96 results on '"Prigmore E"'
Search Results
2. Clinically-relevant postzygotic mosaicism in parents and children with developmental disorders in trio exome sequencing data
- Author
-
Wright, C. F., Prigmore, E., Rajan, D., Handsaker, J., McRae, J., Kaplanis, J., Fitzgerald, T. W., FitzPatrick, D. R., Firth, H. V., and Hurles, M. E.
- Published
- 2019
- Full Text
- View/download PDF
3. Large-scale discovery of novel genetic causes of developmental disorders
- Author
-
Fitzgerald, T. W., Gerety, S. S., Jones, W. D., van Kogelenberg, M., King, D. A., McRae, J., Morley, K. I., Parthiban, V., Al-Turki, S., Ambridge, K., Barrett, D. M., Bayzetinova, T., Clayton, S., Coomber, E. L., Gribble, S., Jones, P., Krishnappa, N., Mason, L. E., Middleton, A., Miller, R., Prigmore, E., Rajan, D., Sifrim, A., Tivey, A. R., Ahmed, M., Akawi, N., Andrews, R., Anjum, U., Archer, H., Armstrong, R., Balasubramanian, M., Banerjee, R., Baralle, D., Batstone, P., Baty, D., Bennett, C., Berg, J., Bernhard, B., Bevan, A. P., Blair, E., Blyth, M., Bohanna, D., Bourdon, L., Bourn, D., Brady, A., Bragin, E., Brewer, C., Brueton, L., Brunstrom, K., Bumpstead, S. J., Bunyan, D. J., Burn, J., Burton, J., Canham, N., Castle, B., Chandler, K., Clasper, S., Clayton-Smith, J., Cole, T., Collins, A., Collinson, M. N., Connell, F., Cooper, N., Cox, H., Cresswell, L., Cross, G., Crow, Y., DʼAlessandro, M., Dabir, T., Davidson, R., Davies, S., Dean, J., Deshpande, C., Devlin, G., Dixit, A., Dominiczak, A., Donnelly, C., Donnelly, D., Douglas, A., Duncan, A., Eason, J., Edkins, S., Ellard, S., Ellis, P., Elmslie, F., Evans, K., Everest, S., Fendick, T., Fisher, R., Flinter, F., Foulds, N., Fryer, A., Fu, B., Gardiner, C., Gaunt, L., Ghali, N., Gibbons, R., Pereira, Gomes S. L., Goodship, J., Goudie, D., Gray, E., Greene, P., Greenhalgh, L., Harrison, L., Hawkins, R., Hellens, S., Henderson, A., Hobson, E., Holden, S., Holder, S., Hollingsworth, G., Homfray, T., Humphreys, M., Hurst, J., Ingram, S., Irving, M., Jarvis, J., Jenkins, L., Johnson, D., Jones, D., Jones, E., Josifova, D., Joss, S., Kaemba, B., Kazembe, S., Kerr, B., Kini, U., Kinning, E., Kirby, G., Kirk, C., Kivuva, E., Kraus, A., Kumar, D., Lachlan, K., Lam, W., Lampe, A., Langman, C., Lees, M., Lim, D., Lowther, G., Lynch, S. A., Magee, A., Maher, E., Mansour, S., Marks, K., Martin, K., Maye, U., McCann, E., McConnell, V., McEntagart, M., McGowan, R., McKay, K., McKee, S., McMullan, D. J., McNerlan, S., Mehta, S., Metcalfe, K., Miles, E., Mohammed, S., Montgomery, T., Moore, D., Morgan, S., Morris, A., Morton, J., Mugalaasi, H., Murday, V., Nevitt, L., Newbury-Ecob, R., Norman, A., OʼShea, R., Ogilvie, C., Park, S., Parker, M. J., Patel, C., Paterson, J., Payne, S., Phipps, J., Pilz, D. T., Porteous, D., Pratt, N., Prescott, K., Price, S., Pridham, A., Procter, A., Purnell, H., Ragge, N., Rankin, J., Raymond, L., Rice, D., Robert, L., Roberts, E., Roberts, G., Roberts, J., Roberts, P., Ross, A., Rosser, E., Saggar, A., Samant, S., Sandford, R., Sarkar, A., Schweiger, S., Scott, C., Scott, R., Selby, A., Seller, A., Sequeira, C., Shannon, N., Sharif, S., Shaw-Smith, C., Shearing, E., Shears, D., Simonic, I., Simpkin, D., Singzon, R., Skitt, Z., Smith, A., Smith, B., Smith, K., Smithson, S., Sneddon, L., Splitt, M., Squires, M., Stewart, F., Stewart, H., Suri, M., Sutton, V., Swaminathan, G. J., Sweeney, E., Tatton-Brown, K., Taylor, C., Taylor, R., Tein, M., Temple, I. K., Thomson, J., Tolmie, J., Torokwa, A., Treacy, B., Turner, C., Turnpenny, P., Tysoe, C., Vandersteen, A., Vasudevan, P., Vogt, J., Wakeling, E., Walker, D., Waters, J., Weber, A., Wellesley, D., Whiteford, M., Widaa, S., Wilcox, S., Williams, D., Williams, N., Woods, G., Wragg, C., Wright, M., Yang, F., Yau, M., Carter, N. P., Parker, M., Firth, H. V., FitzPatrick, D. R., Wright, C. F., Barrett, J. C., and Hurles, M. E.
- Published
- 2015
- Full Text
- View/download PDF
4. Single-cell multi-omics analysis of the immune response in COVID-19
- Author
-
Stephenson, E, Reynolds, G, Botting, RA, Calero-Nieto, FJ, Morgan, MD, Tuong, ZK, Bach, K, Sungnak, W, Worlock, KB, Yoshida, M, Kumasaka, N, Kania, K, Engelbert, J, Olabi, B, Spegarova, JS, Wilson, NK, Mende, N, Jardine, L, Gardner, LCS, Goh, I, Horsfall, D, McGrath, J, Webb, S, Mather, MW, Lindeboom, RGH, Dann, E, Huang, N, Polanski, K, Prigmore, E, Gothe, F, Scott, J, Payne, RP, Baker, KF, Hanrath, AT, van der Loeff, ICDS, Barr, AS, Sanchez-Gonzalez, A, Bergamaschi, L, Mescia, F, Barnes, JL, Kilich, E, de Wilton, A, Saigal, A, Saleh, A, Janes, SM, Smith, CM, Gopee, N, Wilson, C, Coupland, P, Coxhead, JM, Kiselev, VY, van Dongen, S, Bacardit, J, King, HW, Rostron, AJ, Simpson, AJ, Hambleton, S, Laurenti, E, Lyons, PA, Meyer, KB, Nikolic, MZ, Duncan, CJA, Smith, KGC, Teichmann, SA, Clatworthy, MR, Marioni, JC, Gottgens, B, Haniffa, M, Stephenson, E, Reynolds, G, Botting, RA, Calero-Nieto, FJ, Morgan, MD, Tuong, ZK, Bach, K, Sungnak, W, Worlock, KB, Yoshida, M, Kumasaka, N, Kania, K, Engelbert, J, Olabi, B, Spegarova, JS, Wilson, NK, Mende, N, Jardine, L, Gardner, LCS, Goh, I, Horsfall, D, McGrath, J, Webb, S, Mather, MW, Lindeboom, RGH, Dann, E, Huang, N, Polanski, K, Prigmore, E, Gothe, F, Scott, J, Payne, RP, Baker, KF, Hanrath, AT, van der Loeff, ICDS, Barr, AS, Sanchez-Gonzalez, A, Bergamaschi, L, Mescia, F, Barnes, JL, Kilich, E, de Wilton, A, Saigal, A, Saleh, A, Janes, SM, Smith, CM, Gopee, N, Wilson, C, Coupland, P, Coxhead, JM, Kiselev, VY, van Dongen, S, Bacardit, J, King, HW, Rostron, AJ, Simpson, AJ, Hambleton, S, Laurenti, E, Lyons, PA, Meyer, KB, Nikolic, MZ, Duncan, CJA, Smith, KGC, Teichmann, SA, Clatworthy, MR, Marioni, JC, Gottgens, B, and Haniffa, M
- Abstract
Analysis of human blood immune cells provides insights into the coordinated response to viral infections such as severe acute respiratory syndrome coronavirus 2, which causes coronavirus disease 2019 (COVID-19). We performed single-cell transcriptome, surface proteome and T and B lymphocyte antigen receptor analyses of over 780,000 peripheral blood mononuclear cells from a cross-sectional cohort of 130 patients with varying severities of COVID-19. We identified expansion of nonclassical monocytes expressing complement transcripts (CD16+C1QA/B/C+) that sequester platelets and were predicted to replenish the alveolar macrophage pool in COVID-19. Early, uncommitted CD34+ hematopoietic stem/progenitor cells were primed toward megakaryopoiesis, accompanied by expanded megakaryocyte-committed progenitors and increased platelet activation. Clonally expanded CD8+ T cells and an increased ratio of CD8+ effector T cells to effector memory T cells characterized severe disease, while circulating follicular helper T cells accompanied mild disease. We observed a relative loss of IgA2 in symptomatic disease despite an overall expansion of plasmablasts and plasma cells. Our study highlights the coordinated immune response that contributes to COVID-19 pathogenesis and reveals discrete cellular components that can be targeted for therapy.
