366 results on '"Muzny, DM"'
Search Results
2. Comprehensive molecular characterization of human colon and rectal cancer
- Author
-
Collisson, Eric, Muzny, DM, Bainbridge, MN, Chang, K, Dinh, HH, Drummond, JA, Fowler, G, Kovar, CL, Lewis, LR, Morgan, MB, and Newsham, IF
- Abstract
To characterize somatic alterations in colorectal carcinoma, we conducted a genome-scale analysis of 276 samples, analysing exome sequence, DNA copy number, promoter methylation and messenger RNA and microRNA expression. A subset of these samples (97) unde
- Published
- 2012
3. Integrated genomic analyses of ovarian carcinoma
- Author
-
Bell, D, Berchuck, A, Birrer, M, Chien, J, Cramer, DW, Dao, F, Dhir, R, DiSaia, P, Gabra, H, Glenn, P, Godwin, AK, Gross, J, Hartmann, L, Huang, M, Huntsman, DG, Iacocca, M, Imielinski, M, Kalloger, S, Karlan, BY, Levine, DA, Mills, GB, Morrison, C, Mutch, D, Olvera, N, Orsulic, S, Park, K, Petrelli, N, Rabeno, B, Rader, JS, Sikic, BI, Smith-McCune, K, Sood, AK, Bowtell, D, Penny, R, Testa, JR, Chang, K, Creighton, CJ, Dinh, HH, Drummond, JA, Fowler, G, Gunaratne, P, Hawes, AC, Kovar, CL, Lewis, LR, Morgan, MB, Newsham, IF, Santibanez, J, Reid, JG, Trevino, LR, Wu, Y-Q, Wang, M, Muzny, DM, Wheeler, DA, Gibbs, RA, Getz, G, Lawrence, MS, Cibulskis, K, Sivachenko, AY, Sougnez, C, Voet, D, Wilkinson, J, Bloom, T, Ardlie, K, Fennell, T, Baldwin, J, Nichol, R, Fisher, S, Gabriel, S, Lander, ES, Ding, L, Fulton, RS, Koboldt, DC, McLellan, MD, Wylie, T, Walker, J, O’Laughlin, M, Dooling, DJ, Fulton, L, Abbott, R, Dees, ND, Zhang, Q, Kandoth, C, Wendl, M, Schierding, W, Shen, D, Harris, CC, Schmidt, H, Kalicki, J, Delehaunty, KD, Fronick, CC, Demeter, R, Cook, L, Wallis, JW, Lin, L, Magrini, VJ, Hodges, JS, Eldred, JM, Smith, SM, Pohl, CS, and Vandin, F
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Oncology and Carcinogenesis ,Biotechnology ,Rare Diseases ,Human Genome ,Ovarian Cancer ,Cancer ,Aged ,Carcinoma ,DNA Methylation ,Female ,Gene Dosage ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Genomics ,Humans ,MicroRNAs ,Middle Aged ,Mutation ,Ovarian Neoplasms ,RNA ,Messenger ,Cancer Genome Atlas Research Network ,General Science & Technology - Abstract
A catalogue of molecular aberrations that cause ovarian cancer is critical for developing and deploying therapies that will improve patients' lives. The Cancer Genome Atlas project has analysed messenger RNA expression, microRNA expression, promoter methylation and DNA copy number in 489 high-grade serous ovarian adenocarcinomas and the DNA sequences of exons from coding genes in 316 of these tumours. Here we report that high-grade serous ovarian cancer is characterized by TP53 mutations in almost all tumours (96%); low prevalence but statistically recurrent somatic mutations in nine further genes including NF1, BRCA1, BRCA2, RB1 and CDK12; 113 significant focal DNA copy number aberrations; and promoter methylation events involving 168 genes. Analyses delineated four ovarian cancer transcriptional subtypes, three microRNA subtypes, four promoter methylation subtypes and a transcriptional signature associated with survival duration, and shed new light on the impact that tumours with BRCA1/2 (BRCA1 or BRCA2) and CCNE1 aberrations have on survival. Pathway analyses suggested that homologous recombination is defective in about half of the tumours analysed, and that NOTCH and FOXM1 signalling are involved in serous ovarian cancer pathophysiology.
- Published
- 2011
4. DNA methylation patterns identify subgroups of pancreatic neuroendocrine tumors with clinical association
- Author
-
Lakis, V, Lawlor, RT, Newell, F, Patch, AM, Mafficini, A, Sadanandam, A, Koufariotis, LT, Johnston, RL, Leonard, C, Wood, S, Rusev, B, Corbo, V, Luchini, C, Cingarlini, S, Landoni, L, Salvia, R, Milella, M, Chang, D, Bailey, P, Jamieson, NB, Duthie, F, Gingras, MC, Muzny, DM, Wheeler, DA, Gibbs, RA, Milione, M, Chantrill, LA, Timpson, P, Chou, A, Pajic, M, Murphy, A, Dwarte, T, Hermann, D, Vennin, C, Cox, TR, Pereira, B, Ritchie, S, Reed, DA, Chambers, CR, Metcalf, X, Nobis, M, Mukhopadhyay, P, Addala, V, Kazakoff, S, Holmes, O, Xu, Q, Hofmann, O, Samra, JS, Pavlakis, N, Arena, J, High, HA, Asghari, R, Merrett, ND, Pavey, D, Das, A, Cosman, PH, Ismail, K, O’Connnor, C, Stoita, A, Williams, D, Spigellman, A, Lam, VW, McLeod, D, Kirk, J, Kench, JG, Grimison, P, Sandroussi, C, Goodwin, A, Mead, RS, Tucker, K, Andrews, L, Texler, M, Forest, C, Ballal, M, Fletcher, DR, Zeps, N, Nguyen, NQ, Ruszkiewicz, AR, Worthley, C, Chen, J, Brooke-Smith, ME, Papangelis, V, Clouston, AD, Barbour, AP, O’Rourke, TJ, Fawcett, JW, Slater, K, Hatzifotis, M, Hodgkinson, P, Nikfarjam, M, Eshleman, JR, Hruban, RH, Wolfgang, CL, Dixon, J, Scardoni, M, Bassi, C, Grimaldi, S, Cantù, C, Bonizzato, G, Bersani, S, Lakis, V, Lawlor, RT, Newell, F, Patch, AM, Mafficini, A, Sadanandam, A, Koufariotis, LT, Johnston, RL, Leonard, C, Wood, S, Rusev, B, Corbo, V, Luchini, C, Cingarlini, S, Landoni, L, Salvia, R, Milella, M, Chang, D, Bailey, P, Jamieson, NB, Duthie, F, Gingras, MC, Muzny, DM, Wheeler, DA, Gibbs, RA, Milione, M, Chantrill, LA, Timpson, P, Chou, A, Pajic, M, Murphy, A, Dwarte, T, Hermann, D, Vennin, C, Cox, TR, Pereira, B, Ritchie, S, Reed, DA, Chambers, CR, Metcalf, X, Nobis, M, Mukhopadhyay, P, Addala, V, Kazakoff, S, Holmes, O, Xu, Q, Hofmann, O, Samra, JS, Pavlakis, N, Arena, J, High, HA, Asghari, R, Merrett, ND, Pavey, D, Das, A, Cosman, PH, Ismail, K, O’Connnor, C, Stoita, A, Williams, D, Spigellman, A, Lam, VW, McLeod, D, Kirk, J, Kench, JG, Grimison, P, Sandroussi, C, Goodwin, A, Mead, RS, Tucker, K, Andrews, L, Texler, M, Forest, C, Ballal, M, Fletcher, DR, Zeps, N, Nguyen, NQ, Ruszkiewicz, AR, Worthley, C, Chen, J, Brooke-Smith, ME, Papangelis, V, Clouston, AD, Barbour, AP, O’Rourke, TJ, Fawcett, JW, Slater, K, Hatzifotis, M, Hodgkinson, P, Nikfarjam, M, Eshleman, JR, Hruban, RH, Wolfgang, CL, Dixon, J, Scardoni, M, Bassi, C, Grimaldi, S, Cantù, C, Bonizzato, G, and Bersani, S
- Abstract
Here we report the DNA methylation profile of 84 sporadic pancreatic neuroendocrine tumors (PanNETs) with associated clinical and genomic information. We identified three subgroups of PanNETs, termed T1, T2 and T3, with distinct patterns of methylation. The T1 subgroup was enriched for functional tumors and ATRX, DAXX and MEN1 wild-type genotypes. The T2 subgroup contained tumors with mutations in ATRX, DAXX and MEN1 and recurrent patterns of chromosomal losses in half of the genome with no association between regions with recurrent loss and methylation levels. T2 tumors were larger and had lower methylation in the MGMT gene body, which showed positive correlation with gene expression. The T3 subgroup harboured mutations in MEN1 with recurrent loss of chromosome 11, was enriched for grade G1 tumors and showed histological parameters associated with better prognosis. Our results suggest a role for methylation in both driving tumorigenesis and potentially stratifying prognosis in PanNETs.
- Published
- 2021
5. Germline mutation in POLR2A: a heterogeneous, multi-systemic developmental disorder characterized by transcriptional dysregulation.
- Author
-
Hansen, AW, Arora, P, Khayat, MM, Smith, LJ, Lewis, AM, Rossetti, LZ, Jayaseelan, J, Cristian, I, Haynes, D, DiTroia, S, Meeks, N, Delgado, MR, Rosenfeld, JA, Pais, L, White, SM, Meng, Q, Pehlivan, D, Liu, P, Gingras, M-C, Wangler, MF, Muzny, DM, Lupski, JR, Kaplan, CD, Gibbs, RA, Hansen, AW, Arora, P, Khayat, MM, Smith, LJ, Lewis, AM, Rossetti, LZ, Jayaseelan, J, Cristian, I, Haynes, D, DiTroia, S, Meeks, N, Delgado, MR, Rosenfeld, JA, Pais, L, White, SM, Meng, Q, Pehlivan, D, Liu, P, Gingras, M-C, Wangler, MF, Muzny, DM, Lupski, JR, Kaplan, CD, and Gibbs, RA
- Abstract
De novo germline variation in POLR2A was recently reported to associate with a neurodevelopmental disorder. We report twelve individuals harboring putatively pathogenic de novo or inherited variants in POLR2A, detail their phenotypes, and map all known variants to the domain structure of POLR2A and crystal structure of RNA polymerase II. Affected individuals were ascertained from a local data lake, pediatric genetics clinic, and an online community of families of affected individuals. These include six affected by de novo missense variants (including one previously reported individual), four clinical laboratory samples affected by missense variation with unknown inheritance-with yeast functional assays further supporting altered function-one affected by a de novo in-frame deletion, and one affected by a C-terminal frameshift variant inherited from a largely asymptomatic mother. Recurrently observed phenotypes include ataxia, joint hypermobility, short stature, skin abnormalities, congenital cardiac abnormalities, immune system abnormalities, hip dysplasia, and short Achilles tendons. We report a significantly higher occurrence of epilepsy (8/12, 66.7%) than previously reported (3/15, 20%) (p value = 0.014196; chi-square test) and a lower occurrence of hypotonia (8/12, 66.7%) than previously reported (14/15, 93.3%) (p value = 0.076309). POLR2A-related developmental disorders likely represent a spectrum of related, multi-systemic developmental disorders, driven by distinct mechanisms, converging at a single locus.
- Published
- 2021
6. Gene content evolution in the arthropods
- Author
-
Thomas, GWC, Dohmen, E, Hughes, DST, Murali, SC, Poelchau, M, Glastad, K, Anstead, CA, Ayoub, NA, Batterham, P, Bellair, M, Binford, GJ, Chao, H, Chen, YH, Childers, C, Dinh, H, Doddapaneni, HV, Duan, JJ, Dugan, S, Esposito, LA, Friedrich, M, Garb, J, Gasser, RB, Goodisman, MAD, Gundersen-Rindal, DE, Han, Y, Handler, AM, Hatakeyama, M, Hering, L, Hunter, WB, Ioannidis, P, Jayaseelan, JC, Kalra, D, Khila, A, Korhonen, PK, Lee, CE, Lee, SL, Li, Y, Lindsey, ARI, Mayer, G, McGregor, AP, McKenna, DD, Misof, B, Munidasa, M, Munoz-Torres, M, Muzny, DM, Niehuis, O, Osuji-Lacy, N, Palli, SR, Panfilio, KA, Pechmann, M, Perry, T, Peters, RS, Poynton, HC, Prpic, N-M, Qu, J, Rotenberg, D, Schal, C, Schoville, SD, Scully, ED, Skinner, E, Sloan, DB, Stouthamer, R, Strand, MR, Szucsich, NU, Wijeratne, A, Young, ND, Zattara, EE, Benoit, JB, Zdobnov, EM, Pfrender, ME, Hackett, KJ, Werren, JH, Worley, KC, Gibbs, RA, Chipman, AD, Waterhouse, RM, Bornberg-Bauer, E, Hahn, MW, Richards, S, Thomas, GWC, Dohmen, E, Hughes, DST, Murali, SC, Poelchau, M, Glastad, K, Anstead, CA, Ayoub, NA, Batterham, P, Bellair, M, Binford, GJ, Chao, H, Chen, YH, Childers, C, Dinh, H, Doddapaneni, HV, Duan, JJ, Dugan, S, Esposito, LA, Friedrich, M, Garb, J, Gasser, RB, Goodisman, MAD, Gundersen-Rindal, DE, Han, Y, Handler, AM, Hatakeyama, M, Hering, L, Hunter, WB, Ioannidis, P, Jayaseelan, JC, Kalra, D, Khila, A, Korhonen, PK, Lee, CE, Lee, SL, Li, Y, Lindsey, ARI, Mayer, G, McGregor, AP, McKenna, DD, Misof, B, Munidasa, M, Munoz-Torres, M, Muzny, DM, Niehuis, O, Osuji-Lacy, N, Palli, SR, Panfilio, KA, Pechmann, M, Perry, T, Peters, RS, Poynton, HC, Prpic, N-M, Qu, J, Rotenberg, D, Schal, C, Schoville, SD, Scully, ED, Skinner, E, Sloan, DB, Stouthamer, R, Strand, MR, Szucsich, NU, Wijeratne, A, Young, ND, Zattara, EE, Benoit, JB, Zdobnov, EM, Pfrender, ME, Hackett, KJ, Werren, JH, Worley, KC, Gibbs, RA, Chipman, AD, Waterhouse, RM, Bornberg-Bauer, E, Hahn, MW, and Richards, S
- Abstract
BACKGROUND: Arthropods comprise the largest and most diverse phylum on Earth and play vital roles in nearly every ecosystem. Their diversity stems in part from variations on a conserved body plan, resulting from and recorded in adaptive changes in the genome. Dissection of the genomic record of sequence change enables broad questions regarding genome evolution to be addressed, even across hyper-diverse taxa within arthropods. RESULTS: Using 76 whole genome sequences representing 21 orders spanning more than 500 million years of arthropod evolution, we document changes in gene and protein domain content and provide temporal and phylogenetic context for interpreting these innovations. We identify many novel gene families that arose early in the evolution of arthropods and during the diversification of insects into modern orders. We reveal unexpected variation in patterns of DNA methylation across arthropods and examples of gene family and protein domain evolution coincident with the appearance of notable phenotypic and physiological adaptations such as flight, metamorphosis, sociality, and chemoperception. CONCLUSIONS: These analyses demonstrate how large-scale comparative genomics can provide broad new insights into the genotype to phenotype map and generate testable hypotheses about the evolution of animal diversity.
- Published
- 2020
7. Correction: Whole exome sequencing study identifies novel rare and common Alzheimer's-Associated variants involved in immune response and transcriptional regulation.
