molecular delineation of the 9q subtelomeric deletion syndrome supports a major contribution of EHMT1 haploinsufficiency to the core phenotype. J. Med. Genet. 46: 598-606. http://dx.doi.org/10.1136/jmg.2008.062950 PMid:19264732   Kobrynski LJ and Sullivan KE (2007). Velocardiofacial syndrome, DiGeorge syndrome: the chromosome 22q11.2 deletion syndromes. Lancet 370: 1443-1452. http://dx.doi.org/10.1016/S0140-6736(07)61601-8   Myers SM and Challman TD (2004). Congenital heart defects associated with Smith-Magenis syndrome: two cases of total anomalous pulmonary venous return. Am. J. Med. Genet. A 131: 99-100. http://dx.doi.org/10.1002/ajmg.a.30290 PMid:15384100   Neumann SA, Tingley WG, Conklin BR, Shrader CJ, et al. (2009). AKAP10 (I646V) functional polymorphism predicts heart rate and heart rate variability in apparently healthy, middle-aged European-Americans. Psychophysiology 46: 466-472. http://dx.doi.org/10.1111/j.1469-8986.2009.00802.x PMid:19496216 PMCid:2890278   Shi FD and Jia JP (2011). Neurology and neurologic practice in China. Neurology 77: 1986-1992. http://dx.doi.org/10.1212/WNL.0b013e31823a0ed3 PMid:22123780   Slager RE, Newton TL, Vlangos CN, Finucane B, et al. (2003). Mutations in RAI1 associated with Smith-Magenis syndrome. Nat. Genet. 33: 466-468. http://dx.doi.org/10.1038/ng1126 PMid:12652298   Slavotinek AM (2008). Novel microdeletion syndromes detected by chromosome microarrays. Hum. Genet. 124: 1-17. http://dx.doi.org/10.1007/s00439-008-0513-9 PMid:18512078   Smith AC, McGavran L, Waldstein G and Robinson J (1982). Deletion of the 17 short arm in two patients with facial clefts. Am. J. Hum. Genet. 34 (Suppl): 410A.   Smith AC, McGavran L, Robinson J, Waldstein G, et al. (1986). Interstitial deletion of (17)(p11.2p11.2) in nine patients. Am. J. Med. Genet. 24: 393-414. http://dx.doi.org/10.1002/ajmg.1320240303 PMid:2425619   Stratton RF, Dobyns WB, Greenberg F, DeSana JB, et al. (1986). Interstitial deletion of (17)(p11.2p11.2): report of six additional patients with a new chromosome deletion syndrome. Am. J. Med. Genet. 24: 421-432. http://dx.doi.org/10.1002/ajmg.1320240305 PMid:3728561   Sweeney E, Peart I, Tofeig M and Kerr B (1999). Smith-Magenis syndrome and tetralogy of Fallot. J. Med. Genet. 36: 501-502. PMid:10874646 PMCid:1734392   Tan ZP, Huang C, Xu ZB, Yang JF, et al. (2011). Novel ZFPM2/FOG2 variants in patients with double outlet right ventricle. Clin. Genet. DOI 10.1111/j.1399-0004.2011.01787.x. http://dx.doi.org/10.1111/j.1399-0004.2011.01787.x   Wong JT, Chan DK, Wong KY, Tan M, et al. (2003). Smith-Magenis syndrome and cyanotic congenital heart disease: a case report. Clin. Dysmorphol. 12: 73-74. http://dx.doi.org/10.1097/00019605-200301000-00014 PMid:12514371
Y. Bao, Wang, J., He, F., Ma, H., and Wang, H., Molecular cytogenetic identification of a wheat (Triticum aestivum)-American dune grass (Leymus mollis) translocation line resistant to stripe rust, vol. 11, pp. 3198-3206, 2012.