- Published
- 2021
5. Blood and immune development in human fetal bone marrow and Down syndrome
- Author
-
Jardine, L, Webb, S, Goh, I, Londono, MQ, Reynolds, G, Mather, M, Olabi, B, Stephenson, E, Botting, RA, Horsfall, D, Engelbert, J, Maunder, D, Mende, N, Murnane, C, Dann, E, McGrath, J, King, H, Kucinski, I, Queen, R, Carey, CD, Shrubsole, C, Poyner, E, Acres, M, Jones, C, Ness, T, Coulthard, R, Elliott, N, O'Byrne, S, Haltalli, MLR, Lawrence, JE, Lisgo, S, Balogh, P, Meyer, KB, Prigmore, E, Ambridge, K, Jain, MS, Efremova, M, Pickard, K, Creasey, T, Bacardit, J, Henderson, D, Coxhead, J, Filby, A, Hussain, R, Dixon, D, McDonald, D, Popescu, D-M, Kowalczyk, MS, Li, B, Ashenberg, O, Tabaka, M, Dionne, D, Tickle, TL, Slyper, M, Rozenblatt-Rosen, O, Regev, A, Behjati, S, Laurenti, E, Wilson, NK, Roy, A, Goettgens, B, Roberts, I, Teichmann, SA, Haniffa, M, Jardine, L, Webb, S, Goh, I, Londono, MQ, Reynolds, G, Mather, M, Olabi, B, Stephenson, E, Botting, RA, Horsfall, D, Engelbert, J, Maunder, D, Mende, N, Murnane, C, Dann, E, McGrath, J, King, H, Kucinski, I, Queen, R, Carey, CD, Shrubsole, C, Poyner, E, Acres, M, Jones, C, Ness, T, Coulthard, R, Elliott, N, O'Byrne, S, Haltalli, MLR, Lawrence, JE, Lisgo, S, Balogh, P, Meyer, KB, Prigmore, E, Ambridge, K, Jain, MS, Efremova, M, Pickard, K, Creasey, T, Bacardit, J, Henderson, D, Coxhead, J, Filby, A, Hussain, R, Dixon, D, McDonald, D, Popescu, D-M, Kowalczyk, MS, Li, B, Ashenberg, O, Tabaka, M, Dionne, D, Tickle, TL, Slyper, M, Rozenblatt-Rosen, O, Regev, A, Behjati, S, Laurenti, E, Wilson, NK, Roy, A, Goettgens, B, Roberts, I, Teichmann, SA, and Haniffa, M
- Abstract
Haematopoiesis in the bone marrow (BM) maintains blood and immune cell production throughout postnatal life. Haematopoiesis first emerges in human BM at 11-12 weeks after conception1,2, yet almost nothing is known about how fetal BM (FBM) evolves to meet the highly specialized needs of the fetus and newborn. Here we detail the development of FBM, including stroma, using multi-omic assessment of mRNA and multiplexed protein epitope expression. We find that the full blood and immune cell repertoire is established in FBM in a short time window of 6-7 weeks early in the second trimester. FBM promotes rapid and extensive diversification of myeloid cells, with granulocytes, eosinophils and dendritic cell subsets emerging for the first time. The substantial expansion of B lymphocytes in FBM contrasts with fetal liver at the same gestational age. Haematopoietic progenitors from fetal liver, FBM and cord blood exhibit transcriptional and functional differences that contribute to tissue-specific identity and cellular diversification. Endothelial cell types form distinct vascular structures that we show are regionally compartmentalized within FBM. Finally, we reveal selective disruption of B lymphocyte, erythroid and myeloid development owing to a cell-intrinsic differentiation bias as well as extrinsic regulation through an altered microenvironment in Down syndrome (trisomy 21).
- Published
- 2021
6. Applications of combined DNA microarray and chromosome sorting technologies
- Author
-
Gribble, S. M., Fiegler, H., Burford, D. C., Prigmore, E., Yang, F., Carr, P., Ng, B. L., Sun, T., Kamberov, E. S., Makarov, V. L., Langmore, J. P., and Carter, N. P.
- Published
- 2004
- Full Text
- View/download PDF
7. Human Genome Project Resources for Breakpoint Mapping
- Author
-
BURFORD, D, primary, GRIBBLE, S, additional, and PRIGMORE, E, additional
- Published
- 2006
- Full Text
- View/download PDF
8. Large-scale Molecular Analysis of a 34 Mb Interval on Chromosome 6q: Major Refinement of the RP25 Interval
- Author
-
Abd El-Aziz, M. M., Barragan, I., OʼDriscoll, C., Borrego, S., Abu-Safieh, L., Pieras, J. I., El-Ashry, M. F., Prigmore, E., Carter, N., Antinolo, G., and Bhattacharya, S. S.
- Published
- 2008
9. Ultra-high resolution array painting facilitates breakpoint sequencing
- Author
-
Gribble, S M, Kalaitzopoulos, D, Burford, D C, Prigmore, E, Selzer, R R, Ng, B L, Matthews, N S W, Porter, K M, Curley, R, Lindsay, S J, Baptista, J, Richmond, T A, and Carter, N P
- Published
- 2007
10. Quantifying the contribution of recessive coding variation to developmental disorders
- Author
-
Martin, H.C., Jones, W.D., McIntyre, R., Sanchez-Andrade, G., Sanderson, M., Stephenson, J.D., Jones, C.P., Handsaker, J., Gallone, G., Bruntraeger, M., McRae, J.F., Prigmore, E., Short, P., Niemi, M., Kaplanis, J., Radford, E.J., Akavvi, N., Balasubramanian, M., Dean, J., Horton, R., Hulbert, A., Johnson, D.S., Johnson, K., Kumar, D., Lynch, S.A., Mehta, S.G., Morton, J., Parker, M.J., Splitt, M., Turnpenny, P.D., Vasudevan, P.C., Wright, M., Bassett, A., Gerety, S.S., Wright, C.F., FitzPatrick, D.R., Firth, H.V., Hurles, M.E., Barrett, J.C., and Study, D.D.D.
- Subjects
genetic structures ,eye diseases - Abstract
We estimated the genome-wide contribution of recessive coding variation in 6040 families from the Deciphering Developmental Disorders study. The proportion of cases attributable to recessive coding variants was 3.6% in patients of European ancestry, compared with 50% explained by de novo coding mutations. It was higher (31%) in patients with Pakistani ancestry, owing to elevated autozygosity. Half of this recessive burden is attributable to known genes. We identified two genes not previously associated with recessive developmental disorders, KDM5B and EIF3F, and functionally validated them with mouse and cellular models. Our results suggest that recessive coding variants account for a small fraction of currently undiagnosed nonconsanguineous individuals, and that the role of noncoding variants, incomplete penetrance, and polygenic mechanisms need further exploration.