- Author
-
Bis, JC, Jian, X, Kunkle, BW, Chen, Y, Hamilton-Nelson, KL, Bush, WS, Salerno, WJ, Lancour, D, Ma, Y, Renton, AE, Marcora, E, Farrell, JJ, Zhao, Y, Qu, L, Ahmad, S, Amin, N, Amouyel, P, Beecham, GW, Below, JE, Campion, D, Cantwell, L, Charbonnier, C, Chung, J, Crane, PK, Cruchaga, C, Cupples, LA, Dartigues, J-F, Debette, S, Deleuze, J-F, Fulton, L, Gabriel, SB, Genin, E, Gibbs, RA, Goate, A, Grenier-Boley, B, Gupta, N, Haines, JL, Havulinna, AS, Helisalmi, S, Hiltunen, M, Howrigan, DP, Ikram, MA, Kaprio, J, Konrad, J, Kuzma, A, Lander, ES, Lathrop, M, Lehtimäki, T, Lin, H, Mattila, K, Mayeux, R, Muzny, DM, Nasser, W, Neale, B, Nho, K, Nicolas, G, Patel, D, Pericak-Vance, MA, Perola, M, Psaty, BM, Quenez, O, Rajabli, F, Redon, R, Reitz, C, Remes, AM, Salomaa, V, Sarnowski, C, Schmidt, H, Schmidt, M, Schmidt, R, Soininen, H, Thornton, TA, Tosto, G, Tzourio, C, van der Lee, SJ, van Duijn, CM, Valladares, O, Vardarajan, B, Wang, L-S, Wang, W, Wijsman, E, Wilson, RK, Witten, D, Worley, KC, Zhang, X, Alzheimer’s Disease Sequencing Project, Bellenguez, C, Lambert, J-C, Kurki, MI, Palotie, A, Daly, M, Boerwinkle, E, Lunetta, KL, Destefano, AL, Dupuis, J, Martin, ER, Schellenberg, GD, Seshadri, S, Naj, AC, Fornage, M, Farrer, LA, Bis, JC, Jian, X, Kunkle, BW, Chen, Y, Hamilton-Nelson, KL, Bush, WS, Salerno, WJ, Lancour, D, Ma, Y, Renton, AE, Marcora, E, Farrell, JJ, Zhao, Y, Qu, L, Ahmad, S, Amin, N, Amouyel, P, Beecham, GW, Below, JE, Campion, D, Cantwell, L, Charbonnier, C, Chung, J, Crane, PK, Cruchaga, C, Cupples, LA, Dartigues, J-F, Debette, S, Deleuze, J-F, Fulton, L, Gabriel, SB, Genin, E, Gibbs, RA, Goate, A, Grenier-Boley, B, Gupta, N, Haines, JL, Havulinna, AS, Helisalmi, S, Hiltunen, M, Howrigan, DP, Ikram, MA, Kaprio, J, Konrad, J, Kuzma, A, Lander, ES, Lathrop, M, Lehtimäki, T, Lin, H, Mattila, K, Mayeux, R, Muzny, DM, Nasser, W, Neale, B, Nho, K, Nicolas, G, Patel, D, Pericak-Vance, MA, Perola, M, Psaty, BM, Quenez, O, Rajabli, F, Redon, R, Reitz, C, Remes, AM, Salomaa, V, Sarnowski, C, Schmidt, H, Schmidt, M, Schmidt, R, Soininen, H, Thornton, TA, Tosto, G, Tzourio, C, van der Lee, SJ, van Duijn, CM, Valladares, O, Vardarajan, B, Wang, L-S, Wang, W, Wijsman, E, Wilson, RK, Witten, D, Worley, KC, Zhang, X, Alzheimer’s Disease Sequencing Project, Bellenguez, C, Lambert, J-C, Kurki, MI, Palotie, A, Daly, M, Boerwinkle, E, Lunetta, KL, Destefano, AL, Dupuis, J, Martin, ER, Schellenberg, GD, Seshadri, S, Naj, AC, Fornage, M, and Farrer, LA
- Abstract
A correction to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
8. Spectrum of neurodevelopmental disease associated with the GNAO1 guanosine triphosphate-binding region
- Author
-
Kelly, M, Park, M, Mihalek, I, Rochtus, A, Gramm, M, Perez-Palma, E, Axeen, ET, Hung, CY, Olson, H, Swanson, L, Anselm, I, Briere, LC, High, FA, Sweetser, DA, Kayani, S, Snyder, M, Calvert, S, Scheffer, IE, Yang, E, Waugh, JL, Lal, D, Bodamer, O, Poduri, A, Adams, DR, Aday, A, Alejandro, ME, Allard, P, Ashley, EA, Azamian, MS, Bacino, CA, Baker, E, Balasubramanyam, A, Barseghyan, H, Batzli, GF, Beggs, AH, Behnam, B, Bellen, HJ, Bernstein, JA, Bican, A, Bick, DP, Birch, CL, Bonner, D, Boone, BE, Bostwick, BL, Brokamp, E, Brown, DM, Brush, M, Burke, EA, Burrage, LC, Butte, MJ, Chen, S, Clark, GD, Coakley, TR, Cogan, JD, Colley, HA, Cooper, CM, Cope, H, Craigen, WJ, D'Souza, P, Davids, M, Davidson, JM, Dayal, JG, Dell'Angelica, EC, Dhar, SU, Dipple, KM, Donnell-Fink, LA, Dorrani, N, Dorset, DC, Douine, ED, Draper, DD, Dries, AM, Eckstein, DJ, Emrick, LT, Eng, CM, Enns, G-GM, Eskin, A, Esteves, C, Estwick, T, Fairbrother, L, Fernandez, L, Ferreira, C, Fieg, EL, Fisher, PG, Fogel, BL, Friedman, ND, Gahl, WA, Glanton, E, Godfrey, RA, Goldman, AM, Goldstein, DB, Gould, SE, Gourdine, J-PF, Groden, CA, Gropman, AL, Haendel, M, Hamid, R, Hanchard, NA, High, F, Holm, IA, Horn, J, Howerton, EM, Huang, Y, Jamal, F, Jiang, Y-H, Johnston, JM, Jones, AL, Karaviti, L, Koeller, DM, Kohane, IS, Kohler, JN, Konick, S, Koziura, M, Krasnewich, DM, Krier, JB, Kyle, JE, Lalani, SR, Lau, CC, Lazar, J, LeBlanc, K, Lee, BH, Lee, H, Levy, SE, Lewis, RA, Lincoln, SA, Loo, SK, Loscalzo, J, Maas, RL, Macnamara, EF, MacRae, CA, Maduro, VV, Majch-erska, MM, Malicdan, MC, Mamounas, LA, Manolio, TA, Markello, TC, Marom, R, Martin, MG, Martinez-Agosto, JA, Mar-waha, S, May, T, McConkie-Rosell, A, McCormack, CE, McCray, AF, Merker, JD, Metz, TO, Might, M, Moretti, PM, Morimoto, M, Mulvihill, JJ, Murdock, DR, Murphy, JL, Muzny, DM, Nehrebecky, ME, Nelson, SF, Newberry, JS, Newman, JH, Nicholas, SK, Novacic, D, Orange, JS, Orengo, JP, Pallais, JC, Palmer, CGS, Papp, JC, Parker, NH, Pena, LDM, Phillips, JA, Posey, JE, Postlethwait, JH, Potocki, L, Pusey, BN, Reuter, CM, Rives, L, Robertson, AK, Rodan, LH, Rosenfeld, JA, Sampson, JB, Samson, SL, Schoch, K, Scott, DA, Shakachite, L, Sharma, P, Shashi, V, Signer, R, Silverman, EK, Sinsheimer, JS, Smith, KS, Spillmann, RC, Stoler, JM, Stong, N, Sullivan, JA, Tan, QK-G, Tifft, CJ, Toro, C, Tran, AA, Urv, TK, Vilain, E, Vogel, TP, Waggott, DM, Wahl, CE, Walker, M, Walley, NM, Walsh, CA, Wan, J, Wangler, MF, Ward, PA, Waters, KM, Webb-Robertson, B-JM, Westerfield, M, Wheeler, MT, Wise, AL, Wolfe, LA, Worthey, EA, Yamamoto, S, Yang, Y, Yoon, AJ, Yu, G, Zastrow, DB, Zhao, C, Zheng, A, Kelly, M, Park, M, Mihalek, I, Rochtus, A, Gramm, M, Perez-Palma, E, Axeen, ET, Hung, CY, Olson, H, Swanson, L, Anselm, I, Briere, LC, High, FA, Sweetser, DA, Kayani, S, Snyder, M, Calvert, S, Scheffer, IE, Yang, E, Waugh, JL, Lal, D, Bodamer, O, Poduri, A, Adams, DR, Aday, A, Alejandro, ME, Allard, P, Ashley, EA, Azamian, MS, Bacino, CA, Baker, E, Balasubramanyam, A, Barseghyan, H, Batzli, GF, Beggs, AH, Behnam, B, Bellen, HJ, Bernstein, JA, Bican, A, Bick, DP, Birch, CL, Bonner, D, Boone, BE, Bostwick, BL, Brokamp, E, Brown, DM, Brush, M, Burke, EA, Burrage, LC, Butte, MJ, Chen, S, Clark, GD, Coakley, TR, Cogan, JD, Colley, HA, Cooper, CM, Cope, H, Craigen, WJ, D'Souza, P, Davids, M, Davidson, JM, Dayal, JG, Dell'Angelica, EC, Dhar, SU, Dipple, KM, Donnell-Fink, LA, Dorrani, N, Dorset, DC, Douine, ED, Draper, DD, Dries, AM, Eckstein, DJ, Emrick, LT, Eng, CM, Enns, G-GM, Eskin, A, Esteves, C, Estwick, T, Fairbrother, L, Fernandez, L, Ferreira, C, Fieg, EL, Fisher, PG, Fogel, BL, Friedman, ND, Gahl, WA, Glanton, E, Godfrey, RA, Goldman, AM, Goldstein, DB, Gould, SE, Gourdine, J-PF, Groden, CA, Gropman, AL, Haendel, M, Hamid, R, Hanchard, NA, High, F, Holm, IA, Horn, J, Howerton, EM, Huang, Y, Jamal, F, Jiang, Y-H, Johnston, JM, Jones, AL, Karaviti, L, Koeller, DM, Kohane, IS, Kohler, JN, Konick, S, Koziura, M, Krasnewich, DM, Krier, JB, Kyle, JE, Lalani, SR, Lau, CC, Lazar, J, LeBlanc, K, Lee, BH, Lee, H, Levy, SE, Lewis, RA, Lincoln, SA, Loo, SK, Loscalzo, J, Maas, RL, Macnamara, EF, MacRae, CA, Maduro, VV, Majch-erska, MM, Malicdan, MC, Mamounas, LA, Manolio, TA, Markello, TC, Marom, R, Martin, MG, Martinez-Agosto, JA, Mar-waha, S, May, T, McConkie-Rosell, A, McCormack, CE, McCray, AF, Merker, JD, Metz, TO, Might, M, Moretti, PM, Morimoto, M, Mulvihill, JJ, Murdock, DR, Murphy, JL, Muzny, DM, Nehrebecky, ME, Nelson, SF, Newberry, JS, Newman, JH, Nicholas, SK, Novacic, D, Orange, JS, Orengo, JP, Pallais, JC, Palmer, CGS, Papp, JC, Parker, NH, Pena, LDM, Phillips, JA, Posey, JE, Postlethwait, JH, Potocki, L, Pusey, BN, Reuter, CM, Rives, L, Robertson, AK, Rodan, LH, Rosenfeld, JA, Sampson, JB, Samson, SL, Schoch, K, Scott, DA, Shakachite, L, Sharma, P, Shashi, V, Signer, R, Silverman, EK, Sinsheimer, JS, Smith, KS, Spillmann, RC, Stoler, JM, Stong, N, Sullivan, JA, Tan, QK-G, Tifft, CJ, Toro, C, Tran, AA, Urv, TK, Vilain, E, Vogel, TP, Waggott, DM, Wahl, CE, Walker, M, Walley, NM, Walsh, CA, Wan, J, Wangler, MF, Ward, PA, Waters, KM, Webb-Robertson, B-JM, Westerfield, M, Wheeler, MT, Wise, AL, Wolfe, LA, Worthey, EA, Yamamoto, S, Yang, Y, Yoon, AJ, Yu, G, Zastrow, DB, Zhao, C, and Zheng, A
- Abstract
OBJECTIVE: To characterize the phenotypic spectrum associated with GNAO1 variants and establish genotype-protein structure-phenotype relationships. METHODS: We evaluated the phenotypes of 14 patients with GNAO1 variants, analyzed their variants for potential pathogenicity, and mapped them, along with those in the literature, on a three-dimensional structural protein model. RESULTS: The 14 patients in our cohort, including one sibling pair, had 13 distinct, heterozygous GNAO1 variants classified as pathogenic or likely pathogenic. We attributed the same variant in two siblings to parental mosaicism. Patients initially presented with seizures beginning in the first 3 months of life (8/14), developmental delay (4/14), hypotonia (1/14), or movement disorder (1/14). All patients had hypotonia and developmental delay ranging from mild to severe. Nine had epilepsy, and nine had movement disorders, including dystonia, ataxia, chorea, and dyskinesia. The 13 GNAO1 variants in our patients are predicted to result in amino acid substitutions or deletions in the GNAO1 guanosine triphosphate (GTP)-binding region, analogous to those in previous publications. Patients with variants affecting amino acids 207-221 had only movement disorder and hypotonia. Patients with variants affecting the C-terminal region had the mildest phenotypes. SIGNIFICANCE: GNAO1 encephalopathy most frequently presents with seizures beginning in the first 3 months of life. Concurrent movement disorders are also a prominent feature in the spectrum of GNAO1 encephalopathy. All variants affected the GTP-binding domain of GNAO1, highlighting the importance of this region for G-protein signaling and neurodevelopment.
- Published
- 2019
9. Exome Sequencing of a Primary Ovarian Insufficiency Cohort Reveals Common Molecular Etiologies for a Spectrum of Disease
- Author
-
Jolly, A, Bayram, Y, Turan, S, Aycan, Z, Tos, T, Abali, Zy, Hacihamdioglu, B, Akdemir, Zhc, Hijazi, H, Bas, S, Atay, Z, Guran, T, Abali, S, Bas, F, Darendeliler, F, Colombo, Roberto, Barakat, T, Rinne, T, White, Jj, Yesil, G, Gezdirici, A, Gulec, Ey, Karaca, E, Pehlivan, D, Jhangiani, Sn, Muzny, Dm, Poyrazoglu, S, Bereket, A, Gibbs, Ra, Posey, Je, Lupski, Jr, Colombo, R (ORCID:0000-0003-0482-7542), Jolly, A, Bayram, Y, Turan, S, Aycan, Z, Tos, T, Abali, Zy, Hacihamdioglu, B, Akdemir, Zhc, Hijazi, H, Bas, S, Atay, Z, Guran, T, Abali, S, Bas, F, Darendeliler, F, Colombo, Roberto, Barakat, T, Rinne, T, White, Jj, Yesil, G, Gezdirici, A, Gulec, Ey, Karaca, E, Pehlivan, D, Jhangiani, Sn, Muzny, Dm, Poyrazoglu, S, Bereket, A, Gibbs, Ra, Posey, Je, Lupski, Jr, and Colombo, R (ORCID:0000-0003-0482-7542)
- Abstract
Context: Primary ovarian insufficiency (POI) encompasses a spectrum of premature menopause, including both primary and secondary amenorrhea. For 75% to 90% of individuals with hyper-gonadotropic hypogonadism presenting as POI, the molecular etiology is unknown. Common etiologies include chromosomal abnormalities, environmental factors, and congenital disorders affecting ovarian development and function, as well as syndromic and nonsyndromic single gene disorders suggesting POI represents a complex trait.Objective: To characterize the contribution of known disease genes to POI and identify molecular etiologies and biological underpinnings of POI.Design, Setting, and Participants: We applied exome sequencing (ES) and family-based genomics to 42 affected female individuals from 36 unrelated Turkish families, including 31 with reported parental consanguinity.Results: This analysis identified likely damaging, potentially contributing variants and molecular diagnoses in 16 families (44%), including 11 families with likely damaging variants in known genes and five families with predicted deleterious variants in disease genes (IGSF10, MND1, MRPS22, and SOHLH1) not previously associated with POI. Of the 16 families, 2 (13%) had evidence for potentially pathogenic variants at more than one locus. Absence of heterozygosity consistent with identity-by-descent mediated recessive disease burden contributes to molecular diagnosis in 15 of 16 (94%) families. GeneMatcher allowed identification of additional families from diverse genetic backgrounds.Conclusions: ES analysis of a POI cohort further characterized locus heterogeneity, reaffirmed the association of genes integral to meiotic recombination, demonstrated the likely contribution of genes involved in hypothalamic development, and documented multilocus pathogenic variation suggesting the potential for oligogenic inheritance contributing to the development of POI.
- Published
- 2019
10. Unraveling genetic predisposition to familial or early onset gastric cancer using germline whole-exome sequencing
- Author
-
Vogelaar, IP, van der Post, RS, van Krieken, J, Spruijt, L, van Zelst-Stams, W A G, Kets, CM, Lubinski, J, Jakubowska, A, Teodorczyk, U, Aalfs, CM, van Hest, LP (Liselotte), Pinheiro, H, Oliveira, C, Jhangiani, SN, Muzny, DM, Gibbs, RA, Lupski, JR, de Ligt, J, Vissers, L, Hoischen, A, Gilissen, C, van de Vorst, M, Goeman, JJ, Schackert, HK, Ranzani, GN, Molinaro, V, Garcia, EBG, Hes, FJ, Holinski-Feder, E, Genuardi, M, Ausems, M, Sijmons, RH, Wagner, Anja, van der Kolk, LE, Bjornevoll, I, Hoberg-Vetti, H, van Kessel, AG, Kuiper, RP (Roland), Ligtenberg, MJL, Hoogerbrugge, N, Vogelaar, IP, van der Post, RS, van Krieken, J, Spruijt, L, van Zelst-Stams, W A G, Kets, CM, Lubinski, J, Jakubowska, A, Teodorczyk, U, Aalfs, CM, van Hest, LP (Liselotte), Pinheiro, H, Oliveira, C, Jhangiani, SN, Muzny, DM, Gibbs, RA, Lupski, JR, de Ligt, J, Vissers, L, Hoischen, A, Gilissen, C, van de Vorst, M, Goeman, JJ, Schackert, HK, Ranzani, GN, Molinaro, V, Garcia, EBG, Hes, FJ, Holinski-Feder, E, Genuardi, M, Ausems, M, Sijmons, RH, Wagner, Anja, van der Kolk, LE, Bjornevoll, I, Hoberg-Vetti, H, van Kessel, AG, Kuiper, RP (Roland), Ligtenberg, MJL, and Hoogerbrugge, N
- Published
- 2017
11. Initial sequencing and analysis of the human genome
- Author
-
Lander, ES, Linton, LM, Birren, B, Nusbaum, C, Zody, MC, Baldwin, J, Devon, K, Dewar, K, Doyle, M, FitzHugh, W, Funke, R, Gage, D, Harris, K, Heaford, A, Howland, J, Kann, L, Lehoczky, J, LeVine, R, McEwan, P, McKernan, K, Meldrim, J, Mesirov, JP, Miranda, C, Morris, W, Naylor, J, Raymond, C, Rosetti, M, Santos, R, Sheridan, A, Sougnez, C, Stange-Thomann, N, Stojanovic, N, Subramanian, A, Wyman, D, Rogers, J, Sulston, J, Ainscough, R, Beck, S, Bentley, D, Burton, J, Clee, C, Carter, N, Coulson, A, Deadman, R, Deloukas, P, Dunham, A, Dunham, I, Durbin, R, French, L, Grafham, D, Gregory, S, Hubbard, T, Humphray, S, Hunt, A, Jones, M, Lloyd, C, McMurray, A, Matthews, L, Mercer, S, Milne, S, Mullikin, JC, Mungall, A, Plumb, R, Ross, M, Shownkeen, R, Sims, S, Waterston, RH, Wilson, RK, Hillier, LW, McPherson, JD, Marra, MA, Mardis, ER, Fulton, LA, Chinwalla, AT, Pepin, KH, Gish, WR, Chissoe, SL, Wendl, MC, Delehaunty, KD, Miner, TL, Delehaunty, A, Kramer, JB, Cook, LL, Fulton, RS, Johnson, DL, Minx, PJ, Clifton, SW, Hawkins, T, Branscomb, E, Predki, P, Richardson, P, Wenning, S, Slezak, T, Doggett, N, Cheng, JF, Olsen, A, Lucas, S, Elkin, C, Uberbacher, E, Frazier, M, Gibbs, RA, Muzny, DM, Scherer, SE, Bouck, JB, Sodergren, EJ, Worley, KC, Rives, CM, Gorrell, JH, Metzker, ML, Naylor, SL, Kucherlapati, RS, Nelson, DL, Weinstock, GM, Sakaki, Y, Fujiyama, A, Hattori, M, Yada, T, Toyoda, A, Itoh, T, Kawagoe, C, Watanabe, H, Totoki, Y, Taylor, T, Weissenbach, J, Heilig, R, Saurin, W, Artiguenave, F, Brottier, P, Bruls, T, Pelletier, E, Robert, C, Wincker, P, Smith, DR, Doucette-Stamm, L, Rubenfield, M, Weinstock, K, Lee, HM, Dubois, J, Rosenthal, A, Platzer, M, Nyakatura, G, Taudien, S, Rump, A, Yang, H, Yu, J, Wang, J, Huang, G, Gu, J, Hood, L, Rowen, L, Madan, A, Qin, S, Davis, RW, Federspiel, NA, Abola, AP, Proctor, MJ, Myers, RM, Schmutz, J, Dickson, M, Grimwood, J, Cox, DR, Olson, MV, Kaul, R, Shimizu, N, Kawasaki, K, Minoshima, S, Evans, GA, Athanasiou, M, Schultz, R, Roe, BA, Chen, F, Pan, H, Ramser, J, Lehrach, H, Reinhardt, R, McCombie, WR, de la Bastide, M, Dedhia, N, Blöcker, H, Hornischer, K, Nordsiek, G, Agarwala, R, Aravind, L, Bailey, JA, Bateman, A, Batzoglou, S, Birney, E, Bork, P, Brown, DG, Burge, CB, Cerutti, L, Chen, HC, Church, D, Clamp, M, Copley, RR, Doerks, T, Eddy, SR, Eichler, EE, Furey, TS, Galagan, J, Gilbert, JG, Harmon, C, Hayashizaki, Y, Haussler, D, Hermjakob, H, Hokamp, K, Jang, W, Johnson, LS, Jones, TA, Kasif, S, Kaspryzk, A, Kennedy, S, Kent, WJ, Kitts, P, Koonin, EV, Korf, I, Kulp, D, Lancet, D, Lowe, TM, McLysaght, A, Mikkelsen, T, Moran, JV, Mulder, N, Pollara, VJ, Ponting, CP, Schuler, G, Schultz, J, Slater, G, Smit, AF, Stupka, E, Szustakowski, J, Thierry-Mieg, D, Thierry-Mieg, J, Wagner, L, Wallis, J, Wheeler, R, Williams, A, Wolf, YI, Wolfe, KH, Yang, SP, Yeh, RF, Collins, F, Guyer, MS, Peterson, J, Felsenfeld, A, Wetterstrand, KA, Patrinos, A, Morgan, MJ, de Jong, P, Catanese, JJ, Osoegawa, K, Shizuya, H, Choi, S, Chen, YJ, and Szustakowki, J
- Subjects
Genetics ,Cancer genome sequencing ,Chimpanzee genome project ,Multidisciplinary ,Cancer Genome Project ,Gene density ,DNA sequencing theory ,Hybrid genome assembly ,Computational biology ,Biology ,Genome ,Personal genomics - Abstract
The human genome holds an extraordinary trove of information about human development, physiology, medicine and evolution. Here we report the results of an international collaboration to produce and make freely available a draft sequence of the human genome. We also present an initial analysis of the data, describing some of the insights that can be gleaned from the sequence.
- Published
- 2016
- Full Text
- View/download PDF
12. Identification of Intellectual Disability Genes in Female Patients with a Skewed X-Inactivation Pattern
- Author
-
Fieremans N, Van Esch H, Holvoet M, Van Goethem G, Devriendt K, Rosello M, Mayo S, Martinez F, Jhangiani S, Muzny DM, Gibbs RA, Lupski JR, Vermeesch JR, Marynen P, and Froyen G
- Subjects
escape genes ,skewing of X-inactivation ,intellectual disability ,exome sequencing - Abstract
Intellectual disability (ID) is a heterogeneous disorder with an unknown molecular etiology in many cases. Previously, X-linked ID (XLID) studies focused on males because of the hemizygous state of their X chromosome. Carrier females are generally unaffected because of the presence of a second normal allele, or inactivation of the mutant X chromosome in most of their cells (skewing). However, in female ID patients, we hypothesized that the presence of skewing of X-inactivation would be an indicator for an X chromosomal ID cause. We analyzed the X-inactivation patterns of 288 females with ID, and found that 22 (7.6%) had extreme skewing (>90%), which is significantly higher than observed in the general population (3.6%; P=0.029). Whole-exome sequencing of 19 females with extreme skewing revealed causal variants in six females in the XLID genes DDX3X, NHS, WDR45, MECP2, and SMC1A. Interestingly, variants in genes escaping X-inactivation presumably cause both XLID and skewing of X-inactivation in three of these patients. Moreover, variants likely accounting for skewing only were detected in MED12, HDAC8, and TAF9B. All tested candidate causative variants were de novo events. Hence, extreme skewing is a good indicator for the presence of X-linked variants in female patients.