Afzal SN, Haque MI, Ahmedani MS, Rauf A, et al. (2008). Impact of stripe rust on kernel weight of wheat varieties sown in rainfed areas of Pakistan. Pak. J. Bot. 40: 923-929.   Bariana HS and McIntosh RA (1993). Cytogenetic studies in wheat. XV. Location of rust resistance genes in VPM1 and their genetic linkage with other disease resistance genes in chromosome 2A. Genome 36: 476-482. http://dx.doi.org/10.1139/g93-065 PMid:18470001   Cao Z, Deng Z, Wang M, Wang X, et al. (2008). Inheritance and molecular mapping of an alien stripe-rust resistance gene from a wheat-Psathyrostachys huashanica translocation line. Plant Sci. 174: 544-549. http://dx.doi.org/10.1016/j.plantsci.2008.02.007   Chen Q, Conner RL, Ahmad F, Laroche A, et al. (1998). Molecular characterization of the genome composition of partial amphiploids derived from Triticum aestivum × Thinopyrum ponticum and T. aestivum × Th. intermedium as sources of resistance to wheat streak mosaic virus and its vector, Aceria tosichella. Theor. Appl. Genet. 97: 1-8. http://dx.doi.org/10.1007/s001220050860   Chen XM (2005). Epidemiology and control of stripe rust [Puccinia striiformis f. sp. tritici] on wheat. Can. J. Plant. Pathol. 27: 314-337. http://dx.doi.org/10.1080/07060660509507230   Dreisigacker S (2004). Genetic Diversity in Elite Lines and Land Races of CIMMYT Spring Bread Wheat and Hybrid Performance of Crosses Among Elite Germplasm. Ph.D. thesis, Faculty of Agriculture, University of Hohenheim, Hohenheim.   Friebe B, Jiang J, Raupp WJ, McIntosh RA, et al. (1996). Characterization of wheat-alien translocations conferring resistance to diseases and pests: current status. Euphytica 91: 59-87. http://dx.doi.org/10.1007/BF00035277   Fu J, Chen S and Zhang A (1993). Studies of the formation and cytogenetics of octoploid Tritileymus. Acta Genet. Sin. 20: 317-323.   Fu J, Chen S, Zhang A, Hou W, et al. (1996). Cytogenetic studies on the cross progenies between octoploid Tritileymus and Triticum aestivum. Acta Genet. Sin. 23: 24-31.   Fu J, Xu X, Yang Q, Chen S, et al. (1997). Cytogenetic studies on the cross between octoploid Tritileymus and nulllisomic wheat. Acta Genet. Sin. 24: 350-357.   He R, Chang Z, Yang Z, Yuan Z, et al. (2009). Inheritance and mapping of powdery mildew resistance gene Pm43 introgressed from Thinopyrum intermedium into wheat. Theor. Appl. Genet. 118: 1173-1180. http://dx.doi.org/10.1007/s00122-009-0971-z PMid:19214392   Hu LJ, Li GR, Zeng ZX, Chang ZJ, et al. (2011). Molecular cytogenetic identification of a new wheat-Thinopyrum substitution line with stripe rust resistance. Euphytica 177: 169-177. http://dx.doi.org/10.1007/s10681-010-0216-x   Jiang J, Friebe B and Gill BS (1994). Recent advances in alien gene transfer in wheat. Euphytica 73: 199-212. http://dx.doi.org/10.1007/BF00036700   Kang Z, Zhao J, Han D, Zhang H et al. (2010). Status of wheat rust research and control in China. Available at [http:// www.globalrust.org/db/attachments/bgriiwc/24/2/07-kang-ca-A4-embargo.pdf].   Kishii M, Wang RR and Tsujimoto H (2003). Characteristics and behaviour of the chromosomes of Leymus mollis and L. racemosus (Triticeae, Poaceae) during mitosis and meiosis. Chromosome Res 11: 741-748. http://dx.doi.org/10.1023/B:CHRO.0000005774.00726.71 PMid:14712860   Kuraparthy V, Chhuneja P, Dhaliwal HS, Kaur S, et al. (2007a). Characterization and mapping of cryptic alien introgression from Aegilops geniculata with new leaf rust and stripe rust resistance genes Lr57 and Yr40 in wheat. Theor. Appl. Genet. 114: 1379-1389. http://dx.doi.org/10.1007/s00122-007-0524-2 PMid:17356867   Kuraparthy V, Sood S, Chhuneja P, Dhaliwal HS, et al. (2007b). A cryptic wheat-Aegilops triuncialis translocation with leaf rust resistance gene Lr58. Crop Sci. 47: 1995-2003. http://dx.doi.org/10.2135/cropsci2007.01.0038   Li Q, Chen XM, Wang MN and Jing JX (2011). Yr45, a new wheat gene for stripe rust resistance on the long arm of chromosome 3D. Theor. Appl. Genet. 122: 189-197. http://dx.doi.org/10.1007/s00122-010-1435-1 PMid:20838759   Luo PG, Luo HY, Chang ZJ, Zhang HY, et al. (2009). Characterization and chromosomal location of Pm40 in common wheat: a new gene for resistance to powdery mildew derived from Elytrigia intermedium. Theor. Appl. Genet. 