- Published
- 2018
11. The complex nature of constitutional de novo apparently balanced translocations in patients presenting with abnormal phenotypes
- Author
-
Gribble, S M, Prigmore, E, Burford, D C, Porter, K M, Ng, Bee Ling, Douglas, E J, Fiegler, H, Carr, P, Kalaitzopoulos, D, Clegg, S, Carter, N P, Sandstrom, R, Temple, I K, Youings, S A, Thomas, N S, Dennis, N R, Jacobs, P A, and Crolla, J A
- Published
- 2005
12. Array painting: a method for the rapid analysis of aberrant chromosomes using DNA microarrays
- Author
-
Fiegler, H, Gribble, S M, Burford, D C, Carr, P, Prigmore, E, Porter, K M, Clegg, S, Crolla, J A, Dennis, N R, Jacobs, P, and Carter, N P
- Published
- 2003
13. Finding Diagnostically Useful Patterns in Quantitative Phenotypic Data
- Author
-
Aitken, Stuart, primary, Firth, Helen V., additional, McRae, Jeremy, additional, Halachev, Mihail, additional, Kini, Usha, additional, Parker, Michael J., additional, Lees, Melissa M., additional, Lachlan, Katherine, additional, Sarkar, Ajoy, additional, Joss, Shelagh, additional, Splitt, Miranda, additional, McKee, Shane, additional, Németh, Andrea H., additional, Scott, Richard H., additional, Wright, Caroline F., additional, Marsh, Joseph A., additional, Hurles, Matthew E., additional, FitzPatrick, David R., additional, Fitzgerald, T.W., additional, Gerety, S.S., additional, Jones, W.D., additional, van Kogelenberg, M., additional, King, D.A., additional, McRae, J., additional, Morley, K.I., additional, Parthiban, V., additional, Al-Turki, S., additional, Ambridge, K., additional, Barrett, D.M., additional, Bayzetinova, T., additional, Clayton, S., additional, Coomber, E.L., additional, Gribble, S., additional, Jones, P., additional, Krishnappa, N., additional, Mason, L.E., additional, Middleton, A., additional, Miller, R., additional, Prigmore, E., additional, Rajan, D., additional, Sifrim, A., additional, Tivey, A.R., additional, Ahmed, M., additional, Akawi, N., additional, Andrews, R., additional, Anjum, U., additional, Archer, H., additional, Armstrong, R., additional, Balasubramanian, M., additional, Banerjee, R., additional, Barelle, D., additional, Batstone, P., additional, Baty, D., additional, Bennett, C., additional, Berg, J., additional, Bernhard, B., additional, Bevan, A.P., additional, Blair, E., additional, Blyth, M., additional, Bohanna, D., additional, Bourdon, L., additional, Bourn, D., additional, Brady, A., additional, Bragin, E., additional, Brewer, C., additional, Brueton, L., additional, Brunstrom, K., additional, Bumpstead, S.J., additional, Bunyan, D.J., additional, Burn, J., additional, Burton, J., additional, Canham, N., additional, Castle, B., additional, Chandler, K., additional, Clasper, S., additional, Clayton-Smith, J., additional, Cole, T., additional, Collins, A., additional, Collinson, M.N., additional, Connell, F., additional, Cooper, N., additional, Cox, H., additional, Cresswell, L., additional, Cross, G., additional, Crow, Y., additional, D’Alessandro, P.M., additional, Dabir, T., additional, Davidson, R., additional, Davies, S., additional, Dean, J., additional, Deshpande, C., additional, Devlin, G., additional, Dixit, A., additional, Dominiczak, A., additional, Donnelly, C., additional, Donnelly, D., additional, Douglas, A., additional, Duncan, A., additional, Eason, J., additional, Edkins, S., additional, Ellard, S., additional, Ellis, P., additional, Elmslie, F., additional, Evans, K., additional, Everest, S., additional, Fendick, T., additional, Fisher, R., additional, Flinter, F., additional, Foulds, N., additional, Fryer, A., additional, Fu, B., additional, Gardiner, C., additional, Gaunt, L., additional, Ghali, N., additional, Gibbons, R., additional, Pereira, S.L. Gomes, additional, Goodship, J., additional, Goudie, D., additional, Gray, E., additional, Greene, P., additional, Greenhalgh, L., additional, Harrison, L., additional, Hawkins, R., additional, Hellens, S., additional, Henderson, A., additional, Hobson, E., additional, Holden, S., additional, Holder, S., additional, Hollingsworth, G., additional, Homfray, T., additional, Humphreys, M., additional, Hurst, J., additional, Ingram, S., additional, Irving, M., additional, Jarvis, J., additional, Jenkins, L., additional, Johnson, D., additional, Jones, D., additional, Jones, E., additional, Josifova, D., additional, Joss, S., additional, Kaemba, B., additional, Kazembe, S., additional, Kerr, B., additional, Kini, U., additional, Kinning, E., additional, Kirby, G., additional, Kirk, C., additional, Kivuva, E., additional, Kraus, A., additional, Kumar, D., additional, Lachlan, K., additional, Lam, W., additional, Lampe, A., additional, Langman, C., additional, Lees, M., additional, Lim, D., additional, Lowther, G., additional, Lynch, S.A., additional, Magee, A., additional, Maher, E., additional, Mansour, S., additional, Marks, K., additional, Martin, K., additional, Maye, U., additional, McCann, E., additional, McConnell, V., additional, McEntagart, M., additional, McGowan, R., additional, McKay, K., additional, McKee, S., additional, McMullan, D.J., additional, McNerlan, S., additional, Mehta, S., additional, Metcalfe, K., additional, Miles, E., additional, Mohammed, S., additional, Montgomery, T., additional, Moore, D., additional, Morgan, S., additional, Morris, A., additional, Morton, J., additional, Mugalaasi, H., additional, Murday, V., additional, Nevitt, L., additional, Newbury-Ecob, R., additional, Norman, A., additional, O’Shea, R., additional, Ogilvie, C., additional, Park, S., additional, Parker, M.J., additional, Patel, C., additional, Paterson, J., additional, Payne, S., additional, Phipps, J., additional, Pilz, D.T., additional, Porteous, D., additional, Pratt, N., additional, Prescott, K., additional, Price, S., additional, Pridham, A., additional, Proctor, A., additional, Purnell, H., additional, Ragge, N., additional, Rankin, J., additional, Raymond, L., additional, Rice, D., additional, Robert, L., additional, Roberts, E., additional, Roberts, G., additional, Roberts, J., additional, Roberts, P., additional, Ross, A., additional, Rosser, E., additional, Saggar, A., additional, Samant, S., additional, Sandford, R., additional, Sarkar, A., additional, Schweiger, S., additional, Scott, C., additional, Scott, R., additional, Selby, A., additional, Seller, A., additional, Sequeira, C., additional, Shannon, N., additional, Sharif, S., additional, Shaw-Smith, C., additional, Shearing, E., additional, Shears, D., additional, Simonic, I., additional, Simpkin, D., additional, Singzon, R., additional, Skitt, Z., additional, Smith, A., additional, Smith, B., additional, Smith, K., additional, Smithson, S., additional, Sneddon, L., additional, Splitt, M., additional, Squires, M., additional, Stewart, F., additional, Stewart, H., additional, Suri, M., additional, Sutton, V., additional, Swaminathan, G.J., additional, Sweeney, E., additional, Tatton-Brown, K., additional, Taylor, C., additional, Taylor, R., additional, Tein, M., additional, Temple, I.K., additional, Thomson, J., additional, Tolmie, J., additional, Torokwa, A., additional, Treacy, B., additional, Turner, C., additional, Turnpenny, P., additional, Tysoe, C., additional, Vandersteen, A., additional, Vasudevan, P., additional, Vogt, J., additional, Wakeling, E., additional, Walker, D., additional, Waters, J., additional, Weber, A., additional, Wellesley, D., additional, Whiteford, M., additional, Widaa, S., additional, Wilcox, S., additional, Williams, D., additional, Williams, N., additional, Woods, G., additional, Wragg, C., additional, Wright, M., additional, Yang, F., additional, Yau, M., additional, Carter, N.P., additional, Parker, M., additional, Firth, H.V., additional, FitzPatrick, D.R., additional, Wright, C.F., additional, Barrett, J.C., additional, and Hurles, M.E., additional