- Published
- 2016
13. Lucilia cuprina genome unlocks parasitic fly biology to underpin future interventions
- Author
-
Anstead, CA, Korhonen, PK, Young, ND, Hall, RS, Jex, AR, Murali, SC, Hughes, DST, Lee, SF, Perry, T, Stroehlein, AJ, Ansell, BRE, Breugelmans, B, Hofmann, A, Qu, J, Dugan, S, Lee, SL, Chao, H, Dinh, H, Han, Y, Doddapaneni, HV, Worley, KC, Muzny, DM, Ioannidis, P, Waterhouse, RM, Zdobnov, EM, James, PJ, Bagnall, NH, Kotze, AC, Gibbs, RA, Richards, S, Batterham, P, Gasser, RB, Anstead, CA, Korhonen, PK, Young, ND, Hall, RS, Jex, AR, Murali, SC, Hughes, DST, Lee, SF, Perry, T, Stroehlein, AJ, Ansell, BRE, Breugelmans, B, Hofmann, A, Qu, J, Dugan, S, Lee, SL, Chao, H, Dinh, H, Han, Y, Doddapaneni, HV, Worley, KC, Muzny, DM, Ioannidis, P, Waterhouse, RM, Zdobnov, EM, James, PJ, Bagnall, NH, Kotze, AC, Gibbs, RA, Richards, S, Batterham, P, and Gasser, RB
- Abstract
Lucilia cuprina is a parasitic fly of major economic importance worldwide. Larvae of this fly invade their animal host, feed on tissues and excretions and progressively cause severe skin disease (myiasis). Here we report the sequence and annotation of the 458-megabase draft genome of Lucilia cuprina. Analyses of this genome and the 14,544 predicted protein-encoding genes provide unique insights into the fly's molecular biology, interactions with the host animal and insecticide resistance. These insights have broad implications for designing new methods for the prevention and control of myiasis.
- Published
- 2015
14. The genomes of two key bumblebee species with primitive eusocial organization.
- Author
-
Sadd, BM, Barribeau, SM, Bloch, G, de Graaf, DC, Dearden, P, Elsik, CG, Gadau, J, Grimmelikhuijzen, CJP, Hasselmann, M, Lozier, JD, Robertson, HM, Smagghe, G, Stolle, E, Van Vaerenbergh, M, Waterhouse, RM, Bornberg-Bauer, E, Klasberg, S, Bennett, AK, Câmara, F, Guigó, R, Hoff, K, Mariotti, M, Munoz-Torres, M, Murphy, T, Santesmasses, D, Amdam, GV, Beckers, M, Beye, M, Biewer, M, Bitondi, MMG, Blaxter, ML, Bourke, AFG, Brown, MJF, Buechel, SD, Cameron, R, Cappelle, K, Carolan, JC, Christiaens, O, Ciborowski, KL, Clarke, DF, Colgan, TJ, Collins, DH, Cridge, AG, Dalmay, T, Dreier, S, du Plessis, L, Duncan, E, Erler, S, Evans, J, Falcon, T, Flores, K, Freitas, FCP, Fuchikawa, T, Gempe, T, Hartfelder, K, Hauser, F, Helbing, S, Humann, FC, Irvine, F, Jermiin, LS, Johnson, CE, Johnson, RM, Jones, AK, Kadowaki, T, Kidner, JH, Koch, V, Köhler, A, Kraus, FB, Lattorff, HMG, Leask, M, Lockett, GA, Mallon, EB, Antonio, DSM, Marxer, M, Meeus, I, Moritz, RFA, Nair, A, Näpflin, K, Nissen, I, Niu, J, Nunes, FMF, Oakeshott, JG, Osborne, A, Otte, M, Pinheiro, DG, Rossié, N, Rueppell, O, Santos, CG, Schmid-Hempel, R, Schmitt, BD, Schulte, C, Simões, ZLP, Soares, MPM, Swevers, L, Winnebeck, EC, Wolschin, F, Yu, N, Zdobnov, EM, Aqrawi, PK, Blankenburg, KP, Coyle, M, Francisco, L, Hernandez, AG, Holder, M, Hudson, ME, Jackson, L, Jayaseelan, J, Joshi, V, Kovar, C, Lee, SL, Mata, R, Mathew, T, Newsham, IF, Ngo, R, Okwuonu, G, Pham, C, Pu, L-L, Saada, N, Santibanez, J, Simmons, D, Thornton, R, Venkat, A, Walden, KKO, Wu, Y-Q, Debyser, G, Devreese, B, Asher, C, Blommaert, J, Chipman, AD, Chittka, L, Fouks, B, Liu, J, O'Neill, MP, Sumner, S, Puiu, D, Qu, J, Salzberg, SL, Scherer, SE, Muzny, DM, Richards, S, Robinson, GE, Gibbs, RA, Schmid-Hempel, P, Worley, KC, Sadd, BM, Barribeau, SM, Bloch, G, de Graaf, DC, Dearden, P, Elsik, CG, Gadau, J, Grimmelikhuijzen, CJP, Hasselmann, M, Lozier, JD, Robertson, HM, Smagghe, G, Stolle, E, Van Vaerenbergh, M, Waterhouse, RM, Bornberg-Bauer, E, Klasberg, S, Bennett, AK, Câmara, F, Guigó, R, Hoff, K, Mariotti, M, Munoz-Torres, M, Murphy, T, Santesmasses, D, Amdam, GV, Beckers, M, Beye, M, Biewer, M, Bitondi, MMG, Blaxter, ML, Bourke, AFG, Brown, MJF, Buechel, SD, Cameron, R, Cappelle, K, Carolan, JC, Christiaens, O, Ciborowski, KL, Clarke, DF, Colgan, TJ, Collins, DH, Cridge, AG, Dalmay, T, Dreier, S, du Plessis, L, Duncan, E, Erler, S, Evans, J, Falcon, T, Flores, K, Freitas, FCP, Fuchikawa, T, Gempe, T, Hartfelder, K, Hauser, F, Helbing, S, Humann, FC, Irvine, F, Jermiin, LS, Johnson, CE, Johnson, RM, Jones, AK, Kadowaki, T, Kidner, JH, Koch, V, Köhler, A, Kraus, FB, Lattorff, HMG, Leask, M, Lockett, GA, Mallon, EB, Antonio, DSM, Marxer, M, Meeus, I, Moritz, RFA, Nair, A, Näpflin, K, Nissen, I, Niu, J, Nunes, FMF, Oakeshott, JG, Osborne, A, Otte, M, Pinheiro, DG, Rossié, N, Rueppell, O, Santos, CG, Schmid-Hempel, R, Schmitt, BD, Schulte, C, Simões, ZLP, Soares, MPM, Swevers, L, Winnebeck, EC, Wolschin, F, Yu, N, Zdobnov, EM, Aqrawi, PK, Blankenburg, KP, Coyle, M, Francisco, L, Hernandez, AG, Holder, M, Hudson, ME, Jackson, L, Jayaseelan, J, Joshi, V, Kovar, C, Lee, SL, Mata, R, Mathew, T, Newsham, IF, Ngo, R, Okwuonu, G, Pham, C, Pu, L-L, Saada, N, Santibanez, J, Simmons, D, Thornton, R, Venkat, A, Walden, KKO, Wu, Y-Q, Debyser, G, Devreese, B, Asher, C, Blommaert, J, Chipman, AD, Chittka, L, Fouks, B, Liu, J, O'Neill, MP, Sumner, S, Puiu, D, Qu, J, Salzberg, SL, Scherer, SE, Muzny, DM, Richards, S, Robinson, GE, Gibbs, RA, Schmid-Hempel, P, and Worley, KC
- Abstract
BACKGROUND: The shift from solitary to social behavior is one of the major evolutionary transitions. Primitively eusocial bumblebees are uniquely placed to illuminate the evolution of highly eusocial insect societies. Bumblebees are also invaluable natural and agricultural pollinators, and there is widespread concern over recent population declines in some species. High-quality genomic data will inform key aspects of bumblebee biology, including susceptibility to implicated population viability threats. RESULTS: We report the high quality draft genome sequences of Bombus terrestris and Bombus impatiens, two ecologically dominant bumblebees and widely utilized study species. Comparing these new genomes to those of the highly eusocial honeybee Apis mellifera and other Hymenoptera, we identify deeply conserved similarities, as well as novelties key to the biology of these organisms. Some honeybee genome features thought to underpin advanced eusociality are also present in bumblebees, indicating an earlier evolution in the bee lineage. Xenobiotic detoxification and immune genes are similarly depauperate in bumblebees and honeybees, and multiple categories of genes linked to social organization, including development and behavior, show high conservation. Key differences identified include a bias in bumblebee chemoreception towards gustation from olfaction, and striking differences in microRNAs, potentially responsible for gene regulation underlying social and other traits. CONCLUSIONS: These two bumblebee genomes provide a foundation for post-genomic research on these key pollinators and insect societies. Overall, gene repertoires suggest that the route to advanced eusociality in bees was mediated by many small changes in many genes and processes, and not by notable expansion or depauperation.
- Published
- 2015
15. Low-frequency and rare exome chip variants associate with fasting glucose and type 2 diabetes susceptibility
- Author
-
Wessel, J, Chu, AY, Willems, SM, Wang, S, Yaghootkar, H, Brody, JA, Dauriz, M, Hivert, MF, Raghavan, S, Lipovich, L, Hidalgo, B, Fox, K, Huffman, JE, An, P, Lu, YC, Rasmussen-Torvik, LJ, Grarup, N, Ehm, MG, Li, Li, Baldridge, AS, Stancakova, A, Abrol, R, Besse, CL, Boland, A, Bork-Jensen, J, Fornage, M, Freitag, DF, Garcia, ME, Guo, XQ, Hara, K, Isaacs, Aaron, Jakobsdottir, J, Lange, LA, Layton, JC, Li, M, Zhao, JH, Meidtner, K, Morrison, AC, Nalls, MA, Peters, Marjolein, Sabater-Lleal, M, Schurmann, C, Silveira, A, Smith, AV, Southam, L, Stoiber, MH, Strawbridge, RJ, Taylor, KD, Varga, TV, Allin, KH, Amin, Najaf, Aponte, JL, Aung, T, Barbieri, C, Bihlmeyer, NA, Boehnke, M, Bombieri, C, Bowden, DW, Burns, SM, Chen, YN, Chen, YD, Cheng, CY (Ching-Yu), Correa, A, Czajkowski, J, Dehghan, Abbas, Ehret, GB, Eiriksdottir, G, Escher, SA, Farmaki, AE, Franberg, M, Gambaro, G, Giulianini, F, Goddard, WA, Goel, A, Gottesman, O, Grove, ML, Gustafsson, S, Hai, Y, Hallmans, G, Heo, J, Hoffmann, P, Ikram, MK, Jensen, RA, Jorgensen, ME, Jorgensen, T, Karaleftheri, M, Khor, CC, Kirkpatrick, A, Kraja, AT, Kuusisto, J, Lange, Edmee, Lee, IT, Lee, WJ, Leong, A, Liao, JM, Liu, CY, Liu, YM, Lindgren, CM, Linneberg, A, Malerba, G, Mamakou, V, Marouli, E, Maruthur, NM, Matchan, A, McKean-Cowdin, R, McLeod, O, Metcalf, GA, Mohlke, KL, Muzny, DM, Ntalla, I, Palmer, ND, Pasko, D, Peter, A, Rayner, NW, Renstrom, F, Rice, K, Sala, CF, Sennblad, B, Serafetinidis, I, Smith, JA, Soranzo, N, Speliotes, EK, Stahl, EA, Stirrups, K, Tentolouris, N, Thanopoulou, A, Torres, M, Traglia, M, Tsafantakis, E, Javad, S, Yanek, LR, Zengini, E, Becker, DM, Bis, JC, Brown, JB, Cupples, LA, Hansen, T, Ingelsson, E, Karter, AJ, Lorenzo, C, Mathias, RA, Norris, JM, Peloso, GM, Sheu, WHH, Toniolo, D, Vaidya, D, Varma, R, Wagenknecht, LE, Boeing, H, Bottinger, EP, Dedoussis, G, Deloukas, P, Ferrannini, E, Franco Duran, OH, Franks, PW, Gibbs, RA, Gudnason, V, Hamsten, A, Harris, TB, Hattersley, AT, Hayward, C, Hofman, Bert, Jansson, JH, Langenberg, C, Launer, LJ (Lenore), Levy, D, Oostra, Ben, O'Donnell, CJ, O'Rahilly, S, Padmanabhan, S, Pankow, JS, Polasek, O, Province, MA, Rich, SS, Ridker, PM, Rudan, I, Schulze, MB, Smith, BH, Uitterlinden, André, Walker, M, Watkins, H, Wong, TY (Tien Yin), Zeggini, E, Laakso, M, Borecki, IB, Chasman, DI, Pedersen, O, Psaty, BM, Tai, ES, Duijn, Cornelia, Wareham, NJ, Waterworth, DM, Boerwinkle, E, Kao, WHL, Florez, JC, Loos, RJF, Wilson, JG, Frayling, TM, Siscovick, DS, Dupuis, J, Rotter, JI, Meigs, JB, Scott, RA, Goodarzi, MO, Sharp, SJ, Forouhi, NG, Kerrison, ND, Lucarelli, DM, Sims, M, Barroso, I, McCarthy, MI, Arriola, L, Balkau, B, Barricarte, A, Gonzalez, C, Grioni, S, Kaaks, R, Key, TJ, Navarro, C, Nilsson, PM, Overvad, K, Palli, D, Panico, S, Quiros, JR, Rolandsson, O, Sacerdote, C, Sanchez, MJ (Maria-Jose), Slimani, N, Tjonneland, A, Tumino, R, van der A, DL, van der Schouw, YT, Riboli, E, Wessel, J, Chu, AY, Willems, SM, Wang, S, Yaghootkar, H, Brody, JA, Dauriz, M, Hivert, MF, Raghavan, S, Lipovich, L, Hidalgo, B, Fox, K, Huffman, JE, An, P, Lu, YC, Rasmussen-Torvik, LJ, Grarup, N, Ehm, MG, Li, Li, Baldridge, AS, Stancakova, A, Abrol, R, Besse, CL, Boland, A, Bork-Jensen, J, Fornage, M, Freitag, DF, Garcia, ME, Guo, XQ, Hara, K, Isaacs, Aaron, Jakobsdottir, J, Lange, LA, Layton, JC, Li, M, Zhao, JH, Meidtner, K, Morrison, AC, Nalls, MA, Peters, Marjolein, Sabater-Lleal, M, Schurmann, C, Silveira, A, Smith, AV, Southam, L, Stoiber, MH, Strawbridge, RJ, Taylor, KD, Varga, TV, Allin, KH, Amin, Najaf, Aponte, JL, Aung, T, Barbieri, C, Bihlmeyer, NA, Boehnke, M, Bombieri, C, Bowden, DW, Burns, SM, Chen, YN, Chen, YD, Cheng, CY (Ching-Yu), Correa, A, Czajkowski, J, Dehghan, Abbas, Ehret, GB, Eiriksdottir, G, Escher, SA, Farmaki, AE, Franberg, M, Gambaro, G, Giulianini, F, Goddard, WA, Goel, A, Gottesman, O, Grove, ML, Gustafsson, S, Hai, Y, Hallmans, G, Heo, J, Hoffmann, P, Ikram, MK, Jensen, RA, Jorgensen, ME, Jorgensen, T, Karaleftheri, M, Khor, CC, Kirkpatrick, A, Kraja, AT, Kuusisto, J, Lange, Edmee, Lee, IT, Lee, WJ, Leong, A, Liao, JM, Liu, CY, Liu, YM, Lindgren, CM, Linneberg, A, Malerba, G, Mamakou, V, Marouli, E, Maruthur, NM, Matchan, A, McKean-Cowdin, R, McLeod, O, Metcalf, GA, Mohlke, KL, Muzny, DM, Ntalla, I, Palmer, ND, Pasko, D, Peter, A, Rayner, NW, Renstrom, F, Rice, K, Sala, CF, Sennblad, B, Serafetinidis, I, Smith, JA, Soranzo, N, Speliotes, EK, Stahl, EA, Stirrups, K, Tentolouris, N, Thanopoulou, A, Torres, M, Traglia, M, Tsafantakis, E, Javad, S, Yanek, LR, Zengini, E, Becker, DM, Bis, JC, Brown, JB, Cupples, LA, Hansen, T, Ingelsson, E, Karter, AJ, Lorenzo, C, Mathias, RA, Norris, JM, Peloso, GM, Sheu, WHH, Toniolo, D, Vaidya, D, Varma, R, Wagenknecht, LE, Boeing, H, Bottinger, EP, Dedoussis, G, Deloukas, P, Ferrannini, E, Franco Duran, OH, Franks, PW, Gibbs, RA, Gudnason, V, Hamsten, A, Harris, TB, Hattersley, AT, Hayward, C, Hofman, Bert, Jansson, JH, Langenberg, C, Launer, LJ (Lenore), Levy, D, Oostra, Ben, O'Donnell, CJ, O'Rahilly, S, Padmanabhan, S, Pankow, JS, Polasek, O, Province, MA, Rich, SS, Ridker, PM, Rudan, I, Schulze, MB, Smith, BH, Uitterlinden, André, Walker, M, Watkins, H, Wong, TY (Tien Yin), Zeggini, E, Laakso, M, Borecki, IB, Chasman, DI, Pedersen, O, Psaty, BM, Tai, ES, Duijn, Cornelia, Wareham, NJ, Waterworth, DM, Boerwinkle, E, Kao, WHL, Florez, JC, Loos, RJF, Wilson, JG, Frayling, TM, Siscovick, DS, Dupuis, J, Rotter, JI, Meigs, JB, Scott, RA, Goodarzi, MO, Sharp, SJ, Forouhi, NG, Kerrison, ND, Lucarelli, DM, Sims, M, Barroso, I, McCarthy, MI, Arriola, L, Balkau, B, Barricarte, A, Gonzalez, C, Grioni, S, Kaaks, R, Key, TJ, Navarro, C, Nilsson, PM, Overvad, K, Palli, D, Panico, S, Quiros, JR, Rolandsson, O, Sacerdote, C, Sanchez, MJ (Maria-Jose), Slimani, N, Tjonneland, A, Tumino, R, van der A, DL, van der Schouw, YT, and Riboli, E
- Abstract
Fasting glucose and insulin are intermediate traits for type 2 diabetes. Here we explore the role of coding variation on these traits by analysis of variants on the HumanExome BeadChip in 60,564 non-diabetic individuals and in 16,491 T2D cases and 81,877 controls. We identify a novel association of a low-frequency nonsynonymous SNV in GLP1R (A316T; rs10305492; MAF = 1.4%) with lower FG (beta = -0.09 +/- 0.01 mmol l(-1), P = 3.4 x 10(-12)), T2D risk (OR[95% CI] = 0.86[0.76-0.96], P = 0.010), early insulin secretion (beta = -0.07 +/- 0.035 pmol(insulin) mmol(glucose)(-1), P = 0.048), but higher 2-h glucose (beta = 0.16 +/- 0.05 mmol l(-1), P = 4.3 x 10(-4)). We identify a gene-based association with FG at G6PC2 (p(SKAT) = 6.8 x 10(-6)) driven by four rare protein-coding SNVs (H177Y, Y207S, R283X and S324P). We identify rs651007 (MAF = 20%) in the first intron of ABO at the putative promoter of an antisense lncRNA, associating with higher FG (beta = 0.02 +/- 0.004 mmol l(-1), P = 1.3 x 10(-8)). Our approach identifies novel coding variant associations and extends the allelic spectrum of variation underlying diabetes-related quantitative traits and T2D susceptibility.