118: 1059-1064. http://dx.doi.org/10.1007/s00122-009-0962-0 PMid:19194691   Nasuda S, Friebe B, Busch W, Kynast RG, et al. (1998). Structural rearrangement in chromosome 2M of Aegilops comosa has prevented the utilization of the compair and related wheat-Ae. comosa translocations in wheat improvement. Theor. Appl. Genet. 96: 780-785. http://dx.doi.org/10.1007/s001220050802   Singh RP, Nelson JC and Sorrells ME (2000). Mapping Yr28 and other genes for resistance to stripe rust in wheat. Crop Sci. 40: 1148-1155. http://dx.doi.org/10.2135/cropsci2000.4041148x   Sui XX, Wang MN and Chen XM (2009). Molecular mapping of a stripe rust resistance gene in spring wheat cultivar Zak. Phytopathology 99: 1209-1215. http://dx.doi.org/10.1094/PHYTO-99-10-1209 PMid:19740035   Wan AM, Chen XM and He ZH (2007). Wheat stripe rust in China. Aust. J. Agr. Res. 58: 605-619. http://dx.doi.org/10.1071/AR06142   Wang XP, Fu J, Zhang XQ, Jing JK, et al. (2000). Molecular cytogenetic study on genome constitutions of octoploid Tritileymus. Acta Bot. Sin. 42: 582-586.   Yu JK, Dake TM, Singh S, Benscher D, et al. (2004). Development and mapping of EST-derived simple sequence repeat markers for hexaploid wheat. Genome 47: 805-818. http://dx.doi.org/10.1139/g04-057 PMid:15499395   Zhang P, McIntosh RA, Hoxha S and Dong C (2009). Wheat stripe rust resistance genes Yr5 and Yr7 are allelic. Theor. Appl. Genet. 120: 25-29. http://dx.doi.org/10.1007/s00122-009-1156-5 PMid:19763533   Zhou YC, Zhang XQ, Wang XP, Wu LR, et al. (2001). Chromosomal location and molecular marker of resistance gene to Puccinia striiformis west. in Leymus mollis Trin. Hara. Yi Chuan Xue Bao 28: 864-869. PMid:11582747
Q. G. Li, Wu, X. R., Li, X. Z., Yu, J., Xia, Y., Wang, A. P., and Wang, J., Neural-endocrine mechanisms of respiratory syncytial virus-associated asthma in a rat model, vol. 11, pp. 2780-2789, 2012.
Aloe L and Levi-Montalcini R (1979). Nerve growth factor-induced transformation of immature chromaffin cells in vivo into sympathetic neurons: effect of antiserum to nerve growth factor. Proc. Natl. Acad. Sci. U. S. A. 76: 1246-1250. http://dx.doi.org/10.1073/pnas.76.3.1246 PMid:286308 PMCid:383227   Auais A, Adkins B, Napchan G and Piedimonte G (2003). Immunomodulatory effects of sensory nerves during respiratory syncytial virus infection in rats. Am. J. Physiol. Lung Cell Mol. Physiol. 285: L105-L113. PMid:12639840   Bonini S, Lambiase A, Lapucci G, Properzi F, et al. (2002). Nerve growth factor and asthma. Allergy 57 (Suppl 72): 13-15. http://dx.doi.org/10.1034/j.1398-9995.57.s72.3.x PMid:12144547   Braun A, Lommatzsch M, Lewin GR, Virchow JC, et al. (1999). Neurotrophins: a link between airway inflammation and airway smooth muscle contractility in asthma? Int. Arch. Allergy Immunol. 118: 163-165. http://dx.doi.org/10.1159/000024056 PMid:10224367   Dong CC, Yin XJ, Ma JY, Millecchia L, et al. (2005). Effect of diesel exhaust particles on allergic reactions and airway responsiveness in ovalbumin-sensitized brown Norway rats. Toxicol. Sci. 88: 202-212. http://dx.doi.org/10.1093/toxsci/kfi280 PMid:16107553   Feng JT and Hu CP (2005). Dysfunction of releasing adrenaline in asthma by nerve growth factor. Med. Hypotheses 65: 1043-1046. http://dx.doi.org/10.1016/j.mehy.2005.06.029 PMid:16139964   Feng JT, Li XZ, Hu CP, Wang J, et al. (2010). Neural plasticity occurs in the adrenal medulla of asthmatic rats. Chin. Med. J. 123: 1333-1337.   Freund-Michel V and Frossard N (2008). The nerve growth factor and its receptors in airway inflammatory diseases. Pharmacol. Ther. 117: 52-76. http://dx.doi.org/10.1016/j.pharmthera.2007.07.003 PMid:17915332   Greene LA and Tischler AS (1976). Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc. Natl. Acad. Sci. U. S. A. 73: 2424-2428. http://dx.doi.org/10.1073/pnas.73.7.2424 PMid:1065897 PMCid:430592   Hahn C, Islamian AP, Renz H and Nockher WA (2006). Airway epithelial cells produce neurotrophins and promote the survival of eosinophils during allergic airway inflammation. J. Allergy Clin. Immunol. 117: 787-794. http://dx.doi.org/10.1016/j.jaci.2005.12.1339 PMid:16630935   Hu C, Wedde-Beer K, Auais A, Rodriguez MM, et al. (2002). Nerve growth factor and nerve growth factor receptors in respiratory syncytial virus-infected lungs. Am. J. Physiol. Lung Cell Mol. Physiol. 