- Published
- 2019
- Full Text
- View/download PDF
14. Prevalence and architecture of de novo mutations in developmental disorders
- Author
-
McRae, JF, Clayton, S, Fitzgerald, TW, Kaplanis, J, Prigmore, E, Rajan, D, Sifrim, A, Aitken, S, Akawi, N, Alvi, M, Ambridge, K, Barrett, DM, Bayzetinova, T, Jones, P, Jones, WD, King, D, Krishnappa, N, Mason, LE, Singh, T, Tivey, AR, Ahmed, M, Anjum, U, Archer, H, Armstrong, R, Awada, J, Balasubramanian, M, Banka, S, Baralle, D, Barnicoat, A, Batstone, P, Baty, D, Bennett, C, Berg, J, Bernhard, B, Bevan, AP, Bitner-Glindzicz, M, Blair, E, Blyth, M, Bohanna, D, Bourdon, L, Bourn, D, Bradley, L, Brady, A, Brent, S, Brewer, C, Brunstrom, K, Bunyan, DJ, Burn, J, Canham, N, Castle, B, Chandler, K, Chatzimichali, E, Cilliers, D, Clarke, A, Clasper, S, Clayton-Smith, J, Clowes, V, Coates, A, Cole, T, Colgiu, I, Collins, A, Collinson, MN, Connell, F, Cooper, N, Cox, H, Cresswell, L, Cross, G, Crow, Y, D’Alessandro, M, Dabir, T, Davidson, R, Davies, S, de Vries, D, Dean, J, Deshpande, C, Devlin, G, Dixit, A, Dobbie, A, Donaldson, A, Donnai, D, Donnelly, D, Donnelly, C, Douglas, A, Douzgou, S, Duncan, A, Eason, J, Ellard, S, Ellis, I, Elmslie, F, Evans, K, Everest, S, Fendick, T, Fisher, R, Flinter, F, Foulds, N, Fry, A, Fryer, A, Gardiner, C, Gaunt, L, Ghali, N, Gibbons, R, Gill, H, Goodship, J, Goudie, D, Gray, E, Green, A, Greene, P, Greenhalgh, L, Gribble, S, Harrison, R, Harrison, L, Harrison, V, Hawkins, R, He, L, Hellens, S, Henderson, A, Hewitt, S, Hildyard, L, Hobson, E, Holden, S, Holder, M, Holder, S, Hollingsworth, G, Homfray, T, Humphreys, M, Hurst, J, Hutton, B, Ingram, S, Irving, M, Islam, L, Jackson, A, Jarvis, J, Jenkins, L, Johnson, D, Jones, E, Josifova, D, Joss, S, Kaemba, B, Kazembe, S, Kelsell, R, Kerr, B, Kingston, H, Kini, U, Kinning, E, Kirby, G, Kirk, C, Kivuva, E, Kraus, A, Kumar, D, Kumar, VKA, Lachlan, K, Lam, W, Lampe, A, Langman, C, Lees, M, Lim, D, Longman, C, Lowther, G, Lynch, SA, Magee, A, Maher, E, Male, A, Mansour, S, Marks, K, Martin, K, Maye, U, McCann, E, McConnell, V, McEntagart, M, McGowan, R, McKay, K, McKee, S, McMullan, DJ, McNerlan, S, McWilliam, C, Mehta, S, Metcalfe, K, Middleton, A, Miedzybrodzka, Z, Miles, E, Mohammed, S, Montgomery, T, Moore, D, Morgan, S, Morton, J, Mugalaasi, H, Murday, V, Murphy, H, Naik, S, Nemeth, A, Nevitt, L, Newbury-Ecob, R, Norman, A, O’Shea, R, Ogilvie, C, Ong, K-R, Park, S-M, Parker, MJ, Patel, C, Paterson, J, Payne, S, Perrett, D, Phipps, J, Pilz, DT, Pollard, M, Pottinger, C, Poulton, J, Pratt, N, Prescott, K, Price, S, Pridham, A, Procter, A, Purnell, H, Quarrell, O, Ragge, N, Rahbari, R, Randall, J, Rankin, J, Raymond, L, Rice, D, Robert, L, Roberts, E, Roberts, J, Roberts, P, Roberts, G, Ross, A, Rosser, E, Saggar, A, Samant, S, Sampson, J, Sandford, R, Sarkar, A, Schweiger, S, Scott, R, Scurr, I, Selby, A, Seller, A, Sequeira, C, Shannon, N, Sharif, S, Shaw-Smith, C, Shearing, E, Shears, D, Sheridan, E, Simonic, I, Singzon, R, Skitt, Z, Smith, A, Smith, K, Smithson, S, Sneddon, L, Splitt, M, Squires, M, Stewart, F, Stewart, H, Straub, V, Suri, M, Sutton, V, Swaminathan, GJ, Sweeney, E, Tatton-Brown, K, Taylor, C, Taylor, R, Tein, M, Temple, IK, Thomson, J, Tischkowitz, M, Tomkins, S, Torokwa, A, Treacy, B, Turner, C, Turnpenny, P, Tysoe, C, Vandersteen, A, Varghese, V, Vasudevan, P, Vijayarangakannan, P, Vogt, J, Wakeling, E, Wallwark, S, Waters, J, Weber, A, Wellesley, D, Whiteford, M, Widaa, S, Wilcox, S, Wilkinson, E, Williams, D, Williams, N, Wilson, L, Woods, G, Wragg, C, Wright, M, Yates, L, Yau, M, Nellåker, C, Parker, M, Firth, HV, Wright, CF, FitzPatrick, DR, Barrett, JC, and Hurles, ME
- Subjects
Male ,Parents ,Heredity ,Developmental Disabilities ,GRIN2B ,POGZ ,Autoantigens ,SMAD4 ,CASK ,GATAD2B ,0302 clinical medicine ,TRIO ,SMARCA2 ,KCNH1 ,Average Faces ,CTNNB1 ,SCN1A ,Young adult ,Casein Kinase II ,Child ,AUTS2 ,MEF2C ,Exome ,ADNP ,Exome sequencing ,EP300 ,KCNQ2 ,KCNQ3 ,EHMT1 ,CNKSR2 ,CREBBP ,MYT1L ,MED13L ,CSNK2A1 ,Protein Phosphatase 2C ,PPP2R1A ,ZBTB18 ,CDKL5 ,WAC ,HNRNPU ,Cohort ,STXBP1 ,Medical genetics ,SYNGAP1 ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Sex characteristics ,AHDC1 ,SCN8A ,medicine.medical_specialty ,SLC6A1 ,FOXP1 ,USP9X ,Article ,ANKRD11 ,PUF60 ,BRAF ,03 medical and health sciences ,SATB2 ,SMC1A ,Intellectual Disability ,BCL11A ,GABRB3 ,IQSEC2 ,Humans ,TBL1XR1 ,TCF4 ,MSL3 ,TCF20 ,DNM1 ,EEF1A2 ,SUV420H1 ,DYRK1A ,SETD5 ,COL4A3BP ,CTCF ,CHD2 ,R1 ,CHD4 ,030104 developmental biology ,NAA10 ,HDAC8 ,Mutation ,KDM5B ,CHAMP1 ,PhenIcons ,030217 neurology & neurosurgery ,Transcription Factors ,0301 basic medicine ,ZMYND11 ,PTEN ,De novo mutation ,Chromosomal Proteins, Non-Histone ,PTPN11 ,ASXL1 ,Bioinformatics ,medicine.disease_cause ,ASXL3 ,Cohort Studies ,DEAD-box RNA Helicases ,CHD8 ,Prevalence ,QRICH1 ,KIF1A ,Genetics ,Sex Characteristics ,GNAI1 ,Multidisciplinary ,WDR45 ,Middle Aged ,KMT2A ,PPM1D ,MECP2 ,DNA-Binding Proteins ,PPP2R5D ,Phenotype ,PACS1 ,ras GTPase-Activating Proteins ,DDX3X ,Female ,FOXG1 ,SET ,Myeloid-Lymphoid Leukemia Protein ,Developmental Disease ,Adult ,KANSL1 ,Adolescent ,NFIX ,Nerve Tissue Proteins ,PURA ,Biology ,KAT6B ,KAT6A ,NSD1 ,PDHA1 ,ALG13 ,Young Adult ,Seizures ,CDC2 Protein Kinase ,medicine ,Journal Article ,QH426 ,Homeodomain Proteins ,ITPR1 ,DYNC1H1 ,GNAO1 ,Histone-Lysine N-Methyltransferase ,Sequence Analysis, DNA ,ZC4H2 ,ARID1B ,Repressor Proteins ,CNOT3 ,SCN2A ,SLC35A2 ,CDK13 - Abstract
Children with severe, undiagnosed developmental disorders (DDs) are enriched for damaging de novo mutations (DNMs) in developmentally important genes. We exome sequenced 4,294 families with children with DDs, and meta-analysed these data with published data on 3,287 children with similar disorders. We show that the most significant factors influencing the diagnostic yield of de novo mutations are the sex of the child, the relatedness of their parents and the age of both father and mother. We identified 95 genes enriched for damaging de novo mutation at genome-wide significance (P < 5 x 10-7), including fourteen genes for which compelling data for causation was previously lacking. The large number of genome-wide significant findings allow us to demonstrate that, at current cost differentials, exome sequencing has much greater power than genome sequencing for novel gene discovery in genetically heterogeneous disorders. We estimate that 42.5% of our cohort likely carry pathogenic de novo single nucleotide variants (SNVs) and indels in coding sequences, with approximately half operating by a loss-of-function mechanism, and the remainder being gain-of-function. We established that most haploinsufficient developmental disorders have already been identified, but that many gain-of-function disorders remain to be discovered. Extrapolating from the DDD cohort to the general population, we estimate that de novo dominant developmental disorders have an average birth prevalence of 1 in 168 to 1 in 377, depending on parental age.