- Published
- 2015
16. Integrated genomic analyses of ovarian carcinoma
- Author
-
Bell, D, Berchuck, A, Birrer, M, Chien, J, Cramer, DW, Dao, F, Dhir, R, DiSaia, P, Gabra, H, Glenn, P, Godwin, AK, Triche, T, Berman, BP, Van den Berg, DJ, Buckley, J, Baylin, SB, Zhang, J, Spellman, PT, Purdom, E, Iacocca, M, Shelton, T, Voet, D, Neuvial, P, Bengtsson, H, Jakkula, LR, Durinck, S, Han, J, Dorton, S, Marr, H, Zhang, H, Choi, YG, Wang, V, Wilkinson, J, Nguyen, H, Wang, NJ, Imielinski, M, Ngai, J, Conboy, JG, Parvin, B, Feiler, HS, Speed, TP, Gray, JW, Wu, CJ, Bloom, T, Levine, DA, Li, L, Socci, ND, Liang, Y, Taylor, BS, Kalloger, S, Schultz, N, Borsu, L, Lash, AE, Brennan, C, Ardlie, K, Viale, A, Shukla, S, Grimm, D, Sander, C, Ladanyi, M, Hoadley, KA, Meng, S, Du, Y, Karlan, BY, Shi, Y, Fennell, T, Cibulskis, K, Lawrence, MS, Meyerson, M, Hatfield, M, Mills, GB, Sivachenko, A, Jing, R, Park, RW, Liu, Y, Park, PJ, Ramos, AH, Noble, M, Chin, L, Carter, H, Kim, D, Morris, S, Winckler, W, Karchin, R, Morrison, C, Baldwin, J, Korkola, JE, Yena, P, Heiser, LM, Getz, G, Cho, RJ, Hu, Z, Gabriel, S, Mutch, D, Cerami, E, Rhodes, P, Olshen, A, Verhaak, RGW, Lander, ES, Reva, B, Antipin, Y, Shen, R, Olvera, N, Mankoo, P, Sheridan, R, Ciriello, G, Sherman, M, Chang, WK, Bernanke, JA, Hayes, DN, Carter, SL, Haussler, D, Orsulic, S, Benz, CC, Paulauskis, J, Stuart, JM, Zhang, N, Benz, SC, Sanborn, JZ, Vaske, CJ, Mermel, CH, Zhu, J, Szeto, C, Scott, GK, Yau, C, Rabeno, B, Ding, L, Park, K, Balu, S, Perou, CM, Saksena, G, Wilkerson, MD, Millis, S, Kahn, A, Turman, YJ, Fulton, RS, Onofrio, RC, Greene, JM, Sfeir, R, Jensen, MA, Chen, J, Whitmore, J, Alonso, S, Jordan, J, Chu, A, Rader, JS, Koboldt, DC, Zang, D, Gross, J, Barker, A, Compton, C, Eley, G, Ferguson, M, Fielding, P, Gerhard, DS, Myles, R, McLellan, MD, Schaefer, C, Helms, EB, Shaw, KRM, Sikic, BI, Vaught, J, Vockley, JB, Good, PJ, Guyer, MS, Ozenberger, B, Wylie, T, Peterson, J, Thomson, E, Smith-McCune, K, Sood, AK, Bowtell, D, Hubbard, D, Penny, R, Testa, JR, Chang, K, Walker, J, Dinh, HH, Drummond, JA, Fowler, G, Zhou, X, Gunaratne, P, Hawes, AC, Kovar, CL, Lewis, LR, Gupta, S, Morgan, MB, O'Laughlin, M, Newsham, IF, Santibanez, J, Reid, JG, Trevino, LR, Wu, J, Wu, Y-Q, Wang, M, Muzny, DM, Wheeler, DA, Gibbs, RA, Crenshaw, A, Sivachenko, AY, Topal, MD, Sougnez, C, Dooling, DJ, Fulton, L, Akbani, R, Abbott, R, Dees, ND, Zhang, Q, Kandoth, C, Wendl, M, Schierding, W, Shen, D, Harris, CC, Baggerly, KA, Schmidt, H, Wilson, RK, Kalicki, J, Delehaunty, KD, Fronick, CC, Demeter, R, Cook, L, Wallis, JW, Lin, L, Magrini, VJ, Yung, WK, Hodges, JS, Protopopov, A, Eldred, JM, Smith, SM, Pohl, CS, Vandin, F, Raphael, BJ, Weinstock, GM, Mardis, R, Kim, TM, Hartmann, L, Perna, I, Xiao, Y, Ren, G, Sathiamoorthy, N, Petrelli, N, Lee, E, Kucherlapati, R, Absher, DM, Huang, M, Waite, L, Sherlock, G, Brooks, JD, Li, JZ, Weinstein, JN, Xu, J, Myers, RM, Laird, PW, Cope, L, Herman, JG, Shen, H, Huntsman, DG, Weisenberger, DJ, Noushmehr, H, Pan, F, Massachusetts Institute of Technology. Department of Biology, Lander, Eric S., and Meyerson, Matthew L.
- Subjects
endocrine system diseases ,Serous carcinoma ,Messenger ,Gene Dosage ,Cancer Genome Atlas Research Network ,GYNECOLOGIC-ONCOLOGY-GROUP ,GRADE SEROUS CARCINOMA ,0302 clinical medicine ,Ovarian carcinoma ,Aged ,Carcinoma ,DNA Methylation ,Female ,Gene Expression Profiling ,Gene Expression Regulation, Neoplastic ,Humans ,MicroRNAs ,Middle Aged ,Mutation ,Ovarian Neoplasms ,RNA, Messenger ,Genomics ,Multidisciplinary ,Genetics ,HYBRID SELECTION ,0303 health sciences ,female genital diseases and pregnancy complications ,3. Good health ,Multidisciplinary Sciences ,Serous fluid ,BRCA MUTATION CARRIERS ,030220 oncology & carcinogenesis ,DNA methylation ,PARP inhibitor ,Science & Technology - Other Topics ,General Science & Technology ,Biology ,Article ,03 medical and health sciences ,CLEAR-CELL CARCINOMA ,MD Multidisciplinary ,microRNA ,HIGH-THROUGHPUT ANNOTATION ,medicine ,DRIVER MUTATIONS ,Gene ,030304 developmental biology ,Neoplastic ,Science & Technology ,MUTANT-CELLS ,SOMATIC MUTATIONS ,medicine.disease ,CANCER STATISTICS ,Gene Expression Regulation ,Cancer research ,RNA ,Ovarian cancer - Abstract
A catalogue of molecular aberrations that cause ovarian cancer is critical for developing and deploying therapies that will improve patients’ lives. The Cancer Genome Atlas project has analysed messenger RNA expression, microRNA expression, promoter methylation and DNA copy number in 489 high-grade serous ovarian adenocarcinomas and the DNA sequences of exons from coding genes in 316 of these tumours. Here we report that high-grade serous ovarian cancer is characterized by TP53 mutations in almost all tumours (96%); low prevalence but statistically recurrent somatic mutations in nine further genes including NF1, BRCA1, BRCA2, RB1 and CDK12; 113 significant focal DNA copy number aberrations; and promoter methylation events involving 168 genes. Analyses delineated four ovarian cancer transcriptional subtypes, three microRNA subtypes, four promoter methylation subtypes and a transcriptional signature associated with survival duration, and shed new light on the impact that tumours with BRCA1/2 (BRCA1 or BRCA2) and CCNE1 aberrations have on survival. Pathway analyses suggested that homologous recombination is defective in about half of the tumours analysed, and that NOTCH and FOXM1 signalling are involved in serous ovarian cancer pathophysiology., National Institutes of Health (U.S.) (Grant U54HG003067), National Institutes of Health (U.S.) (Grant U54HG003273), National Institutes of Health (U.S.) (Grant U54HG003079), National Institutes of Health (U.S.) (Grant U24CA126543), National Institutes of Health (U.S.) (Grant U24CA126544), National Institutes of Health (U.S.) (Grant U24CA126546), National Institutes of Health (U.S.) (Grant U24CA126551), National Institutes of Health (U.S.) (Grant U24CA126554), National Institutes of Health (U.S.) (Grant U24CA126561), National Institutes of Health (U.S.) (Grant U24CA126563), National Institutes of Health (U.S.) (Grant U24CA143882), National Institutes of Health (U.S.) (Grant U24CA143731), National Institutes of Health (U.S.) (Grant U24CA143835), National Institutes of Health (U.S.) (Grant U24CA143845), National Institutes of Health (U.S.) (Grant U24CA143858), National Institutes of Health (U.S.) (Grant U24CA144025), National Institutes of Health (U.S.) (Grant U24CA143866), National Institutes of Health (U.S.) (Grant U24CA143867), National Institutes of Health (U.S.) (Grant U24CA143848), National Institutes of Health (U.S.) (Grant U24CA143843), National Institutes of Health (U.S.) (Grant R21CA135877)
- Published
- 2010
17. Expanding the phenotypic spectrum of ARID1B-mediated disorders and identification of altered cell-cycle dynamics due to ARID1B haploinsufficiency
- Author
-
Sim, JCH, White, SM, Fitzpatrick, E, Wilson, GR, Gillies, G, Pope, K, Mountford, HS, Torring, PM, Mckee, S, Vulto-van Silfhout, AT, Jhangiani, SN, Muzny, DM, Leventer, RJ, Delatycki, MB, Amor, DJ, Lockhart, PJ, Sim, JCH, White, SM, Fitzpatrick, E, Wilson, GR, Gillies, G, Pope, K, Mountford, HS, Torring, PM, Mckee, S, Vulto-van Silfhout, AT, Jhangiani, SN, Muzny, DM, Leventer, RJ, Delatycki, MB, Amor, DJ, and Lockhart, PJ
- Abstract
BACKGROUND: Mutations in genes encoding components of the Brahma-associated factor (BAF) chromatin remodeling complex have recently been shown to contribute to multiple syndromes characterised by developmental delay and intellectual disability. ARID1B mutations have been identified as the predominant cause of Coffin-Siris syndrome and have also been shown to be a frequent cause of nonsyndromic intellectual disability. Here, we investigate the molecular basis of a patient with an overlapping but distinctive phenotype of intellectual disability, plantar fat pads and facial dysmorphism. METHODS/RESULTS: High density microarray analysis of the patient demonstrated a heterozygous deletion at 6q25.3, which resulted in the loss of four genes including AT Rich Interactive Domain 1B (ARID1B). Subsequent quantitative real-time PCR analysis revealed ARID1B haploinsufficiency in the patient. Analysis of both patient-derived and ARID1B knockdown fibroblasts after serum starvation demonstrated delayed cell cycle re-entry associated with reduced cell number in the S1 phase. Based on the patient's distinctive phenotype, we ascertained four additional patients and identified heterozygous de novo ARID1B frameshift or nonsense mutations in all of them. CONCLUSIONS: This study broadens the spectrum of ARID1B associated phenotypes by describing a distinctive phenotype including plantar fat pads but lacking the hypertrichosis or fifth nail hypoplasia associated with Coffin-Siris syndrome. We present the first direct evidence in patient-derived cells that alterations in cell cycle contribute to the underlying pathogenesis of syndromes associated with ARID1B haploinsufficiency.
- Published
- 2014
18. Finding the missing honey bee genes: Lessons learned from a genome upgrade
- Author
-
Elsik, CG, Worley, KC, Bennett, AK, Beye, M, Camara, F, Childers, CP, de Graaf, DC, Debyser, G, Deng, J, Devreese, B, Elhaik, E, Evans, JD, Foster, LJ, Graur, D, Guigo, R, Hoff, KJ, Holder, ME, Hudson, ME, Hunt, GJ, Jiang, H, Joshi, V, Khetani, RS, Kosarev, P, Kovar, CL, Ma, J, Maleszka, R, Moritz, RFA, Munoz-Torres, MC, Murphy, TD, Muzny, DM, Newsham, IF, Reese, JT, Robertson, HM, Robinson, GE, Rueppell, O, Solovyev, V, Stanke, M, Stolle, E, Tsuruda, JM, Vaerenbergh, MV, Waterhouse, RM, Weaver, DB, Whitfield, CW, Wu, Y, Zdobnov, EM, Zhang, L, Zhu, D, Gibbs, RA, Patil, S, Gubbala, S, Aqrawi, P, Arias, F, Bess, C, Blankenburg, KB, Brocchini, M, Buhay, C, Challis, D, Chang, K, Chen, D, Coleman, P, Drummond, J, English, A, Evani, U, Francisco, L, Fu, Q, Goodspeed, R, Haessly, TH, Hale, W, Han, H, Hu, Y, Jackson, L, Jakkamsetti, A, Jayaseelan, JC, Kakkar, N, Kalra, D, Kandadi, H, Lee, S, Li, H, Liu, Y, Macmil, S, Mandapat, CM, Mata, R, Mathew, T, Matskevitch, T, Munidasa, M, Nagaswamy, U, Najjar, R, Nguyen, N, Niu, J, Opheim, D, Palculict, T, Paul, S, Pellon, M, Perales, L, Pham, C, Pham, P, Pu, LL, Qi, S, Qu, J, Ren, Y, Elsik, CG, Worley, KC, Bennett, AK, Beye, M, Camara, F, Childers, CP, de Graaf, DC, Debyser, G, Deng, J, Devreese, B, Elhaik, E, Evans, JD, Foster, LJ, Graur, D, Guigo, R, Hoff, KJ, Holder, ME, Hudson, ME, Hunt, GJ, Jiang, H, Joshi, V, Khetani, RS, Kosarev, P, Kovar, CL, Ma, J, Maleszka, R, Moritz, RFA, Munoz-Torres, MC, Murphy, TD, Muzny, DM, Newsham, IF, Reese, JT, Robertson, HM, Robinson, GE, Rueppell, O, Solovyev, V, Stanke, M, Stolle, E, Tsuruda, JM, Vaerenbergh, MV, Waterhouse, RM, Weaver, DB, Whitfield, CW, Wu, Y, Zdobnov, EM, Zhang, L, Zhu, D, Gibbs, RA, Patil, S, Gubbala, S, Aqrawi, P, Arias, F, Bess, C, Blankenburg, KB, Brocchini, M, Buhay, C, Challis, D, Chang, K, Chen, D, Coleman, P, Drummond, J, English, A, Evani, U, Francisco, L, Fu, Q, Goodspeed, R, Haessly, TH, Hale, W, Han, H, Hu, Y, Jackson, L, Jakkamsetti, A, Jayaseelan, JC, Kakkar, N, Kalra, D, Kandadi, H, Lee, S, Li, H, Liu, Y, Macmil, S, Mandapat, CM, Mata, R, Mathew, T, Matskevitch, T, Munidasa, M, Nagaswamy, U, Najjar, R, Nguyen, N, Niu, J, Opheim, D, Palculict, T, Paul, S, Pellon, M, Perales, L, Pham, C, Pham, P, Pu, LL, Qi, S, Qu, J, and Ren, Y
- Abstract
Background: The first generation of genome sequence assemblies and annotations have had a significant impact upon our understanding of the biology of the sequenced species, the phylogenetic relationships among species, the study of populations within and across species, and have informed the biology of humans. As only a few Metazoan genomes are approaching finished quality (human, mouse, fly and worm), there is room for improvement of most genome assemblies. The honey bee (Apis mellifera) genome, published in 2006, was noted for its bimodal GC content distribution that affected the quality of the assembly in some regions and for fewer genes in the initial gene set (OGSv1.0) compared to what would be expected based on other sequenced insect genomes. Results: Here, we report an improved honey bee genome assembly (Amel_4.5) with a new gene annotation set (OGSv3.2), and show that the honey bee genome contains a number of genes similar to that of other insect genomes, contrary to what was suggested in OGSv1.0. The new genome assembly is more contiguous and complete and the new gene set includes ~5000 more protein-coding genes, 50% more than previously reported. About 1/6 of the additional genes were due to improvements to the assembly, and the remaining were inferred based on new RNAseq and protein data. Conclusions: Lessons learned from this genome upgrade have important implications for future genome sequencing projects. Furthermore, the improvements significantly enhance genomic resources for the honey bee, a key model for social behavior and essential to global ecology through pollination. © 2014 Elsik et al.; licensee BioMed Central Ltd.
- Published
- 2014
19. Butterfly genome reveals promiscuous exchange of mimicry adaptations among species
- Author
-
Dasmahapatra, KK, Walters, JR, Briscoe, AD, Davey, JW, Whibley, A, Nadeau, NJ, Zimin, AV, Hughes, DST, Ferguson, LC, Martin, SH, Salazar, C, Lewis, JJ, Adler, S, Ahn, S-J, Baker, DA, Baxter, SW, Chamberlain, NL, Chauhan, R, Counterman, BA, Dalmay, T, Gilbert, LE, Gordon, K, Heckel, DG, Hines, HM, Hoff, KJ, Holland, PWH, Jacquin-Joly, E, Jiggins, FM, Jones, RT, Kapan, DD, Kersey, P, Lamas, G, Lawson, D, Mapleson, D, Maroja, LS, Martin, A, Moxon, S, Palmer, WJ, Papa, R, Papanicolaou, A, Pauchet, Y, Ray, DA, Rosser, N, Salzberg, SL, Supple, MA, Surridge, A, Tenger-Trolander, A, Vogel, H, Wilkinson, PA, Wilson, D, Yorke, JA, Yuan, F, Balmuth, AL, Eland, C, Gharbi, K, Thomson, M, Gibbs, RA, Han, Y, Jayaseelan, JC, Kovar, C, Mathew, T, Muzny, DM, Ongeri, F, Pu, L-L, Qu, J, Thornton, RL, Worley, KC, Wu, Y-Q, Linares, M, Blaxter, ML, Ffrench-Constant, RH, Joron, M, Kronforst, MR, Mullen, SP, Reed, RD, Scherer, SE, Richards, S, Mallet, J, McMillan, WO, Jiggins, CD, Dasmahapatra, KK, Walters, JR, Briscoe, AD, Davey, JW, Whibley, A, Nadeau, NJ, Zimin, AV, Hughes, DST, Ferguson, LC, Martin, SH, Salazar, C, Lewis, JJ, Adler, S, Ahn, S-J, Baker, DA, Baxter, SW, Chamberlain, NL, Chauhan, R, Counterman, BA, Dalmay, T, Gilbert, LE, Gordon, K, Heckel, DG, Hines, HM, Hoff, KJ, Holland, PWH, Jacquin-Joly, E, Jiggins, FM, Jones, RT, Kapan, DD, Kersey, P, Lamas, G, Lawson, D, Mapleson, D, Maroja, LS, Martin, A, Moxon, S, Palmer, WJ, Papa, R, Papanicolaou, A, Pauchet, Y, Ray, DA, Rosser, N, Salzberg, SL, Supple, MA, Surridge, A, Tenger-Trolander, A, Vogel, H, Wilkinson, PA, Wilson, D, Yorke, JA, Yuan, F, Balmuth, AL, Eland, C, Gharbi, K, Thomson, M, Gibbs, RA, Han, Y, Jayaseelan, JC, Kovar, C, Mathew, T, Muzny, DM, Ongeri, F, Pu, L-L, Qu, J, Thornton, RL, Worley, KC, Wu, Y-Q, Linares, M, Blaxter, ML, Ffrench-Constant, RH, Joron, M, Kronforst, MR, Mullen, SP, Reed, RD, Scherer, SE, Richards, S, Mallet, J, McMillan, WO, and Jiggins, CD
- Abstract
The evolutionary importance of hybridization and introgression has long been debated. Hybrids are usually rare and unfit, but even infrequent hybridization can aid adaptation by transferring beneficial traits between species. Here we use genomic tools to investigate introgression in Heliconius, a rapidly radiating genus of neotropical butterflies widely used in studies of ecology, behaviour, mimicry and speciation. We sequenced the genome of Heliconius melpomene and compared it with other taxa to investigate chromosomal evolution in Lepidoptera and gene flow among multiple Heliconius species and races. Among 12,669 predicted genes, biologically important expansions of families of chemosensory and Hox genes are particularly noteworthy. Chromosomal organization has remained broadly conserved since the Cretaceous period, when butterflies split from the Bombyx (silkmoth) lineage. Using genomic resequencing, we show hybrid exchange of genes between three co-mimics, Heliconius melpomene, Heliconius timareta and Heliconius elevatus, especially at two genomic regions that control mimicry pattern. We infer that closely related Heliconius species exchange protective colour-pattern genes promiscuously, implying that hybridization has an important role in adaptive radiation.