283: L494-L502. PMid:12114213   Huang EJ and Reichardt LF (2001). Neurotrophins: roles in neuronal development and function. Annu. Rev. Neurosci. 24: 677-736. http://dx.doi.org/10.1146/annurev.neuro.24.1.677 PMid:11520916 PMCid:2758233   Jun W, Cheng-ping H and Wei-jun L (2007). Study on NGF expression and phenotype transformation of AMCC in asthmatic rats. Chin. J. Pract. Int. Med.   Kasprzak A, Zabel M and Biczysko W (2007). Selected markers (chromogranin A, neuron-specific enolase, synaptophysin, protein gene product 9.5) in diagnosis and prognosis of neuroendocrine pulmonary tumours. Pol. J. Pathol. 58: 23-33. PMid:17585539   Lillien LE and Claude P (1985). Nerve growth factor is a mitogen for cultured chromaffin cells. Nature 317: 632-634. http://dx.doi.org/10.1038/317632a0 PMid:4058573   Mohapatra SS and Boyapalle S (2008). Epidemiologic, experimental, and clinical links between respiratory syncytial virus infection and asthma. Clin. Microbiol. Rev. 21: 495-504. http://dx.doi.org/10.1128/CMR.00054-07 PMid:18625684 PMCid:2493089   Psarras S, Papadopoulos NG and Johnston SL (2004). Pathogenesis of respiratory syncytial virus bronchiolitis-related wheezing. Paediatr. Respir. Rev. 5 (Suppl A): S179-S184. http://dx.doi.org/10.1016/S1526-0542(04)90034-6   Shen XY, Pan PH, Wu ES and Hu CP (2006). Effects of respiratory syncytial virus infection on the airway neuronal plasticity and its relationship to the bronchial hyperresponsiveness in rats. Chin. Med. J. 119: 156-159.   Shojo H and Kibayashi K (2006). Changes in localization of synaptophysin following fluid percussion injury in the rat brain. Brain Res. 1078: 198-211. http://dx.doi.org/10.1016/j.brainres.2006.01.063 PMid:16497279   Stensballe LG, Simonsen JB, Thomsen SF, Larsen AM, et al. (2009). The causal direction in the association between respiratory syncytial virus hospitalization and asthma. J. Allergy Clin. Immunol. 123: 131-137. http://dx.doi.org/10.1016/j.jaci.2008.10.042 PMid:19130934   Tortorolo L, Langer A, Polidori G, Vento G, et al. (2005). Neurotrophin overexpression in lower airways of infants with respiratory syncytial virus infection. Am. J. Respir. Crit. Care Med. 172: 233-237. http://dx.doi.org/10.1164/rccm.200412-1693OC PMid:15879412   Unsicker K, Krisch B, Otten U and Thoenen H (1978). Nerve growth factor-induced fiber outgrowth from isolated rat adrenal chromaffin cells: impairment by glucocorticoids. Proc. Natl. Acad. Sci. U. S. A. 75: 3498-3502. http://dx.doi.org/10.1073/pnas.75.7.3498 PMid:28526 PMCid:392805   Unsicker K, Huber K, Schutz G and Kalcheim C (2005). The chromaffin cell and its development. Neurochem. Res. 30: 921-925. http://dx.doi.org/10.1007/s11064-005-6966-5 PMid:16187226   Wang J, Hu C and Feng J (2006). Dysfunction of releasing adrenaline in asthmatic adrenaline medullary chromaffin cells due to functional redundancy primed by nerve growth factor. Zhonghua Jie He Hu Xi Za Zhi 29: 812-815.   Wedde-Beer K, Hu C, Rodriguez MM and Piedimonte G (2002). Leukotrienes mediate neurogenic inflammation in lungs of young rats infected with respiratory syncytial virus. Am. J. Physiol. Lung Cel. Mol. Physiol. 282: L1143-L1150. PMid:11943681   Welliver RC (2003). Respiratory syncytial virus and other respiratory viruses. Pediatr. Infect. Dis. J. 22: S6-10. http://dx.doi.org/10.1097/01.inf.0000053880.92496.db PMid:12671447   Wiesmann C, Ultsch MH, Bass SH and de Vos AM (1999). Crystal structure of nerve growth factor in complex with the ligand-binding domain of the TrkA receptor. Nature 401: 184-188. http://dx.doi.org/10.1038/43705 PMid:10490030   Witzenrath M, Ahrens B, Kube SM, Braun A, et al. (2006). Detection of allergen-induced airway hyperresponsiveness in isolated mouse lungs. Am. J. Physiol. Lung Cell Mol. Physiol. 291: L466-L472. http://dx.doi.org/10.1152/ajplung.00011.2005 PMid:16617097   Ye H, Kuruvilla R, Zweifel LS and Ginty DD (2003). Evidence in support of signaling endosome-based retrograde survival of sympathetic neurons. Neuron 39: 57-68. http://dx.doi.org/10.1016/S0896-6273(03)00266-6
F. H. Bai, Wang, N. J., Wang, J., Yang, L., Zhang, F. M., Yin, F., Liang, J., Wu, K. C., and Fan, D. M., Screening and identification of peritoneal metastasis-related genes of gastric adenocarcinoma using a cDNA microarray, vol. 11, pp. 1682-1689, 2012.