- Published
- 2017
15. Large-scale discovery of novel genetic causes of developmental disorders
- Author
-
Fitzgerald, TW, Gerety, SS, Jones, WD, van Kogelenberg, M, King, DA, McRae, J, Morley, KI, Parthiban, V, Al-Turki, S, Ambridge, K, Barrett, DM, Bayzetinova, T, Clayton, S, Coomber, EL, Gribble, S, Jones, P, Krishnappa, N, Mason, LE, Middleton, A, Miller, R, Prigmore, E, Rajan, D, Sifrim, A, Tivey, AR, Ahmed, M, Akawi, N, Andrews, R, Anjum, U, Archer, H, Armstrong, R, Balasubramanian, M, Banerjee, R, Baralle, D, Batstone, P, Baty, D, Bennett, C, Berg, J, Bernhard, B, Bevan, AP, Blair, E, Blyth, M, Bohanna, D, Bourdon, L, Bourn, D, Brady, A, Bragin, E, Brewer, C, Brueton, L, Brunstrom, K, Bumpstead, SJ, Bunyan, DJ, Burn, J, Burton, J, Canham, N, Castle, B, Chandler, K, Clasper, S, Clayton-Smith, J, Cole, T, Collins, A, Collinson, MN, Connell, F, Cooper, N, Cox, H, Cresswell, L, Cross, G, Crow, Y, D'Alessandro, M, Dabir, T, Davidson, R, Davies, S, Dean, J, Deshpande, C, Devlin, G, Dixit, A, Dominiczak, A, Donnelly, C, Donnelly, D, Douglas, A, Duncan, A, Eason, J, Edkins, S, Ellard, S, Ellis, P, Elmslie, F, Evans, K, Everest, S, Fendick, T, Fisher, R, Flinter, F, Foulds, N, Fryer, A, Fu, B, Gardiner, C, Gaunt, L, Ghali, N, Gibbons, R, Pereira, SLG, Goodship, J, Goudie, D, Gray, E, Greene, P, Greenhalgh, L, Harrison, L, Hawkins, R, Hellens, S, Henderson, A, Hobson, E, Holden, S, Holder, S, Hollingsworth, G, Homfray, T, Humphreys, M, Hurst, J, Ingram, S, Irving, M, Jarvis, J, Jenkins, L, Johnson, D, Jones, D, Jones, E, Josifova, D, Joss, S, Kaemba, B, Kazembe, S, Kerr, B, Kini, U, Kinning, E, Kirby, G, Kirk, C, Kivuva, E, Kraus, A, Kumar, D, Lachlan, K, Lam, W, Lampe, A, Langman, C, Lees, M, Lim, D, Lowther, G, Lynch, SA, Magee, A, Maher, E, Mansour, S, Marks, K, Martin, K, Maye, U, McCann, E, McConnell, V, McEntagart, M, McGowan, R, McKay, K, McKee, S, McMullan, DJ, McNerlan, S, Mehta, S, Metcalfe, K, Miles, E, Mohammed, S, Montgomery, T, Moore, D, Morgan, S, Morris, A, Morton, J, Mugalaasi, H, Murday, V, Nevitt, L, Newbury-Ecob, R, Norman, A, O'Shea, R, Ogilvie, C, Park, S, Parker, MJ, Patel, C, Paterson, J, Payne, S, Phipps, J, Pilz, DT, Porteous, D, Pratt, N, Prescott, K, Price, S, Pridham, A, Procter, A, Purnell, H, Ragge, N, Rankin, J, Raymond, L, Rice, D, Robert, L, Roberts, E, Roberts, G, Roberts, J, Roberts, P, Ross, A, Rosser, E, Saggar, A, Samant, S, Sandford, R, Sarkar, A, Schweier, S, Scott, C, Scott, R, Selby, A, Seller, A, Sequeira, C, Shannon, N, Shanrif, S, Shaw-Smith, C, Shearing, E, Shears, D, Simonic, I, Simpkin, D, Singzon, R, Skitt, Z, Smith, A, Smith, B, Smith, K, Smithson, S, Sneddon, L, Splitt, M, Squires, M, Stewart, F, Stewart, H, Suri, M, Sutton, V, Swaminathan, GJ, Sweeney, E, Tatton-Brown, K, Taylor, C, Taylor, R, Tein, M, Temple, IK, Thomson, J, Tolmie, J, Torokwa, A, Treacy, B, Turner, C, Turnpenny, P, Tysoe, C, Vandersteen, A, Vasudevan, P, Vogt, J, Wakeling, E, Walker, D, Waters, J, Weber, A, Wellesley, D, Whiteford, M, Widaa, S, Wilcox, S, Williams, D, Williams, N, Woods, G, Wragg, C, Wright, M, Yang, F, Yau, M, Carter, NP, Parker, M, Firth, HV, FitzPatrick, DR, Wright, CF, Barrett, JC, Hurles, ME, Fitzgerald, TW, Gerety, SS, Jones, WD, van Kogelenberg, M, King, DA, McRae, J, Morley, KI, Parthiban, V, Al-Turki, S, Ambridge, K, Barrett, DM, Bayzetinova, T, Clayton, S, Coomber, EL, Gribble, S, Jones, P, Krishnappa, N, Mason, LE, Middleton, A, Miller, R, Prigmore, E, Rajan, D, Sifrim, A, Tivey, AR, Ahmed, M, Akawi, N, Andrews, R, Anjum, U, Archer, H, Armstrong, R, Balasubramanian, M, Banerjee, R, Baralle, D, Batstone, P, Baty, D, Bennett, C, Berg, J, Bernhard, B, Bevan, AP, Blair, E, Blyth, M, Bohanna, D, Bourdon, L, Bourn, D, Brady, A, Bragin, E, Brewer, C, Brueton, L, Brunstrom, K, Bumpstead, SJ, Bunyan, DJ, Burn, J, Burton, J, Canham, N, Castle, B, Chandler, K, Clasper, S, Clayton-Smith, J, Cole, T, Collins, A, Collinson, MN, Connell, F, Cooper, N, Cox, H, Cresswell, L, Cross, G, Crow, Y, D'Alessandro, M, Dabir, T, Davidson, R, Davies, S, Dean, J, Deshpande, C, Devlin, G, Dixit, A, Dominiczak, A, Donnelly, C, Donnelly, D, Douglas, A, Duncan, A, Eason, J, Edkins, S, Ellard, S, Ellis, P, Elmslie, F, Evans, K, Everest, S, Fendick, T, Fisher, R, Flinter, F, Foulds, N, Fryer, A, Fu, B, Gardiner, C, Gaunt, L, Ghali, N, Gibbons, R, Pereira, SLG, Goodship, J, Goudie, D, Gray, E, Greene, P, Greenhalgh, L, Harrison, L, Hawkins, R, Hellens, S, Henderson, A, Hobson, E, Holden, S, Holder, S, Hollingsworth, G, Homfray, T, Humphreys, M, Hurst, J, Ingram, S, Irving, M, Jarvis, J, Jenkins, L, Johnson, D, Jones, D, Jones, E, Josifova, D, Joss, S, Kaemba, B, Kazembe, S, Kerr, B, Kini, U, Kinning, E, Kirby, G, Kirk, C, Kivuva, E, Kraus, A, Kumar, D, Lachlan, K, Lam, W, Lampe, A, Langman, C, Lees, M, Lim, D, Lowther, G, Lynch, SA, Magee, A, Maher, E, Mansour, S, Marks, K, Martin, K, Maye, U, McCann, E, McConnell, V, McEntagart, M, McGowan, R, McKay, K, McKee, S, McMullan, DJ, McNerlan, S, Mehta, S, Metcalfe, K, Miles, E, Mohammed, S, Montgomery, T, Moore, D, Morgan, S, Morris, A, Morton, J, Mugalaasi, H, Murday, V, Nevitt, L, Newbury-Ecob, R, Norman, A, O'Shea, R, Ogilvie, C, Park, S, Parker, MJ, Patel, C, Paterson, J, Payne, S, Phipps, J, Pilz, DT, Porteous, D, Pratt, N, Prescott, K, Price, S, Pridham, A, Procter, A, Purnell, H, Ragge, N, Rankin, J, Raymond, L, Rice, D, Robert, L, Roberts, E, Roberts, G, Roberts, J, Roberts, P, Ross, A, Rosser, E, Saggar, A, Samant, S, Sandford, R, Sarkar, A, Schweier, S, Scott, C, Scott, R, Selby, A, Seller, A, Sequeira, C, Shannon, N, Shanrif, S, Shaw-Smith, C, Shearing, E, Shears, D, Simonic, I, Simpkin, D, Singzon, R, Skitt, Z, Smith, A, Smith, B, Smith, K, Smithson, S, Sneddon, L, Splitt, M, Squires, M, Stewart, F, Stewart, H, Suri, M, Sutton, V, Swaminathan, GJ, Sweeney, E, Tatton-Brown, K, Taylor, C, Taylor, R, Tein, M, Temple, IK, Thomson, J, Tolmie, J, Torokwa, A, Treacy, B, Turner, C, Turnpenny, P, Tysoe, C, Vandersteen, A, Vasudevan, P, Vogt, J, Wakeling, E, Walker, D, Waters, J, Weber, A, Wellesley, D, Whiteford, M, Widaa, S, Wilcox, S, Williams, D, Williams, N, Woods, G, Wragg, C, Wright, M, Yang, F, Yau, M, Carter, NP, Parker, M, Firth, HV, FitzPatrick, DR, Wright, CF, Barrett, JC, and Hurles, ME
- Abstract
Despite three decades of successful, predominantly phenotype-driven discovery of the genetic causes of monogenic disorders, up to half of children with severe developmental disorders of probable genetic origin remain without a genetic diagnosis. Particularly challenging are those disorders rare enough to have eluded recognition as a discrete clinical entity, those with highly variable clinical manifestations, and those that are difficult to distinguish from other, very similar, disorders. Here we demonstrate the power of using an unbiased genotype-driven approach to identify subsets of patients with similar disorders. By studying 1,133 children with severe, undiagnosed developmental disorders, and their parents, using a combination of exome sequencing and array-based detection of chromosomal rearrangements, we discovered 12 novel genes associated with developmental disorders. These newly implicated genes increase by 10% (from 28% to 31%) the proportion of children that could be diagnosed. Clustering of missense mutations in six of these newly implicated genes suggests that normal development is being perturbed by an activating or dominant-negative mechanism. Our findings demonstrate the value of adopting a comprehensive strategy, both genome-wide and nationwide, to elucidate the underlying causes of rare genetic disorders.