- Published
- 2012
20. Genome sequence of an Australian kangaroo, Macropus eugenii, provides insight into the evolution of mammalian reproduction and development
- Author
-
Renfree, MB, Papenfuss, AT, Deakin, JE, Lindsay, J, Heider, T, Belov, K, Rens, W, Waters, PD, Pharo, EA, Shaw, G, Wong, ESW, Lefèvre, CM, Nicholas, KR, Kuroki, Y, Wakefield, MJ, Zenger, KR, Wang, C, Ferguson-Smith, M, Nicholas, FW, Hickford, D, Yu, H, Short, KR, Siddle, HV, Frankenberg, SR, Chew, KY, Menzies, BR, Stringer, JM, Suzuki, S, Hore, TA, Delbridge, ML, Mohammadi, A, Schneider, NY, Hu, Y, O'Hara, W, Al Nadaf, S, Wu, C, Feng, ZP, Cocks, BG, Wang, J, Flicek, P, Searle, SMJ, Fairley, S, Beal, K, Herrero, J, Carone, DM, Suzuki, Y, Sugano, S, Toyoda, A, Sakaki, Y, Kondo, S, Nishida, Y, Tatsumoto, S, Mandiou, I, Hsu, A, McColl, KA, Lansdell, B, Weinstock, G, Kuczek, E, McGrath, A, Wilson, P, Men, A, Hazar-Rethinam, M, Hall, A, Davis, J, Wood, D, Williams, S, Sundaravadanam, Y, Muzny, DM, Jhangiani, SN, Lewis, LR, Morgan, MB, Okwuonu, GO, Ruiz, SJ, Santibanez, J, Nazareth, L, Cree, A, Fowler, G, Kovar, CL, Dinh, HH, Joshi, V, Jing, C, Lara, F, Thornton, R, Chen, L, Deng, J, Liu, Y, Shen, JY, Song, XZ, Edson, J, Troon, C, Thomas, D, Stephens, A, Yapa, L, Levchenko, T, Gibbs, RA, Cooper, DW, Speed, TP, Fujiyama, A, M Graves, JA, O'Neill, RJ, Renfree, MB, Papenfuss, AT, Deakin, JE, Lindsay, J, Heider, T, Belov, K, Rens, W, Waters, PD, Pharo, EA, Shaw, G, Wong, ESW, Lefèvre, CM, Nicholas, KR, Kuroki, Y, Wakefield, MJ, Zenger, KR, Wang, C, Ferguson-Smith, M, Nicholas, FW, Hickford, D, Yu, H, Short, KR, Siddle, HV, Frankenberg, SR, Chew, KY, Menzies, BR, Stringer, JM, Suzuki, S, Hore, TA, Delbridge, ML, Mohammadi, A, Schneider, NY, Hu, Y, O'Hara, W, Al Nadaf, S, Wu, C, Feng, ZP, Cocks, BG, Wang, J, Flicek, P, Searle, SMJ, Fairley, S, Beal, K, Herrero, J, Carone, DM, Suzuki, Y, Sugano, S, Toyoda, A, Sakaki, Y, Kondo, S, Nishida, Y, Tatsumoto, S, Mandiou, I, Hsu, A, McColl, KA, Lansdell, B, Weinstock, G, Kuczek, E, McGrath, A, Wilson, P, Men, A, Hazar-Rethinam, M, Hall, A, Davis, J, Wood, D, Williams, S, Sundaravadanam, Y, Muzny, DM, Jhangiani, SN, Lewis, LR, Morgan, MB, Okwuonu, GO, Ruiz, SJ, Santibanez, J, Nazareth, L, Cree, A, Fowler, G, Kovar, CL, Dinh, HH, Joshi, V, Jing, C, Lara, F, Thornton, R, Chen, L, Deng, J, Liu, Y, Shen, JY, Song, XZ, Edson, J, Troon, C, Thomas, D, Stephens, A, Yapa, L, Levchenko, T, Gibbs, RA, Cooper, DW, Speed, TP, Fujiyama, A, M Graves, JA, and O'Neill, RJ
- Abstract
Background: We present the genome sequence of the tammar wallaby, Macropus eugenii, which is a member of the kangaroo family and the first representative of the iconic hopping mammals that symbolize Australia to be sequenced. The tammar has many unusual biological characteristics, including the longest period of embryonic diapause of any mammal, extremely synchronized seasonal breeding and prolonged and sophisticated lactation within a well-defined pouch. Like other marsupials, it gives birth to highly altricial young, and has a small number of very large chromosomes, making it a valuable model for genomics, reproduction and development.Results: The genome has been sequenced to 2 × coverage using Sanger sequencing, enhanced with additional next generation sequencing and the integration of extensive physical and linkage maps to build the genome assembly. We also sequenced the tammar transcriptome across many tissues and developmental time points. Our analyses of these data shed light on mammalian reproduction, development and genome evolution: there is innovation in reproductive and lactational genes, rapid evolution of germ cell genes, and incomplete, locus-specific X inactivation. We also observe novel retrotransposons and a highly rearranged major histocompatibility complex, with many class I genes located outside the complex. Novel microRNAs in the tammar HOX clusters uncover new potential mammalian HOX regulatory elements.Conclusions: Analyses of these resources enhance our understanding of marsupial gene evolution, identify marsupial-specific conserved non-coding elements and critical genes across a range of biological systems, including reproduction, development and immunity, and provide new insight into marsupial and mammalian biology and genome evolution. © 2011 Renfree et al.; licensee BioMed Central Ltd.
- Published
- 2011
21. Genome-wide survey of SNP variation uncovers the genetic structure of cattle breeds.
- Author
-
Bovine Hap Map, Consortium, Gibbs, Ra, Taylor, Jf, Van Tassel, Cp, Barendse, W, Eversole, Ka, Gill, Ca, Green, Rd, Hamernik, Dl, Kappes, Sm, Lien, S, Matukumalli, Lk, Mcevan, Jc, Mazareth, Lv, Schnabel, Rd, Weinstock, Gm, Wheeler, Da, Ajmone Marsan, Paolo, Boettcher, Pj, Caetano, Ar, Garcia, Jf, Hanotte, O, Mariani, P, Skow, Lc, Sonstegard, T, Williams, Jl, Diallo, B, Hailemariam, L, Martinez, Ml, Morris, Ca, Silva, Lo, Spelman, Rj, Malatu, W, Zhao, K, Abbey, Ca, Agaba, M, Araujo, Fr, Bunch, Rj, Burton, J, Gorni, C, Olivier, H, Harrison, Be, Luff, B, Machado, Ma, Mwakaya, J, Plastow, G, Sim, W, Smith, T, Thomas, Mb, Valentini, A, Williams, P, Womack, J, Wolliams, Ja, Liu, Y, Qin, X, Worley, Kc, Gao, C, Jiang, H, Moore, S, Ren, Y, Song, Xz, Bustamante, Cd, Hernandez, Rd, Muzny, Dm, Patil, S, San Lucas, A, Fu, Q, Kent, Mp, Vega, R, Matukumalli, A, Mcwilliam, S, Sclep, G, Bryc, K, Choi, J, Gao, H, Grefenstette, Jj, Murdoch, B, Stella, A, Villa Angulo, R, Wright, M, Aerts, J, Jann, O, Negrini, Riccardo, Goddard, Me, Hayes, Bj, Bradley, Dg, Lau, Lp, Liu, Ge, Lynn, Dj, Panzitta, F, Dodds, Kg, Ajmone Marsan, Paolo (ORCID:0000-0003-3165-4579), Negrini, Riccardo (ORCID:0000-0002-8735-0286), Bovine Hap Map, Consortium, Gibbs, Ra, Taylor, Jf, Van Tassel, Cp, Barendse, W, Eversole, Ka, Gill, Ca, Green, Rd, Hamernik, Dl, Kappes, Sm, Lien, S, Matukumalli, Lk, Mcevan, Jc, Mazareth, Lv, Schnabel, Rd, Weinstock, Gm, Wheeler, Da, Ajmone Marsan, Paolo, Boettcher, Pj, Caetano, Ar, Garcia, Jf, Hanotte, O, Mariani, P, Skow, Lc, Sonstegard, T, Williams, Jl, Diallo, B, Hailemariam, L, Martinez, Ml, Morris, Ca, Silva, Lo, Spelman, Rj, Malatu, W, Zhao, K, Abbey, Ca, Agaba, M, Araujo, Fr, Bunch, Rj, Burton, J, Gorni, C, Olivier, H, Harrison, Be, Luff, B, Machado, Ma, Mwakaya, J, Plastow, G, Sim, W, Smith, T, Thomas, Mb, Valentini, A, Williams, P, Womack, J, Wolliams, Ja, Liu, Y, Qin, X, Worley, Kc, Gao, C, Jiang, H, Moore, S, Ren, Y, Song, Xz, Bustamante, Cd, Hernandez, Rd, Muzny, Dm, Patil, S, San Lucas, A, Fu, Q, Kent, Mp, Vega, R, Matukumalli, A, Mcwilliam, S, Sclep, G, Bryc, K, Choi, J, Gao, H, Grefenstette, Jj, Murdoch, B, Stella, A, Villa Angulo, R, Wright, M, Aerts, J, Jann, O, Negrini, Riccardo, Goddard, Me, Hayes, Bj, Bradley, Dg, Lau, Lp, Liu, Ge, Lynn, Dj, Panzitta, F, Dodds, Kg, Ajmone Marsan, Paolo (ORCID:0000-0003-3165-4579), and Negrini, Riccardo (ORCID:0000-0002-8735-0286)
- Published
- 2009
22. Whole-exome sequencing points to considerable genetic heterogeneity of cerebral palsy.
- Author
-
McMichael, G., Bainbridge, MN., Haan, E., Corbett, M., Gardner, A., Thompson, S., Bon, BWM van, Eyk, CL van, Broadbent, J., Reynolds, C., O'Callaghan, ME., Nguyen, LS., Adelson, DL., Russo, R., Jhangiani, S., Doddapaneni, H., Muzny, DM., Gibbs, RA., Gecz, J., and MacLennan, AH.
- Subjects
CEREBRAL palsy ,CHROMOSOME abnormalities ,NUCLEIC acid isolation methods ,LYMPHOBLASTOID cell lines - Abstract
Cerebral palsy (CP) is a common, clinically heterogeneous group of disorders affecting movement and posture. Its prevalence has changed little in 50 years and the causes remain largely unknown. The genetic contribution to CP causation has been predicted to be ∼ 2%. We performed whole-exome sequencing of 183 cases with CP including both parents (98 cases) or one parent (67 cases) and 18 singleton cases (no parental DNA). We identified and validated 61 de novo protein-altering variants in 43 out of 98 (44%) case-parent trios. Initial prioritization of variants for causality was by mutation type, whether they were known or predicted to be deleterious and whether they occurred in known disease genes whose clinical spectrum overlaps CP. Further, prioritization used two multidimensional frameworks-the Residual Variation Intolerance Score and the Combined Annotation-dependent Depletion score.Ten de novo mutations in three previously identified disease genes (TUBA!A (n = 2), SCN8A (n = 1) and KDM5C (n = 1)) and in six novel candidate CP genes (AGAP1, JHDM1D, MAST1, NAA35, RFX2 and WIPI2) were predicted to be potentially pathogenic for CP. In addition, we identified four predicted pathogenic, hemizygous variants on chromosome χ in two known disease genes, LI CAM and PAK3, and in two novel candidate CP genes, CD99L2 and TENM1. In total, 14% of CP cases, by strict criteria, had a potentially disease-causing gene variant. Half were in novel genes. The genetic heterogeneity highlights the complexity of the genetic contribution to CP. Function and pathway studies are required to establish the causative role of these putative pathogenic CP genes. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
23. Primary immunodeficiency diseases: Genomic approaches delineate heterogeneous Mendelian disorders
- Author
-
Mohammad K. Eldomery, Olaug K. Rødningen, Cecilia Poli, Debra Canter, Berit Flatø, Ketil Heimdal, Nicholas L. Rider, Silje F. Jørgensen, Hasibe Artac, Hans Christian Erichsen, Francisco Javier Espinosa Rosales, Ivan K. Chinn, Alison A. Bertuch, Bo Yuan, Jordan S. Orange, Emily M. Mace, Wojciech Wiszniewski, Robert Lyle, Shalini N. Jhangiani, Tobias Gedde-Dahl, Carla M. Davis, Carl E. Allen, I. Celine Hanson, Magnus K. O. Burstedt, Thomas B. Issekutz, Mari Ann Kulseth, Yavuz Bayram, Eric A. Smith, Tram N. Cao, Stephen Jolles, Andrew C. Issekutz, Pubudu S. Samarakoon, Alice Y. Chan, Gozde Yesil, Eva Holmberg, Børre Fevang, Diana K. Bayer, John W. Belmont, Asbjørg Stray-Pedersen, Timothy J. Vece, Magdalena Walkiewicz, James R. Lupski, Ying Sheng, Trine Prescott, Liv T. N. Osnes, Cecilie F. Rustad, Nina Denisse Guerrero-Cursaru, Juan Carlos Aldave Becerra, Victor Wei Zhang, Philip M. Boone, Mohammad S. Ehlayel, Jason W. Caldwell, Tore G. Abrahamsen, José Luis Franco, Harshal Abhyankar, Henrik Hjorth-Hansen, Liliana Bezrodnik, Vegard Skogen, Nicola A.M. Wright, Lisa R. Forbes, Anne Grete Bechensteen, Christine R. Beck, Saul Oswaldo Lugo Reyes, Lee-Jun C. Wong, Shen Gu, Sarah K. Nicholas, Christina E. West, Filiz O. Seeborg, Mehmed M. Atik, Eric Boerwinkle, Luis A. Pedroza, Caterina Cancrini, Hanne Sørmo Sorte, Yaping Yang, Christine M. Eng, Richard A. Gibbs, Lenora M. Noroski, Alessandro Aiuti, Ender Karaca, Torstein Øverland, Claudia Milena Trujillo Vargas, Jordan K. Abbott, Geir E. Tjønnfjord, William T. Shearer, Javier Chinen, Ingunn Dybedal, Tomasz Gambin, Donna M. Muzny, Pål Aukrust, Ingvild Nordøy, María Soledad Caldirola, Jianhong Hu, Zeynep Coban Akdemir, YEŞİL, Gözde, Stray Pedersen, A, Sorte, H, Samarakoon, P, Gambin, T, Chinn, Ik, Coban Akdemir, Zh, Erichsen, Hc, Forbes, Lr, Gu, S, Yuan, B, Jhangiani, Sn, Muzny, Dm, Rødningen, Ok, Sheng, Y, Nicholas, Sk, Noroski, Lm, Seeborg, Fo, Davis, Cm, Canter, Dl, Mace, Em, Vece, Tj, Allen, Ce, Abhyankar, Ha, Boone, Pm, Beck, Cr, Wiszniewski, W, Fevang, B, Aukrust, P, Tjønnfjord, Ge, Gedde Dahl, T, Hjorth Hansen, H, Dybedal, I, Nordøy, I, Jørgensen, Sf, Abrahamsen, Tg, Øverland, T, Bechensteen, Ag, Skogen, V, Osnes, Lt, Kulseth, Ma, Prescott, Te, Rustad, Cf, Heimdal, Kr, Belmont, Jw, Rider, Nl, Chinen, J, Cao, Tn, Smith, Ea, Caldirola, M, Bezrodnik, L, Lugo Reyes, So, Espinosa Rosales, Fj, Guerrero Cursaru, Nd, Pedroza, La, Poli, Cm, Franco, Jl, Trujillo Vargas, Cm, Aldave Becerra, Jc, Wright, N, Issekutz, Tb, Issekutz, Ac, Abbott, J, Caldwell, Jw, Bayer, Dk, Chan, Ay, Aiuti, Alessandro, Cancrini, C, Holmberg, E, West, C, Burstedt, M, Karaca, E, Yesil, G, Artac, H, Bayram, Y, Atik, Mm, Eldomery, Mk, Ehlayel, M, Jolles, S, Flatø, B, Bertuch, Aa, Hanson, Ic, Zhang, Vw, Wong, Lj, Hu, J, Walkiewicz, M, Yang, Y, Eng, Cm, Boerwinkle, E, Gibbs, Ra, Shearer, Wt, Lyle, R, Orange, J, Lupski, J. R., and Selçuk Üniversitesi
- Subjects
0301 basic medicine ,Male ,Allergy ,Genomic approaches delineate heterogeneous Mendelian disorders-, JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, cilt.139, ss.232-245, 2017 [Stray-Pedersen A., Sorte H. S. , Samarakoon P., Gambin T., Chinn I. K. , Akdemir Z. H. C. , Erichsen H. C. , Forbes L. R. , Gu S., Yuan B., et al., -Primary immunodeficiency diseases] ,0302 clinical medicine ,OMIM : Online Mendelian Inheritance in Man ,Immunology and Allergy ,2.1 Biological and endogenous factors ,Copy-number variation ,Primary immunodeficiency disease ,whole-exome sequencing ,Aetiology ,Child ,Exome sequencing ,Genetics ,screening and diagnosis ,food and beverages ,High-Throughput Nucleotide Sequencing ,Genomics ,Middle Aged ,Settore MED/38 ,Detection ,030220 oncology & carcinogenesis ,Child, Preschool ,Medical genetics ,Female ,Adult ,medicine.medical_specialty ,Adolescent ,DNA Copy Number Variations ,Immunology ,Biology ,03 medical and health sciences ,Young Adult ,Rare Diseases ,Clinical Research ,medicine ,Humans ,Genetic Testing ,Preschool ,Aged ,Severe combined immunodeficiency ,Genetic heterogeneity ,Common variable immunodeficiency ,Prevention ,fungi ,Human Genome ,Immunologic Deficiency Syndromes ,Infant ,medicine.disease ,4.1 Discovery and preclinical testing of markers and technologies ,030104 developmental biology ,Good Health and Well Being ,Primary immunodeficiency ,copy number variants - Abstract
WOS: 000393996800025, PubMed: 27577878, Background: Primary immunodeficiency diseases (PIDDs) are clinically and genetically heterogeneous disorders thus far associated with mutations in more than 300 genes. The clinical phenotypes derived from distinct genotypes can overlap. Genetic etiology can be a prognostic indicator of disease severity and can influence treatment decisions. Objective: We sought to investigate the ability of whole-exome screening methods to detect disease-causing variants in patients with PIDDs. Methods: Patients with PIDDs from 278 families from 22 countries were investigated by using whole-exome sequencing. Computational copy number variant (CNV) prediction pipelines and an exome-tiling chromosomal microarray were also applied to identify intragenic CNVs. Analytic approaches initially focused on 475 known or candidate PIDD genes but were nonexclusive and further tailored based on clinical data, family history, and immunophenotyping. Results: A likely molecular diagnosis was achieved in 110 (40%) unrelated probands. Clinical diagnosis was revised in about half (60/ 110) and management was directly altered in nearly a quarter (26/ 110) of families based on molecular findings. Twelve PIDD-causing CNVs were detected, including 7 smaller than 30 Kb that would not have been detected with conventional diagnostic CNV arrays. Conclusion: This high-throughput genomic approach enabled detection of disease-related variants in unexpected genes; permitted detection of low-grade constitutional, somatic, and revertant mosaicism; and provided evidence of a mutational burden in mixed PIDD immunophenotypes., South-Eastern Norway Health Authority; American Women's club of Oslo; National Human Genome Research InstituteUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Human Genome Research Institute (NHGRI); National Heart, Lung, and BloodUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Heart Lung & Blood Institute (NHLBI) [U54HG006542]; Jeffrey Modell Foundation; NIHUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USA [AI-120989], Funding for the work performed in Oslo was provided by the South-Eastern Norway Health Authority, and A. S.-P. received research scholarship from the American Women's club of Oslo. The BHCMG is supported by the National Human Genome Research Institute and the National Heart, Lung, and Blood (U54HG006542). Funding was also provided by the Jeffrey Modell Foundation and NIH AI-120989 (to J.S.O.).