Arboleda MJ, Lyons JF and Kabbinavar FF (2003). Overexpression of AKT2/protein kinase Bh leads to up-regulation of h1 integrins, increased invasion, and metastasis of human breast and ovarian cancer cells. Cancer Res. 63: 196-206. PMid:12517798   Bai F, Liang J, Wang J, Shi Y, et al. (2007). Inhibitory effects of a specific phage-displayed peptide on high peritoneal metastasis of gastric cancer. J. Mol. Med. 85: 169-180. http://dx.doi.org/10.1007/s00109-006-0115-8 PMid:17043801   Brito M, Malta-Vacas J, Carmona B, Aires C, et al. (2005). Polyglycine expansions in eRF3/GSPT1 are associated with gastric cancer susceptibility. Carcinogenesis 26: 2046-2049. http://dx.doi.org/10.1093/carcin/bgi168 PMid:15987717   Cho YG, Nam SW, Kim TY, Kim YS, et al. (2003). Overexpression of S100A4 is closely related to the aggressiveness of gastric cancer. APMIS 111: 539-545. http://dx.doi.org/10.1034/j.1600-0463.2003.1110502.x PMid:12887505   Choi MG, Sung CO, Noh JH, Kim KM, et al. (2010). Mucinous gastric cancer presents with more advanced tumor stage and weaker beta-catenin expression than nonmucinous cancer. Ann. Surg. Oncol. 17: 3053-3058. http://dx.doi.org/10.1245/s10434-010-1184-z PMid:20645013   Davidson B, Zhang Z, Kleinberg L, Li M, et al. (2006). Gene expression signatures differentiate ovarian/peritoneal serous carcinoma from diffuse malignant peritoneal mesothelioma. Clin. Cancer Res. 12: 5944-5950. http://dx.doi.org/10.1158/1078-0432.CCR-06-1059 PMid:17062665   Hippo Y, Yashiro M, Ishii M, Taniguchi H, et al. (2001). Differential gene expression profiles of scirrhous gastric cancer cells with high metastatic potential to peritoneum or lymph nodes. Cancer Res. 61: 889-895. PMid:11221876   Huerta S, Harris DM, Jazirehi A, Bonavida B, et al. (2003). Gene expression profile of metastatic colon cancer cells resistant to cisplatin-induced apoptosis. Int. J. Oncol. 22: 663-670. PMid:12579322   Jemal A, Murray T, Ward E, Samuels A, et al. (2005). Cancer statistics, 2005. CA Cancer J. Clin. 55: 10-30. http://dx.doi.org/10.3322/canjclin.55.1.10 PMid:15661684   Kang YH, Lee HS and Kim WH (2002). Promoter methylation and silencing of PTEN in gastric carcinoma. Lab. Invest. 82: 285-291. http://dx.doi.org/10.1038/labinvest.3780422 PMid:11896207   Lazăr D, Raica M, Sporea I, Tăban S, et al. (2006). Tumor angiogenesis in gastric cancer. Rom. J. Morphol. Embryol. 47: 5-13. PMid:16838051   Lee SS, Jeong HE, Liu KH, Ryu JY, et al. (2005). Identification and functional characterization of novel CYP2J2 variants: G312R variant causes loss of enzyme catalytic activity. Pharmacogenet. Genomics 15: 105-113. http://dx.doi.org/10.1097/01213011-200502000-00006 PMid:15861034   Li DW, Wu Q, Peng ZH, Yang ZR, et al. (2007). Expression and significance of Notch1 and PTEN in gastric cancer. Ai Zheng 26: 1183-1187. PMid:17991315   Li J, Wu Y, Qian X and Sha B (2006). Crystal structure of yeast Sis1 peptide-binding fragment and Hsp70 Ssa1 C-terminal complex. Biochem. J. 398: 353-360. http://dx.doi.org/10.1042/BJ20060618 PMid:16737444 PMCid:1559466   Mori K, Aoyagi K, Ueda T, Danjoh I, et al. (2004). Highly specific marker genes for detecting minimal gastric cancer cells in cytology negative peritoneal washings. Biochem. Biophys. Res. Commun. 