- Published
- 2015
16. The DNA sequence and biological annotation of human chromosome 1
- Author
-
Gregory, S. G., Barlow, K. F., McLay, K. E., Kaul, R., Swarbreck, D., Dunham, A., Scott, C. E., Howe, K. L., Woodfine, K., Spencer, C. C. A., Jones, M. C., Gillson, C., Searle, S., Zhou, Y., Kokocinski, F., McDonald, L., Evans, R., Phillips, K., Atkinson, A., Cooper, R., Jones, C., Hall, R. E., Andrews, T. D., Lloyd, C., Ainscough, R., Almeida, J. P., Ambrose, K. D., Anderson, F., Andrew, R. W., Ashwell, R. I. S., Aubin, K., Babbage, A. K., Bagguley, C. L., Bailey, J., Banerjee, R., Beasley, H., Bethel, G., Bird, C. P., Bray-Allen, S., Brown, J. Y., Brown, A. J., Bryant, S. P., Buckley, D., Burford, D. C., Burrill, W. D. H., Burton, J., Bye, J., Carder, C., Chapman, J. C., Clark, S. Y., Clarke, G., Clee, C., Clegg, S. M., Cobley, V., Collier, R. E., Corby, N., Coville, G. J., Davies, J., Deadman, R., Dhami, P., Dovey, O., Dunn, M., Earthrowl, M., Ellington, A. G., Errington, H., Faulkner, L. M., Frankish, A., Frankland, J., French, L., Garner, P., Garnett, J., Gay, L., Ghori, M. R. J., Gibson, R., Gilby, L. M., Gillett, W., Glithero, R. J., Grafham, D. V., Gribble, S. M., Griffiths, C., Griffiths-Jones, S., Grocock, R., Hammond, S., Harrison, E. S. I., Hart, E., Haugen, E., Heath, P. D., Holmes, S., Holt, K., Howden, P. J., Hunt, A. R., Hunt, S. E., Hunter, G., Isherwood, J., James, R., Johnson, C., Johnson, D., Joy, A., Kay, M., Kershaw, J. K., Kibukawa, M., Kimberley, A. M., King, A., Knights, A. J., Lad, H., Laird, G., Langford, C. F., Lawlor, S., Leongamornlert, D. A., Lloyd, D. M., Loveland, J., Lovell, J., Lush, M. J., Lyne, R., Martin, S., Mashreghi-Mohammadi, M., Matthews, L., Matthews, N. S. W., McLaren, S., Milne, S., Mistry, S., oore, M. J. F. M., Nickerson, T., O'Dell, C. N., Oliver, K., Palmeiri, A., Palmer, S. A., Pandian, R. D., Parker, A., Patel, D., Pearce, A. V., Peck, A. I., Pelan, S., Phelps, K., Phillimore, B. J., Plumb, R., Porter, K. M., Prigmore, E., Rajan, J., Raymond, C., Rouse, G., Saenphimmachak, C., Sehra, H. K., Sheridan, E., Shownkeen, R., Sims, S., Skuce, C. D., Smith, M., Steward, C., Subramanian, S., Sycamore, N., Tracey, A., Tromans, A., Van Helmond, Z., Wall J. M. Wallis, M., White, S., Whitehead, S. L., Wilkinson, J. E., Willey, D. L., Williams, H., Wilming, L., Wray, P. W., Wu, Z., Coulson, A., Vaudin, M., Sulston, J. E., Durbin, R., Hubbard, T., Wooster, R., Dunham, I., Carter, N. P., McVean, G., Ross, M. T., Harrow, J., Olson, M. V., Beck, S., Rogers, J., and Bentley, D. R.
- Subjects
Environmental issues ,Science and technology ,Zoology and wildlife conservation - Abstract
Author(s): S. G. Gregory [1]; K. F. Barlow [1]; K. E. McLay [1]; R. Kaul [1]; D. Swarbreck [1]; A. Dunham [1]; C. E. Scott [1]; K. L. Howe [1]; [...]
- Published
- 2006
- Full Text
- View/download PDF
17. Erratum: The DNA sequence and biological annotation of human chromosome 1
- Author
-
Gregory, S. G., primary, Barlow, K. F., additional, McLay, K. E., additional, Kaul, R., additional, Swarbreck, D., additional, Dunham, A., additional, Scott, C. E., additional, Howe, K. L., additional, Woodfine, K., additional, Spencer, C. C. A., additional, Jones, M. C., additional, Gillson, C., additional, Searle, S., additional, Zhou, Y., additional, Kokocinski, F., additional, McDonald, L., additional, Evans, R., additional, Phillips, K., additional, Atkinson, A., additional, Cooper, R., additional, Jones, C., additional, Hall, R. E., additional, Andrews, T. D., additional, Lloyd, C., additional, Ainscough, R., additional, Almeida, J. P., additional, Ambrose, K. D., additional, Anderson, F., additional, Andrew, R. W., additional, Ashwell, R. I. S., additional, Aubin, K., additional, Babbage, A. K., additional, Bagguley, C. L., additional, Bailey, J., additional, Banerjee, R., additional, Beasley, H., additional, Bethel, G., additional, Bird, C. P., additional, Bray-Allen, S., additional, Brown, J. Y., additional, Brown, A. J., additional, Bryant, S. P., additional, Buckley, D., additional, Burford, D. C., additional, Burrill, W. D. H., additional, Burton, J., additional, Bye, J., additional, Carder, C., additional, Chapman, J. C., additional, Clark, S. Y., additional, Clarke, G., additional, Clee, C., additional, Clegg, S. M., additional, Cobley, V., additional, Collier, R. E., additional, Corby, N., additional, Coville, G. J., additional, Davies, J., additional, Deadman, R., additional, Dhami, P., additional, Dovey, O., additional, Dunn, M., additional, Earthrowl, M., additional, Ellington, A. G., additional, Errington, H., additional, Faulkner, L. M., additional, Frankish, A., additional, Frankland, J., additional, French, L., additional, Garner, P., additional, Garnett, J., additional, Gay, L., additional, Ghori, M. R. J., additional, Gibson, R., additional, Gilby, L. M., additional, Gillett, W., additional, Glithero, R. J., additional, Grafham, D. V., additional, Gribble, S. M., additional, Griffiths, C., additional, Griffiths-Jones, S., additional, Grocock, R., additional, Hammond, S., additional, Harrison, E. S. I., additional, Hart, E., additional, Haugen, E., additional, Heath, P. D., additional, Holmes, S., additional, Holt, K., additional, Howden, P. J., additional, Hunt, A. R., additional, Hunt, S. E., additional, Hunter, G., additional, Isherwood, J., additional, James, R., additional, Johnson, C., additional, Johnson, D., additional, Joy, A., additional, Kay, M., additional, Kershaw, J. K., additional, Kibukawa, M., additional, Kimberley, A. M., additional, King, A., additional, Knights, A. J., additional, Lad, H., additional, Laird, G., additional, Langford, C. F., additional, Lawlor, S., additional, Leongamornlert, D. A., additional, Lloyd, D. M., additional, Loveland, J., additional, Lovell, J., additional, Lush, M. J., additional, Lyne, R., additional, Martin, S., additional, Mashreghi-Mohammadi, M., additional, Matthews, L., additional, Matthews, N. S. W., additional, McLaren, S., additional, Milne, S., additional, Mistry, S., additional, oore, M. J. F. M., additional, Nickerson, T., additional, O'Dell, C. N., additional, Oliver, K., additional, Palmeiri, A., additional, Palmer, S. A., additional, Pandian, R. D., additional, Parker, A., additional, Patel, D., additional, Pearce, A. V., additional, Peck, A. I., additional, Pelan, S., additional, Phelps, K., additional, Phillimore, B. J., additional, Plumb, R., additional, Porter, K. M., additional, Prigmore, E., additional, Rajan, J., additional, Raymond, C., additional, Rouse, G., additional, Saenphimmachak, C., additional, Sehra, H. K., additional, Sheridan, E., additional, Shownkeen, R., additional, Sims, S., additional, Skuce, C. D., additional, Smith, M., additional, Steward, C., additional, Subramanian, S., additional, Sycamore, N., additional, Tracey, A., additional, Tromans, A., additional, Van Helmond, Z., additional, Wall J. M. Wallis, M., additional, White, S., additional, Whitehead, S. L., additional, Wilkinson, J. E., additional, Willey, D. L., additional, Williams, H., additional, Wilming, L., additional, Wray, P. W., additional, Wu, Z., additional, Coulson, A., additional, Vaudin, M., additional, Sulston, J. E., additional, Durbin, R., additional, Hubbard, T., additional, Wooster, R., additional, Dunham, I., additional, Carter, N. P., additional, McVean, G., additional, Ross, M. T., additional, Harrow, J., additional, Olson, M. V., additional, Beck, S., additional, Rogers, J., additional, and Bentley, D. R., additional
- Published
- 2006
- Full Text
- View/download PDF
18. Ultra-high resolution array painting facilitates breakpoint sequencing
- Author
-
Gribble, S M, primary, Kalaitzopoulos, D, additional, Burford, D C, additional, Prigmore, E, additional, Selzer, R R, additional, Ng, B L, additional, Matthews, N S W, additional, Porter, K M, additional, Curley, R, additional, Lindsay, S J, additional, Baptista, J, additional, Richmond, T A, additional, and Carter, N P, additional