- Published
- 2017
24. Somatic mutations affect key pathways in lung adenocarcinoma
- Author
-
Aldi T. Kraja, Brian H. Dunford-Shore, Tittu Mathew, Otis Hall, Barbara A. Weir, Timothy Fennell, William Pao, Jack A. Roth, Alicia Hawes, Heidi Greulich, Steven E. Scherer, Xiaoqi Shi, Giovanni Tonon, Manuel L. Gonzalez-Garay, Yuzhu Tang, Mark B. Orringer, Qunyuan Zhang, Bruce E. Johnson, Li Ding, David G. Beer, Amit Dutt, Margaret R. Spitz, Carrie A. Haipek, Michael A. Province, Yiming Zhu, Liuda Ziaugra, Lucian R. Chirieac, Ken Chen, Rachel Abbott, William D. Travis, George M. Weinstock, Harold E. Varmus, Lucinda Fulton, Daniel C. Koboldt, Kristian Cibulskis, Carrie Sougnez, Christopher S. Sawyer, Richard A. Gibbs, Bradley A. Ozenberger, Thomas J. Giordano, Heather Schmidt, Ling Lin, Jennifer Baldwin, Elaine R. Mardis, Rick Meyer, Tracie L. Miner, David E. Larson, Ignacio I. Wistuba, Jiqiang Yao, Margaret Morgan, Andrew C. Chang, Akihiko Yoshizawa, Shalini N. Jhangiani, Xiaojun Zhao, David A. Wheeler, Stephen R. Broderick, Jody S. Robinson, Kerstin Clerc, Eric S. Lander, Richard K. Wilson, Ginger A. Fewell, Hua Shen, David J. Dooling, Robert S. Fulton, Aleksandar Milosavljevic, John R. Osborne, Gad Getz, Donna M. Muzny, Yanru Ren, Wendy Winckler, Roman K. Thomas, Mark A. Watson, Peter J. Good, Sacha N. Sander, Megan Hanna, Michael D. McLellan, Ginger A. Metcalf, Brian Ng, Michael C. Wendl, Lora Lewis, Seth D. Crosby, Michael C. Zody, Matthew Meyerson, Robert C. Onofrio, Michael S. Lawrence, Marc Ladanyi, Aniko Sabo, Craig Pohl, Stacey Gabriel, Tammi L. Vickery, Ding, L, Getz, G, Wheeler, Da, Mardis, Ea, Mclellan, Md, Cibulskis, K, Sougnez, C, Greulich, H, Muzny, Dm, Morgan, Mb, Fulton, L, Fulton, R, Zhang, Q, Wendl, Mc, Lawrence, M, Larson, De, Chen, K, Dooling, Dj, Sabo, A, Hawes, Ac, Shen, H, Jhangiani, Sh, Lewis, Lr, Hall, O, Zhu, Y, Mathew, T, Ren, Y, Yao, J, Scherer, Se, Clerc, K, Metcalf, Ga, Ng, B, Milosavljevic, A, Gonzalez-Garay, Ml, Osborne, Jr, Meyer, R, Shi, X, Tang, Y, Koboldt, Dc, Lin, L, Abbott, R, Miner, Tl, Pohl, C, Fewell, G, Haipek, C, Schmidt, H, Dunford-Shore, Bh, Kraja, A, Crosby, Sd, Sawyer, C, Vickery, T, Sander, S, Robinson, J, Winckler, W, Baldwin, J, Chirieac, Lr, Dutt, A, Fennell, T, Hanna, M, Johnson, Be, Onofrio, Rc, Thomas, Rk, Tonon, G, Weir, Ba, Zhao, X, Ziaugra, L, Zody, Mc, Giordano, T, Orringer, Mb, Roth, Ja, Spitz, Mr, Wistuba, Ii, Ozenberger, B, Good, Pj, Chang, Ac, Beer, Dg, Watson, Ma, Ladanyi, M, Broderick, S, Yoshizawa, A, Travis, Wd, Pao, W, Province, Ma, Weinstock, Gm, Varmus, He, Gabriel, Sb, Lander, E, Gibbs, Ra, Meyerson, M, and Wilson, Rk.
- Subjects
Male ,Genetics ,Mutation ,Lung Neoplasms ,Multidisciplinary ,Tumor suppressor gene ,DNA repair ,Gene Dosage ,Adenocarcinoma, Bronchiolo-Alveolar ,Biology ,medicine.disease ,medicine.disease_cause ,Article ,Gene Expression Regulation, Neoplastic ,Germline mutation ,Proto-Oncogenes ,medicine ,Humans ,Adenocarcinoma ,Female ,Genes, Tumor Suppressor ,Carcinogenesis ,Lung cancer ,Gene - Abstract
Determining the genetic basis of cancer requires comprehensive analyses of large collections of histopathologically well-classified primary tumours. Here we report the results of a collaborative study to discover somatic mutations in 188 human lung adenocarcinomas. DNA sequencing of 623 genes with known or potential relationships to cancer revealed more than 1,000 somatic mutations across the samples. Our analysis identified 26 genes that are mutated at significantly high frequencies and thus are probably involved in carcinogenesis. The frequently mutated genes include tyrosine kinases, among them the EGFR homologue ERBB4; multiple ephrin receptor genes, notably EPHA3; vascular endothelial growth factor receptor KDR; and NTRK genes. These data provide evidence of somatic mutations in primary lung adenocarcinoma for several tumour suppressor genes involved in other cancers--including NF1, APC, RB1 and ATM--and for sequence changes in PTPRD as well as the frequently deleted gene LRP1B. The observed mutational profiles correlate with clinical features, smoking status and DNA repair defects. These results are reinforced by data integration including single nucleotide polymorphism array and gene expression array. Our findings shed further light on several important signalling pathways involved in lung adenocarcinoma, and suggest new molecular targets for treatment.
- Published
- 2008
25. VizCNV: An integrated platform for concurrent phased BAF and CNV analysis with trio genome sequencing data.
- Author
-
Du H, Lun MY, Gagarina L, Mehaffey MG, Hwang JP, Jhangiani SN, Bhamidipati SV, Muzny DM, Poli MC, Ochoa S, Chinn IK, Linstrand A, Posey JE, Gibbs RA, Lupski JR, and Carvalho CMB
- Abstract
Background: Copy number variation (CNV) is a class of genomic Structural Variation (SV) that underlie genomic disorders and can have profound implications for health. Short-read genome sequencing (sr-GS) enables CNV calling for genomic intervals of variable size and across multiple phenotypes. However, unresolved challenges include an overwhelming number of false-positive calls due to systematic biases from non-uniform read coverage and collapsed calls resulting from the abundance of paralogous segments and repetitive elements in the human genome., Methods: To address these interpretative challenges, we developed VizCNV. The VizCNV computational tool for inspecting CNV calls uses various data signal sources from sr-GS data, including read depth, phased B-allele frequency, as well as benchmarking signals from other SV calling methods. The interactive features and view modes are adept for analyzing both chromosomal abnormalities [e.g., aneuploidy, segmental aneusomy, and chromosome translocations], gene exonic CNV and non-coding gene regulatory regions. In addition, VizCNV includes a built-in filter schema for trio genomes, prioritizing the detection of impactful germline CNVs, such as de novo CNVs. Upon computational optimization by fine-tuning parameters to maximize sensitivity and specificity, VizCNV demonstrated approximately 83.8% recall and 77.2% precision on the 1000 Genome Project data with an average coverage read depth of 30x., Results: We applied VizCNV to 39 families with primary immunodeficiency disease without a molecular diagnosis. With implemented build-in filter, we identified two de novo CNVs and 90 inherited CNVs >10 kb per trio. Genotype-phenotype analyses revealed that a compound heterozygous combination of a paternal 12.8 kb deletion of exon 5 and a maternal missense variant allele of DOCK8 are likely the molecular cause of one proband., Conclusions: VizCNV provides a robust platform for genome-wide relevant CNV discovery and visualization of such CNV using sr-GS data.
- Published
- 2024
- Full Text
- View/download PDF
26. Identification of allele-specific KIV-2 repeats and impact on Lp(a) measurements for cardiovascular disease risk.
- Author
-
Behera S, Belyeu JR, Chen X, Paulin LF, Nguyen NQH, Newman E, Mahmoud M, Menon VK, Qi Q, Joshi P, Marcovina S, Rossi M, Roller E, Han J, Onuchic V, Avery CL, Ballantyne CM, Rodriguez CJ, Kaplan RC, Muzny DM, Metcalf GA, Gibbs RA, Yu B, Boerwinkle E, Eberle MA, and Sedlazeck FJ
- Subjects
- Humans, DNA Copy Number Variations, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide, Cardiovascular Diseases genetics, Alleles, Lipoprotein(a) genetics, Lipoprotein(a) blood
- Abstract
The abundance of Lp(a) protein holds significant implications for the risk of cardiovascular disease (CVD), which is directly impacted by the copy number (CN) of KIV-2, a 5.5 kbp sub-region. KIV-2 is highly polymorphic in the population and accurate analysis is challenging. In this study, we present the DRAGEN KIV-2 CN caller, which utilizes short reads. Data across 166 WGS show that the caller has high accuracy, compared to optical mapping and can further phase approximately 50% of the samples. We compared KIV-2 CN numbers to 24 previously postulated KIV-2 relevant SNVs, revealing that many are ineffective predictors of KIV-2 copy number. Population studies, including USA-based cohorts, showed distinct KIV-2 CN, distributions for European-, African-, and Hispanic-American populations and further underscored the limitations of SNV predictors. We demonstrate that the CN estimates correlate significantly with the available Lp(a) protein levels and that phasing is highly important., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
27. Development and extensive sequencing of a broadly-consented Genome in a Bottle matched tumor-normal pair.
- Author
-
McDaniel JH, Patel V, Olson ND, He HJ, He Z, Cole KD, Schmitt A, Sikkink K, Sedlazeck FJ, Doddapaneni H, Jhangiani SN, Muzny DM, Gingras MC, Mehta H, Paulin LF, Hastie AR, Yu HC, Weigman V, Rojas A, Kennedy K, Remington J, Gonzalez I, Sudkamp M, Wiseman K, Lajoie BR, Levy S, Jain M, Akeson S, Narzisi G, Steinsnyder Z, Reeves C, Shelton J, Kingan SB, Lambert C, Bayabyan P, Wenger AM, McLaughlin IJ, Adamson A, Kingsley C, Wescott M, Kim Y, Paten B, Park J, Violich I, Miga KH, Gardner J, McNulty B, Rosen G, McCoy R, Brundu F, Sayyari E, Scheffler K, Truong S, Catreux S, Hannah LC, Lipson D, Benjamin H, Iremadze N, Soifer I, Eacker S, Wood M, Cross E, Husar G, Gross S, Vernich M, Kolmogorov M, Ahmad T, Keskus A, Bryant A, Thibaud-Nissen F, Trow J, Proszynski J, Hirschberg JW, Ryon K, Mason CE, Wagner J, Xiao C, Liss AS, and Zook JM
- Abstract
The Genome in a Bottle Consortium (GIAB), hosted by the National Institute of Standards and Technology (NIST), is developing new matched tumor-normal samples, the first to be explicitly consented for public dissemination of genomic data and cell lines. Here, we describe a comprehensive genomic dataset from the first individual, HG008, including DNA from an adherent, epithelial-like pancreatic ductal adenocarcinoma (PDAC) tumor cell line and matched normal cells from duodenal and pancreatic tissues. Data for the tumor-normal matched samples comes from thirteen distinct state-of-the-art whole genome measurement technologies, including high depth short and long-read bulk whole genome sequencing (WGS), single cell WGS, and Hi-C, and karyotyping. These data will be used by the GIAB Consortium to develop matched tumor-normal benchmarks for somatic variant detection. We expect these data to facilitate innovation for whole genome measurement technologies, de novo assembly of tumor and normal genomes, and bioinformatic tools to identify small and structural somatic mutations. This first-of-its-kind broadly consented open-access resource will facilitate further understanding of sequencing methods used for cancer biology., Competing Interests: Competing interests A.S. and K.S. are employees of Arima Genomics. L.F.P. from BCM, was sponsored by Genentech Inc until September 2023. F.J.S from BCM, received research support from Illumina, ONT and Pacbio. A.R.H and H-C.Y. are employees of Bionano Genomics and own stock shares and options of Bionano Genomics, Inc. V.W., K.K., J.R., and I.G. are employees of BioSkryb Genomics. M.S., K.B., B.R.L. and S.L. are employees of Element Biosciences. S.B.K., C.L., P.B., A.M.W., I.J.M., A.A., C.K., M.W., and Y.K. are employees and shareholders of PacBio, Inc. D.L., H.B., N.I., and I.S. are employees and shareholders of Ultima Genomics. S.E. and M.W. are employees of Phase Genomics. E.C., G.H., S.G., and M.V. are employees of KromaTiD, Inc, E.C. is also a shareholder. F.B., E.S., K.S., S.T. and S.C. are employees of Illumina, Inc. All other authors have no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
28. Complete Genomic Characterization of Global Pathogens, Respiratory Syncytial Virus (RSV), and Human Norovirus (HuNoV) Using Probe-based Capture Enrichment.
- Author
-
Bhamidipati SV, Surathu A, Chao H, Agustinho DP, Xiang Q, Kottapalli K, Santhanam A, Momin Z, Walker K, Menon VK, Weissenberger G, Emerick N, Mahjabeen F, Meng Q, Hu J, Sucgang R, Henke D, Sedlazeck FJ, Khan Z, Metcalf GA, Avadhanula V, Piedra PA, Ramani S, Atmar RL, Estes MK, Petrosino JF, Gibbs RA, Muzny DM, Cregeen SJ, and Doddapaneni H
- Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide, while human noroviruses (HuNoV) are a leading cause of epidemic and sporadic acute gastroenteritis. Generating full-length genome sequences for these viruses is crucial for understanding viral diversity and tracking emerging variants. However, obtaining high-quality sequencing data is often challenging due to viral strain variability, quality, and low titers. Here, we present a set of comprehensive oligonucleotide probe sets designed from 1,570 RSV and 1,376 HuNoV isolate sequences in GenBank. Using these probe sets and a capture enrichment sequencing workflow, 85 RSV positive nasal swab samples and 55 (49 stool and six human intestinal enteroids) HuNoV positive samples encompassing major subtypes and genotypes were characterized. The Ct values of these samples ranged from 17.0-29.9 for RSV, and from 20.2-34.8 for HuNoV, with some HuNoV having below the detection limit. The mean percentage of post-processing reads mapped to viral genomes was 85.1% for RSV and 40.8% for HuNoV post-capture, compared to 0.08% and 1.15% in pre-capture libraries, respectively. Full-length genomes were>99% complete in all RSV positive samples and >96% complete in 47/55 HuNoV positive samples-a significant improvement over genome recovery from pre-capture libraries. RSV transcriptome (subgenomic mRNAs) sequences were also characterized from this data. Probe-based capture enrichment offers a comprehensive approach for RSV and HuNoV genome sequencing and monitoring emerging variants.
- Published
- 2024
- Full Text
- View/download PDF
29. INTRA- AND INTER-HOST EVOLUTION OF HUMAN NOROVIRUS IN HEALTHY ADULTS.
- Author
-
Ramani S, Javornik Cregeen SJ, Surathu A, Neill FH, Muzny DM, Doddapaneni H, Menon VK, Hoffman KL, Ross MC, Metcalf G, Opekun AR, Graham DY, Gibbs RA, Petrosino JF, Estes MK, and Atmar RL
- Abstract
Background: Human noroviruses are a leading cause of acute and sporadic gastroenteritis worldwide. The evolution of human noroviruses in immunocompromised persons has been evaluated in many studies. Much less is known about the evolutionary dynamics of human norovirus in healthy adults., Methods: We used sequential samples collected from a controlled human infection study with GI.1/Norwalk/US/68 virus to evaluate intra- and inter-host evolution of a human norovirus in healthy adults. Up to 12 samples from day 1 to day 56 post-challenge were sequenced using a norovirus-specific capture probe method., Results: Complete genomes were assembled, even in samples that were below the limit of detection of standard RT-qPCR assays, up to 28 days post-challenge. Analysis of 123 complete genomes showed changes in the GI.1 genome in all persons, but there were no conserved changes across all persons. Single nucleotide variants resulting in non-synonymous amino acid changes were observed in all proteins, with the capsid VP1 and nonstructural protein NS3 having the largest numbers of changes., Conclusions: These data highlight the potential of a new capture-based sequencing approach to assemble human norovirus genomes with high sensitivity and demonstrate limited conserved immune pressure-driven evolution of GI.1 virus in healthy adults.
- Published
- 2024
- Full Text
- View/download PDF
30. Examining intra-host genetic variation of RSV by short read high-throughput sequencing.
- Author
-
Henke D, Piedra FA, Avadhanula V, Doddapaneni H, Muzny DM, Menon VK, Hoffman KL, Ross MC, Javornik Cregeen SJ, Metcalf G, Gibbs RA, Petrosino JF, and Piedra PA
- Abstract
Every viral infection entails an evolving population of viral genomes. High-throughput sequencing technologies can be used to characterize such populations, but to date there are few published examples of such work. In addition, mixed sequencing data are sometimes used to infer properties of infecting genomes without discriminating between genome-derived reads and reads from the much more abundant, in the case of a typical active viral infection, transcripts. Here we apply capture probe-based short read high-throughput sequencing to nasal wash samples taken from a previously described group of adult hematopoietic cell transplant (HCT) recipients naturally infected with respiratory syncytial virus (RSV). We separately analyzed reads from genomes and transcripts for the levels and distribution of genetic variation by calculating per position Shannon entropies. Our analysis reveals a low level of genetic variation within the RSV infections analyzed here, but with interesting differences between genomes and transcripts in 1) average per sample Shannon entropies; 2) the genomic distribution of variation 'hotspots'; and 3) the genomic distribution of hotspots encoding alternative amino acids. In all, our results suggest the importance of separately analyzing reads from genomes and transcripts when interpreting high-throughput sequencing data for insight into intra-host viral genome replication, expression, and evolution.
- Published
- 2024
- Full Text
- View/download PDF
31. Inverted triplications formed by iterative template switches generate structural variant diversity at genomic disorder loci.
- Author
-
Grochowski CM, Bengtsson JD, Du H, Gandhi M, Lun MY, Mehaffey MG, Park K, Höps W, Benito E, Hasenfeld P, Korbel JO, Mahmoud M, Paulin LF, Jhangiani SN, Hwang JP, Bhamidipati SV, Muzny DM, Fatih JM, Gibbs RA, Pendleton M, Harrington E, Juul S, Lindstrand A, Sedlazeck FJ, Pehlivan D, Lupski JR, and Carvalho CMB
- Subjects
- Humans, Comparative Genomic Hybridization, Genomic Structural Variation genetics, Genome, Human genetics, Gene Duplication genetics, Haplotypes genetics
- Abstract
The duplication-triplication/inverted-duplication (DUP-TRP/INV-DUP) structure is a complex genomic rearrangement (CGR). Although it has been identified as an important pathogenic DNA mutation signature in genomic disorders and cancer genomes, its architecture remains unresolved. Here, we studied the genomic architecture of DUP-TRP/INV-DUP by investigating the DNA of 24 patients identified by array comparative genomic hybridization (aCGH) on whom we found evidence for the existence of 4 out of 4 predicted structural variant (SV) haplotypes. Using a combination of short-read genome sequencing (GS), long-read GS, optical genome mapping, and single-cell DNA template strand sequencing (strand-seq), the haplotype structure was resolved in 18 samples. The point of template switching in 4 samples was shown to be a segment of ∼2.2-5.5 kb of 100% nucleotide similarity within inverted repeat pairs. These data provide experimental evidence that inverted low-copy repeats act as recombinant substrates. This type of CGR can result in multiple conformers generating diverse SV haplotypes in susceptible dosage-sensitive loci., Competing Interests: Declaration of interests Baylor College of Medicine and Miraca Holdings have formed a joint venture with shared ownership and governance of BG, which performs clinical microarray analysis, clinical ES, and clinical biochemical studies. J.R.L. serves on the scientific advisory board of the BG. J.R.L. has stock ownership in 23andMe, is a paid consultant for Genomics International, and is a co-inventor on multiple US and European patents related to molecular diagnostics for inherited neuropathies, eye diseases, genomic disorders, and bacterial genomic fingerprinting. E.H. and S.J. are employees of ONT and shareholders and/or share option holders of ONT. D.P. provides consulting services for Ionis Pharmaceuticals. F.J.S. receives research support from Genetech, Illumina, Pacbio, and ONT., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
32. Somatic mutations of esophageal adenocarcinoma: a comparison between Black and White patients.
- Author
-
Lim H, Gingras MC, Zhao J, Byun J, Castro PD, Tsavachidis S, Hu J, Doddapaneni H, Han Y, Muzny DM, Gibbs RA, Amos CI, and Thrift AP
- Subjects
- Female, Humans, Male, Mutation, Black or African American, White, Exome Sequencing, Adenocarcinoma genetics, Adenocarcinoma pathology, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology
- Abstract
Esophageal adenocarcinoma is the most common histological subtype of esophageal cancer in Western countries and shows poor prognosis with rapid growth. EAC is characterized by a strong male predominance and racial disparity. EAC is up to fivefold more common among Whites than Blacks, yet Black patients with EAC have poorer survival rates. The racial disparity remains largely unknown, and there is limited knowledge of mutations in EAC regarding racial disparities. We used whole-exome sequencing to show somatic mutation profiles derived from tumor samples from 18 EAC male patients. We identified three molecular subgroups based on the pre-defined esophageal cancer-specific mutational signatures. Group 1 is associated with age and NTHL1 deficiency-related signatures. Group 2 occurs primarily in Black patients and is associated with signatures related to DNA damage from oxidative stress and NTHL1 deficiency-related signatures. Group 3 is associated with defective homologous recombination-based DNA often caused by BRCA mutation in White patients. We observed significantly mutated race related genes (LCE2B in Black, SDR39U1 in White) were (q-value < 0.1). Our findings underscore the possibility of distinct molecular mutation patterns in EAC among different races. Further studies are needed to validate our findings, which could contribute to precision medicine in EAC., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
33. NODAL variants are associated with a continuum of laterality defects from simple D-transposition of the great arteries to heterotaxy.