313: 931-937. http://dx.doi.org/10.1016/j.bbrc.2003.12.025 PMid:14706632   Motoori M, Takemasa I, Doki Y, Saito S, et al. (2006). Prediction of peritoneal metastasis in advanced gastric cancer by gene expression profiling of the primary site. Eur. J. Cancer 42: 1897-1903. http://dx.doi.org/10.1016/j.ejca.2006.04.007 PMid:16831544   Ong CK, Ng CY, Leong C, Ng CP, et al. (2004). Genomic structure of human OKL38 gene and its differential expression in kidney carcinogenesis. J. Biol. Chem. 279: 743-754. http://dx.doi.org/10.1074/jbc.M308668200 PMid:14570898   Retterspitz MF, Monig SP, Schreckenberg S, Schneider PM, et al. (2010). Expression of {beta}-catenin, MUC1 and c-met in diffuse-type gastric carcinomas: correlations with tumour progression and prognosis. Anticancer Res. 30: 4635-4641. PMid:21115917   Schena M, Shalon D, Davis RW and Brown PO (1995). Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science 270: 467-470. http://dx.doi.org/10.1126/science.270.5235.467 PMid:7569999   Schieren G, Rumberger B, Klein M, Kreutz C, et al. (2006). Gene profiling of polycystic kidneys. Nephrol. Dial. Transplant 21: 1816-1824. http://dx.doi.org/10.1093/ndt/gfl071 PMid:16520345   Shapira M, Ben-Izhak O, Bishara B, Futerman B, et al. (2004). Alterations in the expression of the cell cycle regulatory protein cyclin kinase subunit 1 in colorectal carcinoma. Cancer 100: 1615-1621. http://dx.doi.org/10.1002/cncr.20172 PMid:15073847   Wang J, Wu K, Bai F, Zhai H, et al. (2006). Celecoxib could reverse the hypoxia-induced Angiopoietin-2 upregulation in gastric cancer. Cancer Lett. 242: 20-27. http://dx.doi.org/10.1016/j.canlet.2005.10.030 PMid:16338068   Wang YY, Ye ZY, Zhao ZS, Tao HQ, et al. (2010). High-level expression of S100A4 correlates with lymph node metastasis and poor prognosis in patients with gastric cancer. Ann. Surg. Oncol. 17: 89-97. http://dx.doi.org/10.1245/s10434-009-0722-z PMid:19820999   Yanagihara K, Takigahira M, Tanaka H, Komatsu T, et al. (2005). Development and biological analysis of peritoneal metastasis mouse models for human scirrhous stomach cancer. Cancer Sci. 96: 323-332. http://dx.doi.org/10.1111/j.1349-7006.2005.00054.x PMid:15958054   Yonemura Y, Endou Y, Kimura K, Fushida S, et al. (2000). Inverse expression of S100A4 and E-cadherin is associated with metastatic potential in gastric cancer. Clin. Cancer Res. 6: 4234-4242. PMid:11106237   Yonemura Y, Endo Y, Obata T and Sasaki T (2007). Recent advances in the treatment of peritoneal dissemination of gastrointestinal cancers by nucleoside antimetabolites. Cancer Sci. 98: 11-18. http://dx.doi.org/10.1111/j.1349-7006.2006.00350.x PMid:17052255   Yoon CS, Hyung WJ, Lee JH, Chae YS, et al. (2008). Expression of S100A4, E-cadherin, alpha- and beta-catenin in gastric adenocarcinoma. Hepatogastroenterology 55: 1916-1920. PMid:19102422   Yoshikawa T, Yanoma S, Tsuburaya A, Kobayashi O, et al. (2006). Expression of MMP-7 and MT1-MMP in peritoneal dissemination of gastric cancer. Hepatogastroenterology 53: 964-967. PMid:17153464
L. H. Xu, Wang, C. H., Wang, J., Dong, Z. J., Ma, Y. Q., and Yang, X. X., Selection pressures have driven population differentiation of domesticated and wild common carp (Cyprinus carpio L.), vol. 11, pp. 3222-3235, 2012.