- Published
- 2006
- Full Text
- View/download PDF
19. Ultra-high resolution array painting facilitates breakpoint sequencing.
- Author
-
Gribbie, S. M., Kalaitzopoulos, D., Burford, D. C., Prigmore, E., Selzer, R. R., Ng, B. L., Matthews, N. S. W., Porter, K. M., Curley, R., Lindsay, S. J., Baptista, J., Richmond, T. A., and Carter, N. P.
- Subjects
HUMAN gene mapping ,CELL physiology ,GENETIC polymorphisms ,CLINICAL trials ,GENE mapping ,GENOMICS - Abstract
Objective: To describe a considerably advanced method of array painting, which allows the rapid, ultra-high resolution mapping of translocation breakpoints such that rearrangement junction fragments can be amplified directly and sequenced. Method: Ultra-high resolution array painting involves the hybridisation of probes generated by the amplification of small numbers of flow-sorted derivative chromosomes to oligonucleotide arrays designed to tile breakpoint regions at extremely high resolution. Results and discussion: How ultra-high resolution array painting of four balanced translocation cases rapidly and efficiently maps breakpoints to a point where junction fragments can be amplified easily and sequenced is demonstrated. With this new development, breakpoints can be mapped using just two array experiments: the first using whole-genome array painting to tiling resolution large insert clone arrays, the second using ultra-high-resolution oligonucleotide arrays targeted to the breakpoint regions. In this way, breakpoints can be mapped and then sequenced in a few weeks. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
20. Cryptic Rac-binding and p21(Cdc42Hs/Rac)-activated kinase phosphorylation sites of NADPH oxidase component p67(phox).
- Author
-
Ahmed, S, Prigmore, E, Govind, S, Veryard, C, Kozma, R, Wientjes, F B, Segal, A W, and Lim, L
- Abstract
Rac1 is a member of the Rho family of small molecular mass GTPases that act as molecular switches to control actin-based cell morphology as well as cell growth and differentiation. Rac1 and Rac2 are specifically required for superoxide formation by components of the NADPH oxidase. In binding assays, Rac1 interacts directly with p67(phox), but not with the other oxidase components: cytochrome b, p40(phox), or p47(phox) (Prigmore, E., Ahmed, S., Best, A., Kozma, R. , Manser, E., Segal, A. W., and Lim, L. (1995) J. Biol. Chem. 270, 10717-10722). Here, the Rac1/2 interaction with p67(phox) has been characterized further. Rac1 and Rac2 can bind to p67(phox) amino acid residues 170-199, and the N terminus (amino acids 1-192) of p67(phox) can be used as a specific inhibitor of Rac signaling. Deletion of p67(phox) C-terminal sequences (amino acids 193-526), the C-terminal SH3 domain (amino acids 470-526), or the polyproline-rich motif (amino acids 226-236) stimulates Rac1 binding by approximately 8-fold. p21(Cdc42Hs/Rac)-activated kinase (PAK) phosphorylates p67(phox) amino acid residues adjacent to the Rac1/2-binding site, and this phosphorylation is stimulated by deletion of the C-terminal SH3 domain or the polyproline-rich motif. These data suggest a role for cryptic Rac-binding and PAK phosphorylation sites of p67(phox) in control of the NADPH oxidase.
- Published
- 1998
21. A 68-kDa kinase and NADPH oxidase component p67phox are targets for Cdc42Hs and Rac1 in neutrophils.
- Author
-
Prigmore, E, Ahmed, S, Best, A, Kozma, R, Manser, E, Segal, A W, and Lim, L
- Abstract
Cdc42Hs and Rac1 are members of the Ras superfamily of small molecular weight (p21) GTP binding proteins. Cdc42Hs induces filopodia formation in Swiss 3T3 fibroblasts while Rac1 induces membrane ruffling. Rac1 also activates superoxide production by the components (cytochrome b, p40phox, p67phox, and p47phox) of the neutrophil oxidase. To isolate target proteins involved in these signaling pathways, we have probed proteins from neutrophil cytosol immobilized on nitrocellulose with Cdc42Hs labeled with [gamma-32P]GTP. Cdc42Hs probe detected binding protein(s) of 66-68 kDa in neutrophil cytosol. Rac1 probe also detected the 66-68-kDa proteins, suggesting the possibility that p67phox may be a binding protein for both of these p21 proteins. Indeed, Cdc42Hs and Rac1 were found to bind specifically to purified recombinant p67phox but not the other oxidase components. A 68-kDa Cdc42Hs binding protein was purified from neutrophil cytosol and found to be related to the recently described p65pak kinase from brain. These results suggest that the p68 kinase and p67phox are targets for Cdc42Hs and Rac1 in neutrophils.
- Published
- 1995
22. The complex nature of constitutional de novo apparently balanced translocations in patients presenting with abnormal phenotypes
- Author
-
Gribble, S. M., Prigmore, E., Burford, D. C., Porter, K. M., Nq, B. L., Douglas, E. J., Fiegler, H. C., Carr, P., Kalaitzopoulos, D., Clegg, S., Sandstrom, R., Temple, I. K., Youings, S. I., Thomas, N. S., Dennis, N. R., Jacobs, P. A., Crolla, J. A., Carter, N. P., Gribble, S. M., Prigmore, E., Burford, D. C., Porter, K. M., Nq, B. L., Douglas, E. J., Fiegler, H. C., Carr, P., Kalaitzopoulos, D., Clegg, S., Sandstrom, R., Temple, I. K., Youings, S. I., Thomas, N. S., Dennis, N. R., Jacobs, P. A., Crolla, J. A., and Carter, N. P.
- Abstract
OBJECTIVE: To describe the systematic analysis of constitutional de novo apparently balanced translocations in patients presenting with abnormal phenotypes, characterise the structural chromosome rearrangements, map the translocation breakpoints, and report detectable genomic imbalances. METHODS: DNA microarrays were used with a resolution of 1 Mb for the detailed genome-wide analysis of the patients. Array CGH was used to screen for genomic imbalance and array painting to map chromosome breakpoints rapidly. These two methods facilitate rapid analysis of translocation breakpoints and screening for cryptic chromosome imbalance. Breakpoints of rearrangements were further refined (to the level of spanning clones) using fluorescence in situ hybridisation where appropriate. RESULTS: Unexpected additional complexity or genome imbalance was found in six of 10 patients studied. The patients could be grouped according to the general nature of the karyotype rearrangement as follows: (A) three cases with complex multiple rearrangements including deletions, inversions, and insertions at or near one or both breakpoints; (B) three cases in which, while the translocations appeared to be balanced, microarray analysis identified previously unrecognised imbalance on chromosomes unrelated to the translocation; (C) four cases in which the translocation breakpoints appeared simple and balanced at the resolution used. CONCLUSIONS: This high level of unexpected rearrangement complexity, if generally confirmed in the study of further patients, will have an impact on current diagnostic investigations of this type and provides an argument for the more widespread adoption of microarray analysis or other high resolution genome-wide screens for chromosome imbalance and rearrangement.
23. Mapping of an Renal cell carcinoma (RCC) Associated (3;6) Translocation and Identification of RCC Candidate Genes.
- Author
-
Adulrahman, Mahera, Foster, R. E., Morris, M. R., Prigmore, E., Gribble, S., Gentle, D., Chu, C., Weston, P. M. T., Davison, V., Carter, N., Latif, F., and Maher, E. R.