- Author
-
Dardas Z, Fatih JM, Jolly A, Dawood M, Du H, Grochowski CM, Jones EG, Jhangiani SN, Wehrens XHT, Liu P, Bi W, Boerwinkle E, Posey JE, Muzny DM, Gibbs RA, Lupski JR, Coban-Akdemir Z, and Morris SA
- Subjects
- Animals, Humans, Arteries, Comparative Genomic Hybridization, Phenotype, Heart Defects, Congenital genetics, Heterotaxy Syndrome genetics, Transposition of Great Vessels
- Abstract
Background: NODAL signaling plays a critical role in embryonic patterning and heart development in vertebrates. Genetic variants resulting in perturbations of the TGF-β/NODAL signaling pathway have reproducibly been shown to cause laterality defects in humans. To further explore this association and improve genetic diagnosis, the study aims to identify and characterize a broader range of NODAL variants in a large number of individuals with laterality defects., Methods: We re-analyzed a cohort of 321 proband-only exomes of individuals with clinically diagnosed laterality congenital heart disease (CHD) using family-based, rare variant genomic analyses. To this cohort we added 12 affected subjects with known NODAL variants and CHD from institutional research and clinical cohorts to investigate an allelic series. For those with candidate contributory variants, variant allele confirmation and segregation analysis were studied by Sanger sequencing in available family members. Array comparative genomic hybridization and droplet digital PCR were utilized for copy number variants (CNV) validation and characterization. We performed Human Phenotype Ontology (HPO)-based quantitative phenotypic analyses to dissect allele-specific phenotypic differences., Results: Missense, nonsense, splice site, indels, and/or structural variants of NODAL were identified as potential causes of heterotaxy and other laterality defects in 33 CHD cases. We describe a recurrent complex indel variant for which the nucleic acid secondary structure predictions implicate secondary structure mutagenesis as a possible mechanism for formation. We identified two CNV deletion alleles spanning NODAL in two unrelated CHD cases. Furthermore, 17 CHD individuals were found (16/17 with known Hispanic ancestry) to have the c.778G > A:p.G260R NODAL missense variant which we propose reclassification from variant of uncertain significance (VUS) to likely pathogenic. Quantitative HPO-based analyses of the observed clinical phenotype for all cases with p.G260R variation, including heterozygous, homozygous, and compound heterozygous cases, reveal clustering of individuals with biallelic variation. This finding provides evidence for a genotypic-phenotypic correlation and an allele-specific gene dosage model., Conclusion: Our data further support a role for rare deleterious variants in NODAL as a cause for sporadic human laterality defects, expand the repertoire of observed anatomical complexity of potential cardiovascular anomalies, and implicate an allele specific gene dosage model., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
34. Genetic sex validation for sample tracking in next-generation sequencing clinical testing.
- Author
-
Hu J, Korchina V, Zouk H, Harden MV, Murdock D, Macbeth A, Harrison SM, Lennon N, Kovar C, Balasubramanian A, Zhang L, Chandanavelli G, Pasham D, Rowley R, Wiley K, Smith ME, Gordon A, Jarvik GP, Sleiman P, Kelly MA, Bland HT, Murugan M, Venner E, Boerwinkle E, Prows C, Mahanta L, Rehm HL, Gibbs RA, and Muzny DM
- Subjects
- Humans, Bone Marrow Transplantation, Genotype, Laboratories, High-Throughput Nucleotide Sequencing, Clinical Laboratory Services
- Abstract
Objective: Data from DNA genotyping via a 96-SNP panel in a study of 25,015 clinical samples were utilized for quality control and tracking of sample identity in a clinical sequencing network. The study aimed to demonstrate the value of both the precise SNP tracking and the utility of the panel for predicting the sex-by-genotype of the participants, to identify possible sample mix-ups., Results: Precise SNP tracking showed no sample swap errors within the clinical testing laboratories. In contrast, when comparing predicted sex-by-genotype to the provided sex on the test requisition, we identified 110 inconsistencies from 25,015 clinical samples (0.44%), that had occurred during sample collection or accessioning. The genetic sex predictions were confirmed using additional SNP sites in the sequencing data or high-density genotyping arrays. It was determined that discrepancies resulted from clerical errors (49.09%), samples from transgender participants (3.64%) and stem cell or bone marrow transplant patients (7.27%) along with undetermined sample mix-ups (40%) for which sample swaps occurred prior to arrival at genome centers, however the exact cause of the events at the sampling sites resulting in the mix-ups were not able to be determined., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
35. Inter and intra-host diversity of RSV in hematopoietic stem cell transplant adults with normal and delayed viral clearance.
- Author
-
Avadhanula V, Agustinho DP, Menon VK, Chemaly RF, Shah DP, Qin X, Surathu A, Doddapaneni H, Muzny DM, Metcalf GA, Cregeen SJ, Gibbs RA, Petrosino JF, Sedlazeck FJ, and Piedra PA
- Abstract
Respiratory syncytial virus (RSV) infection in immunocompromised individuals often leads to prolonged illness, progression to severe lower respiratory tract infection, and even death. How the host immune environment of the hematopoietic stem cell transplant (HCT) adults can affect viral genetic variation during an acute infection is not understood well. In the present study, we performed whole genome sequencing of RSV/A or RSV/B from samples collected longitudinally from HCT adults with normal (<14 days) and delayed (≥14 days) RSV clearance who were enrolled in a ribavirin trial. We determined the inter-host and intra-host genetic variation of RSV and the effect of mutations on putative glycosylation sites. The inter-host variation of RSV is centered in the attachment (G) and fusion (F) glycoprotein genes followed by polymerase (L) and matrix (M) genes. Interestingly, the overall genetic variation was constant between normal and delayed clearance groups for both RSV/A and RSV/B. Intra-host variation primarily occurred in the G gene followed by non-structural protein (NS1) and L genes; however, gain or loss of stop codons and frameshift mutations appeared only in the G gene and only in the delayed viral clearance group. Potential gain or loss of O-linked glycosylation sites in the G gene occurred both in RSV/A and RSV/B isolates. For RSV F gene, loss of N-linked glycosylation site occurred in three RSV/B isolates within an antigenic epitope. Both oral and aerosolized ribavirin did not cause any mutations in the L gene. In summary, prolonged viral shedding and immune deficiency resulted in RSV variation, especially in structural mutations in the G gene, possibly associated with immune evasion. Therefore, sequencing and monitoring of RSV isolates from immunocompromised patients are crucial as they can create escape mutants that can impact the effectiveness of upcoming vaccines and treatments., Competing Interests: F.J.S. has research funding from Illumina, Pacbio, Genentech and Oxford Nanopore. The remaining authors declare no competing interests, (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
36. Dominant negative variants in KIF5B cause osteogenesis imperfecta via down regulation of mTOR signaling.
- Author
-
Marom R, Zhang B, Washington ME, Song IW, Burrage LC, Rossi VC, Berrier AS, Lindsey A, Lesinski J, Nonet ML, Chen J, Baldridge D, Silverman GA, Sutton VR, Rosenfeld JA, Tran AA, Hicks MJ, Murdock DR, Dai H, Weis M, Jhangiani SN, Muzny DM, Gibbs RA, Caswell R, Pottinger C, Cilliers D, Stals K, Eyre D, Krakow D, Schedl T, Pak SC, and Lee BH
- Subjects
- Animals, Humans, Mice, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Carrier Proteins genetics, Down-Regulation, NIH 3T3 Cells, Proteomics, Signal Transduction genetics, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Kinesins genetics, Kinesins metabolism, Osteogenesis Imperfecta
- Abstract
Background: Kinesin motor proteins transport intracellular cargo, including mRNA, proteins, and organelles. Pathogenic variants in kinesin-related genes have been implicated in neurodevelopmental disorders and skeletal dysplasias. We identified de novo, heterozygous variants in KIF5B, encoding a kinesin-1 subunit, in four individuals with osteogenesis imperfecta. The variants cluster within the highly conserved kinesin motor domain and are predicted to interfere with nucleotide binding, although the mechanistic consequences on cell signaling and function are unknown., Methods: To understand the in vivo genetic mechanism of KIF5B variants, we modeled the p.Thr87Ile variant that was found in two patients in the C. elegans ortholog, unc-116, at the corresponding position (Thr90Ile) by CRISPR/Cas9 editing and performed functional analysis. Next, we studied the cellular and molecular consequences of the recurrent p.Thr87Ile variant by microscopy, RNA and protein analysis in NIH3T3 cells, primary human fibroblasts and bone biopsy., Results: C. elegans heterozygous for the unc-116 Thr90Ile variant displayed abnormal body length and motility phenotypes that were suppressed by additional copies of the wild type allele, consistent with a dominant negative mechanism. Time-lapse imaging of GFP-tagged mitochondria showed defective mitochondria transport in unc-116 Thr90Ile neurons providing strong evidence for disrupted kinesin motor function. Microscopy studies in human cells showed dilated endoplasmic reticulum, multiple intracellular vacuoles, and abnormal distribution of the Golgi complex, supporting an intracellular trafficking defect. RNA sequencing, proteomic analysis, and bone immunohistochemistry demonstrated down regulation of the mTOR signaling pathway that was partially rescued with leucine supplementation in patient cells., Conclusion: We report dominant negative variants in the KIF5B kinesin motor domain in individuals with osteogenesis imperfecta. This study expands the spectrum of kinesin-related disorders and identifies dysregulated signaling targets for KIF5B in skeletal development., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Marom et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
37. Break-induced replication underlies formation of inverted triplications and generates unexpected diversity in haplotype structures.
- Author
-
Grochowski CM, Bengtsson JD, Du H, Gandhi M, Lun MY, Mehaffey MG, Park K, Höps W, Benito-Garagorri E, Hasenfeld P, Korbel JO, Mahmoud M, Paulin LF, Jhangiani SN, Muzny DM, Fatih JM, Gibbs RA, Pendleton M, Harrington E, Juul S, Lindstrand A, Sedlazeck FJ, Pehlivan D, Lupski JR, and Carvalho CMB
- Abstract
Background: The duplication-triplication/inverted-duplication (DUP-TRP/INV-DUP) structure is a type of complex genomic rearrangement (CGR) hypothesized to result from replicative repair of DNA due to replication fork collapse. It is often mediated by a pair of inverted low-copy repeats (LCR) followed by iterative template switches resulting in at least two breakpoint junctions in cis . Although it has been identified as an important mutation signature of pathogenicity for genomic disorders and cancer genomes, its architecture remains unresolved and is predicted to display at least four structural variation (SV) haplotypes., Results: Here we studied the genomic architecture of DUP-TRP/INV-DUP by investigating the genomic DNA of 24 patients with neurodevelopmental disorders identified by array comparative genomic hybridization (aCGH) on whom we found evidence for the existence of 4 out of 4 predicted SV haplotypes. Using a combination of short-read genome sequencing (GS), long- read GS, optical genome mapping and StrandSeq the haplotype structure was resolved in 18 samples. This approach refined the point of template switching between inverted LCRs in 4 samples revealing a DNA segment of ∼2.2-5.5 kb of 100% nucleotide similarity. A prediction model was developed to infer the LCR used to mediate the non-allelic homology repair., Conclusions: These data provide experimental evidence supporting the hypothesis that inverted LCRs act as a recombinant substrate in replication-based repair mechanisms. Such inverted repeats are particularly relevant for formation of copy-number associated inversions, including the DUP-TRP/INV-DUP structures. Moreover, this type of CGR can result in multiple conformers which contributes to generate diverse SV haplotypes in susceptible loci .
- Published
- 2023
- Full Text
- View/download PDF
38. Genetic Sex Validation for Sample Tracking in Clinical Testing.
- Author
-
Hu J, Korchina V, Zouk H, Harden MV, Murdock D, Macbeth A, Harrison SM, Lennon N, Kovar C, Balasubramanian A, Zhang L, Chandanavelli G, Pasham D, Rowley R, Wiley K, Smith ME, Gordon A, Jarvik GP, Sleiman P, Kelly MA, Bland HT, Murugan M, Venner E, Boerwinkle E, Prows C, Mahanta L, Rehm HL, Gibbs RA, and Muzny DM
- Abstract
Objective: Data from DNA genotyping via a 96-SNP panel in a study of 25,015 clinical samples were utilized for quality control and tracking of sample identity in a clinical sequencing network. The study aimed to demonstrate the value of both the precise SNP tracking and the utility of the panel for predicting the sex-by-genotype of the participants, to identify possible sample mix-ups., Results: Precise SNP tracking showed no sample swap errors within the clinical testing laboratories. In contrast, when comparing predicted sex-by-genotype to the provided sex on the test requisition, we identified 110 inconsistencies from 25,015 clinical samples (0.44%), that had occurred during sample collection or accessioning. The genetic sex predictions were confirmed using additional SNP sites in the sequencing data or high-density genotyping arrays. It was determined that discrepancies resulted from clerical errors, samples from transgender participants and stem cell or bone marrow transplant patients along with undetermined sample mix-ups., Competing Interests: Competing interests JH, DM, MM, RAG, DMM disclose that the Baylor Genetics Laboratory is co-owned by Baylor College of Medicine. EV is cofounder of Codified Genomics, which provides variant interpretation services. DM has received consulting fees from Illumina. The remaining authors disclose they have no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
39. Functional Genomics of Gastrointestinal Escherichia coli Isolated from Patients with Cancer and Diarrhea.
- Author
-
Carter H, Clark J, Carlin LG, Vaughan E, Rajan A, Olvera A, Yu X, Zeng XL, Kambal A, Holder M, Qin X, Gibbs RA, Petrosino JF, Muzny DM, Doddapaneni H, Menon VK, Hoffman KL, Meng Q, Ross MC, Javornik Cregeen SJ, Metcalf G, Jenq R, Blutt S, Estes MK, Maresso A, and Okhuysen PC
- Abstract
We describe the epidemiology and clinical characteristics of 29 patients with cancer and diarrhea in whom Enteroaggregative Escherichia coli (EAEC) was initially identified by GI BioFire panel multiplex. E. coli strains were successfully isolated from fecal cultures in 14 of 29 patients. Six of the 14 strains were identified as EAEC and 8 belonged to other diverse E. coli groups of unknown pathogenesis. We investigated these strains by their adherence to human intestinal organoids, cytotoxic responses, antibiotic resistance profile, full sequencing of their genomes, and annotation of their functional virulome. Interestingly, we discovered novel and enhanced adherence and aggregative patterns for several diarrheagenic pathotypes that were not previously seen when co-cultured with immortalized cell lines. EAEC isolates displayed exceptional adherence and aggregation to human colonoids compared not only to diverse GI E. coli , but also compared to prototype strains of other diarrheagenic E. coli . Some of the diverse E. coli strains that could not be classified as a conventional pathotype also showed an enhanced aggregative and cytotoxic response. Notably, we found a high carriage rate of antibiotic resistance genes in both EAEC strains and diverse GI E. coli isolates and observed a positive correlation between adherence to colonoids and the number of metal acquisition genes carried in both EAEC and the diverse E. coli strains. This work indicates that E. coli from cancer patients constitute strains of remarkable pathotypic and genomic divergence, including strains of unknown disease etiology with unique virulomes. Future studies will allow for the opportunity to re-define E. coli pathotypes with greater diagnostic accuracy and into more clinically relevant groupings.
- Published
- 2023
- Full Text
- View/download PDF
40. Longitudinal host transcriptional responses to SARS-CoV-2 infection in adults with extremely high viral load.
- Author
-
Avadhanula V, Creighton CJ, Ferlic-Stark L, Sucgang R, Zhang Y, Nagaraj D, Nicholson EG, Rajan A, Menon VK, Doddapaneni H, Muzny DM, Metcalf G, Cregeen SJJ, Hoffman KL, Gibbs RA, Petrosino J, and Piedra PA
- Abstract
Current understanding of viral dynamics of SARS-CoV-2 and host responses driving the pathogenic mechanisms in COVID-19 is rapidly evolving. Here, we conducted a longitudinal study to investigate gene expression patterns during acute SARS-CoV-2 illness. Cases included SARS-CoV-2 infected individuals with extremely high viral loads early in their illness, individuals having low SARS-CoV-2 viral loads early in their infection, and individuals testing negative for SARS-CoV-2. We could identify widespread transcriptional host responses to SARS-CoV-2 infection that were initially most strongly manifested in patients with extremely high initial viral loads, then attenuating within the patient over time as viral loads decreased. Genes correlated with SARS-CoV-2 viral load over time were similarly differentially expressed across independent datasets of SARS-CoV-2 infected lung and upper airway cells, from both in vitro systems and patient samples. We also generated expression data on the human nose organoid model during SARS-CoV-2 infection. The human nose organoid-generated host transcriptional response captured many aspects of responses observed in the above patient samples, while suggesting the existence of distinct host responses to SARS-CoV-2 depending on the cellular context, involving both epithelial and cellular immune responses. Our findings provide a catalog of SARS-CoV-2 host response genes changing over time., Competing Interests: Conflict of Interest statement: The authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
41. The genomic landscape of familial glioma.
- Author
-
Choi DJ, Armstrong G, Lozzi B, Vijayaraghavan P, Plon SE, Wong TC, Boerwinkle E, Muzny DM, Chen HC, Gibbs RA, Ostrom QT, Melin B, Deneen B, Bondy ML, Bainbridge MN, Amos CI, Barnholtz-Sloan JS, Bernstein JL, Claus EB, Houlston RS, Il'yasova D, Jenkins RB, Johansen C, Lachance D, Lai R, Melin BS, Merrell RT, Olson SH, Sadetzki S, Schildkraut J, Shete S, Ambrose JC, Arumugam P, Bevers R, Bleda M, Boardman-Pretty F, Boustred CR, Brittain H, Brown MA, Caulfield MJ, Chan GC, Giess A, Griffin JN, Hamblin A, Henderson S, Hubbard TJP, Jackson R, Jones LJ, Kasperaviciute D, Kayikci M, Kousathanas A, Lahnstein L, Lakey A, Leigh SEA, Leong IUS, Lopez FJ, Maleady-Crowe F, McEntagart M, Minneci F, Mitchell J, Moutsianas L, Mueller M, Murugaesu N, Need AC, O'Donovan P, Odhams CA, Patch C, Perez-Gil D, Pereira MB, Pullinger J, Rahim T, Rendon A, Rogers T, Savage K, Sawant K, Scott RH, Siddiq A, Sieghart A, Smith SC, Sosinsky A, Stuckey A, Tanguy M, Taylor Tavares AL, Thomas ERA, Thompson SR, Tucci A, Welland MJ, Williams E, Witkowska K, Wood SM, and Zarowiecki M
- Subjects
- Humans, Genomics, Genetic Predisposition to Disease, Whole Genome Sequencing, Calcium-Binding Proteins genetics, DNA-Binding Proteins genetics, Tumor Suppressor Proteins genetics, Glioma genetics, Glioma pathology, Brain Neoplasms genetics, Brain Neoplasms pathology
- Abstract
Glioma is a rare brain tumor with a poor prognosis. Familial glioma is a subset of glioma with a strong genetic predisposition that accounts for approximately 5% of glioma cases. We performed whole-genome sequencing on an exploratory cohort of 203 individuals from 189 families with a history of familial glioma and an additional validation cohort of 122 individuals from 115 families. We found significant enrichment of rare deleterious variants of seven genes in both cohorts, and the most significantly enriched gene was HERC2 ( P = 0.0006). Furthermore, we identified rare noncoding variants in both cohorts that were predicted to affect transcription factor binding sites or cause cryptic splicing. Last, we selected a subset of discovered genes for validation by CRISPR knockdown screening and found that DMBT1, HP1BP3 , and ZCH7B3 have profound impacts on proliferation. This study performs comprehensive surveillance of the genomic landscape of familial glioma.