Appio KT and Weber LI (2007). Temporal genetic differentiation in cultured and natural beds of the brown mussel Perna perna (Mytilidae). Genet. Mol. Res. 6: 127-136. PMid:17469062   Aung O, Nguyen TTT, Poompuang S and Kamonrat W (2010). Microsatellite DNA markers revealed genetic population structure among captive stocks and wild populations of mrigal, Cirrhinus cirrhosus in Myanmar. Aquaculture 299: 37-43. http://dx.doi.org/10.1016/j.aquaculture.2009.12.010   Balon E (1995). Origin and domestication of the wild carp, Cyprinus carpio: from Roman gourmets to the swimming flowers. Aquaculture 129: 3-48. http://dx.doi.org/10.1016/0044-8486(94)00227-F   Barker JSF, Frydenberg J, González J, Davies HI, et al. (2009). Bottlenecks, population differentiation and apparent selection at microsatellite loci in Australian Drosophila buzzatii. Heredity 102: 389-401. http://dx.doi.org/10.1038/hdy.2008.127 PMid:19142202   Bazin E, Glemin S and Galtier N (2006). Population size does not influence mitochondrial genetic diversity in animals. Science 312: 570-572. http://dx.doi.org/10.1126/science.1122033 PMid:16645093   Beaumont MA and Nichols RA (1996). Evaluating loci for use in the genetic analysis of population structure. Proc. R. Soc. Lond. B 263: 1619-1626. http://dx.doi.org/10.1098/rspb.1996.0237   Beaumont MA and Balding DJ (2004). Identifying adaptive genetic divergence among populations from genome scans. Mol. Ecol. 13: 969-980. http://dx.doi.org/10.1111/j.1365-294X.2004.02125.x PMid:15012769   Carlson SM, Edeline E, Asbjorn VL, Haugen TO, et al. (2007). Four decades of opposing natural and human-induced artificial selection acting on Windermere pike (Esox lucius). Ecol. Lett. 10: 512-521. http://dx.doi.org/10.1111/j.1461-0248.2007.01046.x PMid:17498150   Cheng Y, Wakefield M, Siddle HV, Coggill PC, et al. (2009). Isolation and characterization of 10 MHC Class I-associated microsatellite loci in tammar wallaby (Macropus eugenii). Mol. Ecol. Resour. 9: 346-349. http://dx.doi.org/10.1111/j.1755-0998.2008.02401.x PMid:21564646   Chistiakov DA and Voronova NV (2009). Genetic evolution and diversity of common carp Cyprinus carpio L. Cent. Eur. J. Biol. 4: 304-312. http://dx.doi.org/10.2478/s11535-009-0024-2   Coombs JA, Letcher BH and Nislow KH (2008). CREATE: a software to create input files from diploid genotypic data for 52 genetic software programs. Mol. Ecol. Resour. 8: 578-580. http://dx.doi.org/10.1111/j.1471-8286.2007.02036.x PMid:21585837   Cornuet JM and Luikart G (1996). Description and power analysis of two tests for detecting recent population bottlenecks from allele frequency data. Genetics 144: 2001-2014. PMid:8978083 PMCid:1207747   Dupanloup I, Schneider S and Excoffier L (2002). A simulated annealing approach to define the genetic structure of populations. Mol. Ecol. 11: 2571-2581. http://dx.doi.org/10.1046/j.1365-294X.2002.01650.x PMid:12453240   Excoffier L and Lischer HEL (2010). Arlequin suite ver 3.5: a new series of programs to perform population genetics analyses under Linux and Windows. Mol. Ecol. Resour. 10: 564-567. http://dx.doi.org/10.1111/j.1755-0998.2010.02847.x PMid:21565059   Excoffier L, Hofer T and Foll M (2009). Detecting loci under selection in a hierarchically structured population. Heredity 103: 285-298. http://dx.doi.org/10.1038/hdy.2009.74 PMid:19623208   Fraser DJ, Cook AM, Eddington JD, Bentzen P, et al. (2008). Mixed evidence for reduced local adaptation in wild salmon resulting from interbreeding with escaped farmed salmon: complexities in hybrid fitness. Evol. Appl. 1: 501-512. http://dx.doi.org/10.1111/j.1752-4571.2008.00037.x PMCid:3352379   Galtier N, Nabholz B, Glemin S and Hurst GD (2009). Mitochondrial DNA as a marker of molecular diversity: a reappraisal. Mol. Ecol. 18: 4541-4550. http://dx.doi.org/10.1111/j.1365-294X.2009.04380.x PMid:19821901   Goudet J (1995). FSTAT (Version 1.2): A computer program to calculate F-statistics. J. Heredity 86: 485-486.   Hall TA (1999). BioEdit: A user-friendly biological sequence alignment editor and analysis program for Windows 95/98/ NT. Nucleic Acids Symp. Ser. 41: 95-98.   Hansen MM, Meier K and Mensberg KLD (2010). Identifying footprints of selection in stocked brown trout populations: a spatio-temporal approach. Mol. Ecol. 19: 1787-1800. http://dx.doi.org/10.1111/j.1365-294X.2010.04615.x PMid:20345684   Innan H and Kim Y (2004). Pattern of polymorphism after strong artificial selection in a domestication event. Proc. Natl. Acad. Sci. U. S. A. 101: 10667-10672. http://dx.doi.org/10.1073/pnas.0401720101 PMid:15249682 PMCid:489992   Kauer MO, Dieringer D and Schlotterer C (2003). A microsatellite variability screen for positive selection associated with the "out of Africa" habitat expansion of Drosophila melanogaster. Genetics 165: 1137-1148. PMid:14668371 PMCid:1462820   Kohlmann K, Kersten P and Flajšhans M (2005). Microsatellite-based genetic variability and differentiation of domesticated, wild and feral common carp (Cyprinus carpio L.) populations. Aquaculture 247: 253-266. http://dx.doi.org/10.1016/j.aquaculture.2005.02.024   Kohlmann K, Kersten P and Flajshans M (2007). Comparison of microsatellite variability in wild and cultured tench (Tinca tinca). Aquaculture 272: S147-S151. http://dx.doi.org/10.1016/j.aquaculture.2007.08.003   Kuhner MK and Smith LP (2007). Comparing likelihood and Bayesian coalescent estimation of population parameters. Genetics 175: 155-165. http://dx.doi.org/10.1534/genetics.106.056457 PMid:16510781 PMCid:1775024   Li Q, Shu J, Yu RH and Tian CY (2007). Genetic variability of cultured populations of the Pacific abalone (Haliotis discus hannai Ino) in China based on microsatellites. Aquac. Res. 38: 981-990. http://dx.doi.org/10.1111/j.1365-2109.2007.01764.x   Lind CE, Evans BS, Knauer J, Taylor JJU, et al. (2009). Decreased genetic diversity and a reduced effective population size in cultured silver-lipped pearl oysters (Pinctada maxima). Aquaculture 286: 12-19. http://dx.doi.org/10.1016/j.aquaculture.2008.09.009   Ljungqvist M, Åkesson M and Hansson B (2010). Do microsatellites reflect genome-wide genetic diversity in natural populations? Mol. Ecol. 19: 851-855. http://dx.doi.org/10.1111/j.1365-294X.2010.04522.x PMid:20102512   Lou YD and Sun JC (2001). Progress on studies of origin and genetic diversity of three breeds of red carp in Jiangxi province. J. Fish. China 25: 570-575.   McGinnity P, Prodohl P, Ferguson A, Hynes R, et al. (2003). Fitness reduction and potential extinction of wild populations of Atlantic salmon, Salmo salar, as a result of interactions with escaped farm salmon. Proc. Biol. Sci. 270: 2443-2450. http://dx.doi.org/10.1098/rspb.2003.2520 PMid:14667333 PMCid:1691531   Nei M (1987). Molecular Evolution Genetics. Columbia University Press, New York.   Nielsen R (2005). Molecular signatures of natural selection. Annu. Rev. Genet. 39: 197-218. http://dx.doi.org/10.1146/annurev.genet.39.073003.112420 PMid:16285858   Peakall R and Smouse PE (2006). GENALEX 6: genetic analysis in Excel. Population genetic software for teaching and research. Mol. Ecol. Notes 6: 288-295. http://dx.doi.org/10.1111/j.1471-8286.2005.01155.x   Peel D, Ovenden JR and Peel SL (2004). NeEstimator: Software for Estimating Effective Population Size. Version 1.3. Department of Primary Industries and Fisheries, Queensland Government, Brisbane.   Piry S, Luikart G and Cornuet JM (1999). BOTTLENECK: a computer program for detecting recent reductions in the effective size using allele frequency data. J. Heredity 90: 502-503. http://dx.doi.org/10.1093/jhered/90.4.502   Posada D (2006). Collapse: Describing Haplotypes from Sequence Alignments. In: Computational Evolutionary Biology Laboratory, University of Vigo, Vigo.   Rahman SMZ, Khan MR, Islam S and Alam S (2009). Genetic variation of wild and hatchery populations of the catla Indian major carp (Catla catla Hamilton 1822: Cypriniformes, Cyprinidae) revealed by RAPD markers. Genet. Mol. Biol. 32: 197-201. http://dx.doi.org/10.1590/S1415-47572009005000013 PMid:21637668 PMCid:3032962   Raymond M and Rousset F (1995). GENEPOP (version 1.2): Population genetics software for exact tests and ecumenicism. Heredity 86: 248-249.   Rogell B, Eklund M, Thorngren H, Laurila A, et al. (2010). The effects of selection, drift and genetic variation on life-history trait divergence among insular populations of natterjack toad, Bufo calamita. Mol. Ecol. 19: 2229-2240. http://dx.doi.org/10.1111/j.1365-294X.2010.04642.x PMid:20465584   Sambrook Jl and Russell DW (2001). Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, New York.   Van Oosterhout C, Hutchinson WF, Wills DPM and Shipley P (2004). MICRO-CHECKER: software for identifying and correcting genotyping errors in microsatellite data. Mol. Ecol. Notes 4: 535-538. http://dx.doi.org/10.1111/j.1471-8286.2004.00684.x   Vigouroux Y, McMullen M, Hittinger CT, Houchins K, et al. (2002). Identifying genes of agronomic importance in maize by screening microsatellites for evidence of selection during domestication. Proc. Natl. Acad. Sci. U. S. A. 99: 9650- 9655. http://dx.doi.org/10.1073/pnas.112324299 PMid:12105270 PMCid:124964   Wang CH, Li SF, Nagy ZT, Lehoczky I, et al. (2010). Molecular genetic structure and relationship of Chinese and Hungarian common carp (Cyprinus carpio L.) strains based on mitochondrial sequence. Aquac. Res. 41: 1339-1347. http://dx.doi.org/10.1111/j.1365-2109.2009.02422.x   Zhang JS and Sun XY (2006). Open and analysis of breeding techniques for Jian common carp. China Fish. 9: 69-72.

Pages