- Published
- 2023
- Full Text
- View/download PDF
42. Identification of allele-specific KIV-2 repeats and impact on Lp(a) measurements for cardiovascular disease risk.
- Author
-
Behera S, Belyeu JR, Chen X, Paulin LF, Nguyen NQH, Newman E, Mahmoud M, Menon VK, Qi Q, Joshi P, Marcovina S, Rossi M, Roller E, Han J, Onuchic V, Avery CL, Ballantyne CM, Rodriguez CJ, Kaplan RC, Muzny DM, Metcalf GA, Gibbs R, Yu B, Boerwinkle E, Eberle MA, and Sedlazeck FJ
- Abstract
The abundance of Lp(a) protein holds significant implications for the risk of cardiovascular disease (CVD), which is directly impacted by the copy number (CN) of KIV-2, a 5.5 kbp sub-region. KIV-2 is highly polymorphic in the population and accurate analysis is challenging. In this study, we present the DRAGEN KIV-2 CN caller, which utilizes short reads. Data across 166 WGS show that the caller has high accuracy, compared to optical mapping and can further phase ~50% of the samples. We compared KIV-2 CN numbers to 24 previously postulated KIV-2 relevant SNVs, revealing that many are ineffective predictors of KIV-2 copy number. Population studies, including USA-based cohorts, showed distinct KIV-2 CN, distributions for European-, African-, and Hispanic-American populations and further underscored the limitations of SNV predictors. We demonstrate that the CN estimates correlate significantly with the available Lp(a) protein levels and that phasing is highly important., Competing Interests: Ethics declarations Competing Interests FJS receives research support from Illumina, PacBio and ONT. LP is funded from Genentech. JB, EN, MR, ER, JH and VO are employees from Illumina. XC and ME are employees from PacBio. VM is employed now at Genentech.
- Published
- 2023
- Full Text
- View/download PDF
43. Rare variant enrichment analysis supports GREB1L as a contributory driver gene in the etiology of Mayer-Rokitansky-Küster-Hauser syndrome.
- Author
-
Jolly A, Du H, Borel C, Chen N, Zhao S, Grochowski CM, Duan R, Fatih JM, Dawood M, Salvi S, Jhangiani SN, Muzny DM, Koch A, Rouskas K, Glentis S, Deligeoroglou E, Bacopoulou F, Wise CA, Dietrich JE, Van den Veyver IB, Dimas AS, Brucker S, Sutton VR, Gibbs RA, Antonarakis SE, Wu N, Coban-Akdemir ZH, Zhu L, Posey JE, and Lupski JR
- Subjects
- Female, Humans, Uterus abnormalities, 46, XX Disorders of Sex Development genetics, Urogenital Abnormalities
- Abstract
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is characterized by aplasia of the female reproductive tract; the syndrome can include renal anomalies, absence or dysgenesis, and skeletal anomalies. While functional models have elucidated several candidate genes, only WNT4 (MIM: 603490) variants have been definitively associated with a subtype of MRKH with hyperandrogenism (MIM: 158330). DNA from 148 clinically diagnosed MRKH probands across 144 unrelated families and available family members from North America, Europe, and South America were exome sequenced (ES) and by family-based genomics analyzed for rare likely deleterious variants. A replication cohort consisting of 442 Han Chinese individuals with MRKH was used to further reproduce GREB1L findings in diverse genetic backgrounds. Proband and OMIM phenotypes annotated using the Human Phenotype Ontology were analyzed to quantitatively delineate the phenotypic spectrum associated with GREB1L variant alleles found in our MRKH cohort and those previously published. This study reports 18 novel GREB1L variant alleles, 16 within a multiethnic MRKH cohort and two within a congenital scoliosis cohort. Cohort-wide analyses for a burden of rare variants within a single gene identified likely damaging variants in GREB1L (MIM: 617782), a known disease gene for renal hypoplasia and uterine abnormalities (MIM: 617805), in 16 of 590 MRKH probands. GREB1L variant alleles, including a CNV null allele, were found in 8 MRKH type 1 probands and 8 MRKH type II probands. This study used quantitative phenotypic analyses in a worldwide multiethnic cohort to identify and strengthen the association of GREB1L to isolated uterine agenesis (MRKH type I) and syndromic MRKH type II., Competing Interests: J.R.L. has stock ownership in 23andMe and is a co-inventor on multiple U.S. and European patents related to molecular diagnostics for inherited neuropathies, genomic disorders, eye diseases, and bacterial genomic fingerprinting. The Department of Molecular and Human Genetics at Baylor College of Medicine derives revenue from the chromosomal microarray analysis and clinical genomic sequencing (both ES and WGS) offered in the Baylor Genetics Laboratory (http://bmgl.com). J.R.L. and I.B.V. serve on the Scientific Advisory Board of BG. S.E.A. is the co-founder and CEO of Medigenome, The Swiss Institute of Genomic Medicine., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
44. Modeling nonsegmented negative-strand RNA virus (NNSV) transcription with ejective polymerase collisions and biased diffusion.
- Author
-
Piedra FA, Henke D, Rajan A, Muzny DM, Doddapaneni H, Menon VK, Hoffman KL, Ross MC, Javornik Cregeen SJ, Metcalf G, Gibbs RA, Petrosino JF, Avadhanula V, and Piedra PA
- Abstract
Infections by non-segmented negative-strand RNA viruses (NNSV) are widely thought to entail gradient gene expression from the well-established existence of a single promoter at the 3' end of the viral genome and the assumption of constant transcriptional attenuation between genes. But multiple recent studies show viral mRNA levels in infections by respiratory syncytial virus (RSV), a major human pathogen and member of NNSV, that are inconsistent with a simple gradient. Here we integrate known and newly predicted phenomena into a biophysically reasonable model of NNSV transcription. Our model succeeds in capturing published observations of respiratory syncytial virus and vesicular stomatitis virus (VSV) mRNA levels. We therefore propose a novel understanding of NNSV transcription based on the possibility of ejective polymerase-polymerase collisions and, in the case of RSV, biased polymerase diffusion., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Piedra, Henke, Rajan, Muzny, Doddapaneni, Menon, Hoffman, Ross, Javornik Cregeen, Metcalf, Gibbs, Petrosino, Avadhanula and Piedra.)
- Published
- 2023
- Full Text
- View/download PDF
45. xAtlas: scalable small variant calling across heterogeneous next-generation sequencing experiments.
- Author
-
Farek J, Hughes D, Salerno W, Zhu Y, Pisupati A, Mansfield A, Krasheninina O, English AC, Metcalf G, Boerwinkle E, Muzny DM, Gibbs R, Khan Z, and Sedlazeck FJ
- Subjects
- Genome, INDEL Mutation, High-Throughput Nucleotide Sequencing methods, Polymorphism, Single Nucleotide, Algorithms, Software
- Abstract
Background: The growing volume and heterogeneity of next-generation sequencing (NGS) data complicate the further optimization of identifying DNA variation, especially considering that curated high-confidence variant call sets frequently used to validate these methods are generally developed from the analysis of comparatively small and homogeneous sample sets., Findings: We have developed xAtlas, a single-sample variant caller for single-nucleotide variants (SNVs) and small insertions and deletions (indels) in NGS data. xAtlas features rapid runtimes, support for CRAM and gVCF file formats, and retraining capabilities. xAtlas reports SNVs with 99.11% recall and 98.43% precision across a reference HG002 sample at 60× whole-genome coverage in less than 2 CPU hours. Applying xAtlas to 3,202 samples at 30× whole-genome coverage from the 1000 Genomes Project achieves an average runtime of 1.7 hours per sample and a clear separation of the individual populations in principal component analysis across called SNVs., Conclusions: xAtlas is a fast, lightweight, and accurate SNV and small indel calling method. Source code for xAtlas is available under a BSD 3-clause license at https://github.com/jfarek/xatlas., (© The Author(s) 2023. Published by Oxford University Press GigaScience.)
- Published
- 2022
- Full Text
- View/download PDF
46. Long read sequencing and expression studies of AHDC1 deletions in Xia-Gibbs syndrome reveal a novel genetic regulatory mechanism.
- Author
-
Chander V, Mahmoud M, Hu J, Dardas Z, Grochowski CM, Dawood M, Khayat MM, Li H, Li S, Jhangiani S, Korchina V, Shen H, Weissenberger G, Meng Q, Gingras MC, Muzny DM, Doddapaneni H, Posey JE, Lupski JR, Sabo A, Murdock DR, Sedlazeck FJ, and Gibbs RA
- Subjects
- Humans, DNA-Binding Proteins genetics, Endoribonucleases, Phosphoprotein Phosphatases, Qa-SNARE Proteins, RNA-Binding Proteins, Sphingomyelin Phosphodiesterase, Abnormalities, Multiple genetics, Intellectual Disability genetics, Musculoskeletal Abnormalities, Neurodevelopmental Disorders genetics
- Abstract
Xia-Gibbs syndrome (XGS; MIM# 615829) is a rare mendelian disorder characterized by Development Delay (DD), intellectual disability (ID), and hypotonia. Individuals with XGS typically harbor de novo protein-truncating mutations in the AT-Hook DNA binding motif containing 1 (AHDC1) gene, although some missense mutations can also cause XGS. Large de novo heterozygous deletions that encompass the AHDC1 gene have also been ascribed as diagnostic for the disorder, without substantial evidence to support their pathogenicity. We analyzed 19 individuals with large contiguous deletions involving AHDC1, along with other genes. One individual bore the smallest known contiguous AHDC1 deletion (∼350 Kb), encompassing eight other genes within chr1p36.11 (Feline Gardner-Rasheed, IFI6, FAM76A, STX12, PPP1R8, THEMIS2, RPA2, SMPDL3B) and terminating within the first intron of AHDC1. The breakpoint junctions and phase of the deletion were identified using both short and long read sequencing (Oxford Nanopore). Quantification of RNA expression patterns in whole blood revealed that AHDC1 exhibited a mono-allelic expression pattern with no deficiency in overall AHDC1 expression levels, in contrast to the other deleted genes, which exhibited a 50% reduction in mRNA expression. These results suggest that AHDC1 expression in this individual is compensated by a novel regulatory mechanism and advances understanding of mutational and regulatory mechanisms in neurodevelopmental disorders., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
47. Proteogenomic Markers of Chemotherapy Resistance and Response in Triple-Negative Breast Cancer.
- Author
-
Anurag M, Jaehnig EJ, Krug K, Lei JT, Bergstrom EJ, Kim BJ, Vashist TD, Huynh AMT, Dou Y, Gou X, Huang C, Shi Z, Wen B, Korchina V, Gibbs RA, Muzny DM, Doddapaneni H, Dobrolecki LE, Rodriguez H, Robles AI, Hiltke T, Lewis MT, Nangia JR, Nemati Shafaee M, Li S, Hagemann IS, Hoog J, Lim B, Osborne CK, Mani DR, Gillette MA, Zhang B, Echeverria GV, Miles G, Rimawi MF, Carr SA, Ademuyiwa FO, Satpathy S, and Ellis MJ
- Subjects
- Humans, Carboplatin, Proteomics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoadjuvant Therapy, X-ray Repair Cross Complementing Protein 1, Triple Negative Breast Neoplasms drug therapy, Proteogenomics
- Abstract
Microscaled proteogenomics was deployed to probe the molecular basis for differential response to neoadjuvant carboplatin and docetaxel combination chemotherapy for triple-negative breast cancer (TNBC). Proteomic analyses of pretreatment patient biopsies uniquely revealed metabolic pathways, including oxidative phosphorylation, adipogenesis, and fatty acid metabolism, that were associated with resistance. Both proteomics and transcriptomics revealed that sensitivity was marked by elevation of DNA repair, E2F targets, G2-M checkpoint, interferon-gamma signaling, and immune-checkpoint components. Proteogenomic analyses of somatic copy-number aberrations identified a resistance-associated 19q13.31-33 deletion where LIG1, POLD1, and XRCC1 are located. In orthogonal datasets, LIG1 (DNA ligase I) gene deletion and/or low mRNA expression levels were associated with lack of pathologic complete response, higher chromosomal instability index (CIN), and poor prognosis in TNBC, as well as carboplatin-selective resistance in TNBC preclinical models. Hemizygous loss of LIG1 was also associated with higher CIN and poor prognosis in other cancer types, demonstrating broader clinical implications., Significance: Proteogenomic analysis of triple-negative breast tumors revealed a complex landscape of chemotherapy response associations, including a 19q13.31-33 somatic deletion encoding genes serving lagging-strand DNA synthesis (LIG1, POLD1, and XRCC1), that correlate with lack of pathologic response, carboplatin-selective resistance, and, in pan-cancer studies, poor prognosis and CIN. This article is highlighted in the In This Issue feature, p. 2483., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
48. Clinical and molecular features of pediatric cancer patients with Lynch syndrome.
- Author
-
Scollon S, Eldomery MK, Reuther J, Lin FY, Potter SL, Desrosiers L, McClain KL, Smith V, Su JM, Venkatramani R, Hu J, Korchina V, Zarrin-Khameh N, Gibbs RA, Muzny DM, Eng C, Roy A, Parsons DW, and Plon SE
- Subjects
- Brain Neoplasms, Child, Colorectal Neoplasms, DNA Mismatch Repair genetics, DNA-Binding Proteins genetics, Germ-Line Mutation, Humans, MutL Protein Homolog 1 genetics, MutS Homolog 2 Protein genetics, Neoplastic Syndromes, Hereditary, Colorectal Neoplasms, Hereditary Nonpolyposis diagnosis, Colorectal Neoplasms, Hereditary Nonpolyposis genetics
- Abstract
Background: The association of childhood cancer with Lynch syndrome is not established compared with the significant pediatric cancer risk in recessive constitutional mismatch repair deficiency syndrome (CMMRD)., Procedure: We describe the clinical features, germline analysis, and tumor genomic profiling of patients with Lynch syndrome among patients enrolled in pediatric cancer genomic studies., Results: There were six of 773 (0.8%) pediatric patients with solid tumors identified with Lynch syndrome, defined as a germline heterozygous pathogenic variant in one of the mismatch repair (MMR) genes (three with MSH6, two with MLH1, and one with MSH2). Tumor analysis demonstrated evidence for somatic second hits and/or increased tumor mutation burden in three of four patients with available tumor with potential implications for therapy and identification of at-risk family members. Only one patient met current guidelines for pediatric cancer genetics evaluation at the time of tumor diagnosis., Conclusion: Approximately 1% of children with cancer have Lynch syndrome, which is missed with current referral guidelines, suggesting the importance of adding MMR genes to tumor and hereditary pediatric cancer panels. Tumor analysis may provide the first suggestion of an underlying cancer predisposition syndrome and is useful in distinguishing between Lynch syndrome and CMMRD., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
49. Author Correction: Comparative and demographic analysis of orang-utan genomes.
- Author
-
Locke DP, Hillier LW, Warren WC, Worley KC, Nazareth LV, Muzny DM, Yang SP, Wang Z, Chinwalla AT, Minx P, Mitreva M, Cook L, Delehaunty KD, Fronick C, Schmidt H, Fulton LA, Fulton RS, Nelson JO, Magrini V, Pohl C, Graves TA, Markovic C, Cree A, Dinh HH, Hume J, Kovar CL, Fowler GR, Lunter G, Meader S, Heger A, Ponting CP, Marques-Bonet T, Alkan C, Chen L, Cheng Z, Kidd JM, Eichler EE, White S, Searle S, Vilella AJ, Chen Y, Flicek P, Ma J, Raney B, Suh B, Burhans R, Herrero J, Haussler D, Faria R, Fernando O, Darré F, Farré D, Gazave E, Oliva M, Navarro A, Roberto R, Capozzi O, Archidiacono N, Della Valle G, Purgato S, Rocchi M, Konkel MK, Walker JA, Ullmer B, Batzer MA, Smit AFA, Hubley R, Casola C, Schrider DR, Hahn MW, Quesada V, Puente XS, Ordoñez GR, López-Otín C, Vinar T, Brejova B, Ratan A, Harris RS, Miller W, Kosiol C, Lawson HA, Taliwal V, Martins AL, Siepel A, RoyChoudhury A, Ma X, Degenhardt J, Bustamante CD, Gutenkunst RN, Mailund T, Dutheil JY, Hobolth A, Schierup MH, Ryder OA, Yoshinaga Y, de Jong PJ, Weinstock GM, Rogers J, Mardis ER, Gibbs RA, and Wilson RK
- Published
- 2022
- Full Text
- View/download PDF
50. Phenotypic and mutational spectrum of ROR2-related Robinow syndrome.
- Author
-
Lima AR, Ferreira BM, Zhang C, Jolly A, Du H, White JJ, Dawood M, Lins TC, Chiabai MA, van Beusekom E, Cordoba MS, Caldas Rosa ECC, Kayserili H, Kimonis V, Wu E, Mellado C, Aggarwal V, Richieri-Costa A, Brunoni D, Canó TM, Jorge AAL, Kim CA, Honjo R, Bertola DR, Dandalo-Girardi RM, Bayram Y, Gezdirici A, Yilmaz-Gulec E, Gumus E, Yilmaz GC, Okamoto N, Ohashi H, Coban-Akdemir Z, Mitani T, Jhangiani SN, Muzny DM, Regattieri NAP, Pogue R, Pereira RW, Otto PA, Gibbs RA, Ali BR, van Bokhoven H, Brunner HG, Sutton VR, Lupski JR, Vianna-Morgante AM, Carvalho CMB, and Mazzeu JF
- Subjects
- Genes, Recessive, Humans, Male, Phenotype, Craniofacial Abnormalities diagnosis, Craniofacial Abnormalities genetics, Dwarfism diagnosis, Dwarfism genetics, Limb Deformities, Congenital diagnosis, Limb Deformities, Congenital genetics, Receptor Tyrosine Kinase-like Orphan Receptors genetics, Urogenital Abnormalities diagnosis, Urogenital Abnormalities genetics
- Abstract
Robinow syndrome is characterized by a triad of craniofacial dysmorphisms, disproportionate-limb short stature, and genital hypoplasia. A significant degree of phenotypic variability seems to correlate with different genes/loci. Disturbances of the noncanonical WNT-pathway have been identified as the main cause of the syndrome. Biallelic variants in ROR2 cause an autosomal recessive form of the syndrome with distinctive skeletal findings. Twenty-two patients with a clinical diagnosis of autosomal recessive Robinow syndrome were screened for variants in ROR2 using multiple molecular approaches. We identified 25 putatively pathogenic ROR2 variants, 16 novel, including single nucleotide variants and exonic deletions. Detailed phenotypic analyses revealed that all subjects presented with a prominent forehead, hypertelorism, short nose, abnormality of the nasal tip, brachydactyly, mesomelic limb shortening, short stature, and genital hypoplasia in male patients. A total of 19 clinical features were present in more than 75% of the subjects, thus pointing to an overall uniformity of the phenotype. Disease-causing variants in ROR2, contribute to a clinically recognizable autosomal recessive trait phenotype with multiple skeletal defects. A comprehensive quantitative clinical evaluation of this cohort delineated the phenotypic spectrum of ROR2-related Robinow syndrome. The identification of exonic deletion variant alleles further supports the contention of a loss-of-function mechanism in the etiology of the syndrome., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.