- Subjects
RENAL cell carcinoma - Abstract
Presents an abstract of the article "Mapping of an Renal Cell Carcinoma (RCC) Associated (3;6) Translocation and Identification of RCC Candidate Genes," by Mahera Adulrahman, R. E. Foster, M. R. Morris, E. Prigmore, S. Gribble, D. Gentle, C. Chu, P. M. T. Weston, V. Davison, N. Carter, F. Latif., and E. R. Maher.
- Published
- 2005
24. Update on Detection of Submicroscopic Chromosomal Imbalances in Patients With Learning Disability and Dysmorphic Features by Array-based Comparative Genomic Hybridization (Array-CGH) at 1 Mb Resolution.
- Author
-
Shaw-Smith, Charles, Rickman, L., Gribble, S., Willatt, L., Prigmore, E., Porter, K., Curley, R., Whittaker, J., Dunn, C., Firth, H., Wilson, L., Clayton-Smith, J., Temple, K., Fryer, A., and Carter, N.
- Subjects
LEARNING disabilities - Abstract
Presents an abstract of the article "Update on Detection of Submicroscopic Chromosomal Imbalances in Patients With Learning Disability and Dysmorphic Features by Array-based Comparative Genomic Hybridization (Array-CGH) at 1 Mb Resolution," by Charles Shaw-Smith, L. Rickman, S. Gribble, L. Willatt, E. Prigmore, K. Porter, R. Curley, j. Whittaker, C. Dunn, H. Firth, L. Wilson, J. Clayton-Smith, K. Temple, A. Fryer, and N. Carter.
- Published
- 2005
25. Inherited Duplications/Deletions Detected by Array CGH - Chromosomal Variants or Clinically Significant?
- Author
-
Willatt, Lionel, Kerr, E., Carter, N. P., Cumming, S., Gribble, S., Prigmore, E., Rickman, L., Simon, I., Whittaker, J., Lunt, P., Deshpande, C., Wilson, L., and Shaw-Smith, C.
- Subjects
COMPARATIVE genomic hybridization - Abstract
Presents an abstract of the article "Inherited Duplications/Deletions Detected by Array CGH - Chromosomal Variants or Clinically Significant?" by Lionel Willatt, E. Kerr, N.P. Carter, S. Cumming, S. Gribble, E. Prigmore, L. Rickman, I. Simon, J. Whittaker, P. Lunt, C. Deshpande, l. Wilson, and C. Shaw-Smith.
- Published
- 2005
26. An integrated single-cell reference atlas of the human endometrium.
- Author
-
Marečková M, Garcia-Alonso L, Moullet M, Lorenzi V, Petryszak R, Sancho-Serra C, Oszlanczi A, Icoresi Mazzeo C, Wong FCK, Kelava I, Hoffman S, Krassowski M, Garbutt K, Gaitskell K, Yancheva S, Woon EV, Male V, Granne I, Hellner K, Mahbubani KT, Saeb-Parsy K, Lotfollahi M, Prigmore E, Southcombe J, Dragovic RA, Becker CM, Zondervan KT, and Vento-Tormo R
- Subjects
- Humans, Female, Transcriptome, Stromal Cells metabolism, Epithelial Cells metabolism, Genome-Wide Association Study, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta genetics, Gene Expression Profiling methods, Signal Transduction genetics, Fibroblasts metabolism, Endometrium metabolism, Endometrium cytology, Single-Cell Analysis methods, Endometriosis genetics, Endometriosis pathology, Endometriosis metabolism
- Abstract
The complex and dynamic cellular composition of the human endometrium remains poorly understood. Previous endometrial single-cell atlases profiled few donors and lacked consensus in defining cell types. We introduce the Human Endometrial Cell Atlas (HECA), a high-resolution single-cell reference atlas (313,527 cells) combining published and new endometrial single-cell transcriptomics datasets of 63 women with and without endometriosis. HECA assigns consensus and identifies previously unreported cell types, mapped in situ using spatial transcriptomics and validated using a new independent single-nuclei dataset (312,246 nuclei, 63 donors). In the functionalis, we identify intricate stromal-epithelial cell coordination via transforming growth factor beta (TGFβ) signaling. In the basalis, we define signaling between fibroblasts and an epithelial population expressing progenitor markers. Integration of HECA with large-scale endometriosis genome-wide association study data pinpoints decidualized stromal cells and macrophages as most likely dysregulated in endometriosis. The HECA is a valuable resource for studying endometrial physiology and disorders, and for guiding microphysiological in vitro systems development., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
27. Author Correction: Human SARS-CoV-2 challenge uncovers local and systemic response dynamics.
- Author
-
Lindeboom RGH, Worlock KB, Dratva LM, Yoshida M, Scobie D, Wagstaffe HR, Richardson L, Wilbrey-Clark A, Barnes JL, Kretschmer L, Polanski K, Allen-Hyttinen J, Mehta P, Sumanaweera D, Boccacino JM, Sungnak W, Elmentaite R, Huang N, Mamanova L, Kapuge R, Bolt L, Prigmore E, Killingley B, Kalinova M, Mayer M, Boyers A, Mann A, Swadling L, Woodall MNJ, Ellis S, Smith CM, Teixeira VH, Janes SM, Chambers RC, Haniffa M, Catchpole A, Heyderman R, Noursadeghi M, Chain B, Mayer A, Meyer KB, Chiu C, Nikolić MZ, and Teichmann SA
- Published
- 2024
- Full Text
- View/download PDF
28. Human SARS-CoV-2 challenge uncovers local and systemic response dynamics.
- Author
-
Lindeboom RGH, Worlock KB, Dratva LM, Yoshida M, Scobie D, Wagstaffe HR, Richardson L, Wilbrey-Clark A, Barnes JL, Kretschmer L, Polanski K, Allen-Hyttinen J, Mehta P, Sumanaweera D, Boccacino JM, Sungnak W, Elmentaite R, Huang N, Mamanova L, Kapuge R, Bolt L, Prigmore E, Killingley B, Kalinova M, Mayer M, Boyers A, Mann A, Swadling L, Woodall MNJ, Ellis S, Smith CM, Teixeira VH, Janes SM, Chambers RC, Haniffa M, Catchpole A, Heyderman R, Noursadeghi M, Chain B, Mayer A, Meyer KB, Chiu C, Nikolić MZ, and Teichmann SA
- Subjects
- Female, Humans, Male, Epithelial Cells immunology, Gene Expression Profiling, Interferons immunology, Macrophages immunology, Macrophages virology, Nasopharynx virology, Nasopharynx immunology, T-Lymphocytes cytology, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes virology, Time Factors, Virus Replication, COVID-19 genetics, COVID-19 immunology, COVID-19 pathology, COVID-19 virology, Multiomics, SARS-CoV-2 growth & development, SARS-CoV-2 immunology, SARS-CoV-2 pathogenicity, SARS-CoV-2 physiology, Single-Cell Analysis
- Abstract
The COVID-19 pandemic is an ongoing global health threat, yet our understanding of the dynamics of early cellular responses to this disease remains limited
1 . Here in our SARS-CoV-2 human challenge study, we used single-cell multi-omics profiling of nasopharyngeal swabs and blood to temporally resolve abortive, transient and sustained infections in seronegative individuals challenged with pre-Alpha SARS-CoV-2. Our analyses revealed rapid changes in cell-type proportions and dozens of highly dynamic cellular response states in epithelial and immune cells associated with specific time points and infection status. We observed that the interferon response in blood preceded the nasopharyngeal response. Moreover, nasopharyngeal immune infiltration occurred early in samples from individuals with only transient infection and later in samples from individuals with sustained infection. High expression of HLA-DQA2 before inoculation was associated with preventing sustained infection. Ciliated cells showed multiple immune responses and were most permissive for viral replication, whereas nasopharyngeal T cells and macrophages were infected non-productively. We resolved 54 T cell states, including acutely activated T cells that clonally expanded while carrying convergent SARS-CoV-2 motifs. Our new computational pipeline Cell2TCR identifies activated antigen-responding T cells based on a gene expression signature and clusters these into clonotype groups and motifs. Overall, our detailed time series data can serve as a Rosetta stone for epithelial and immune cell responses and reveals early dynamic responses associated with protection against infection., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
29. NUDCD3 deficiency disrupts V(D)J recombination to cause SCID and Omenn syndrome.
- Author
-
Chen R, Lukianova E, van der Loeff IS, Spegarova JS, Willet JDP, James KD, Ryder EJ, Griffin H, IJspeert H, Gajbhiye A, Lamoliatte F, Marin-Rubio JL, Woodbine L, Lemos H, Swan DJ, Pintar V, Sayes K, Ruiz-Morales ER, Eastham S, Dixon D, Prete M, Prigmore E, Jeggo P, Boyes J, Mellor A, Huang L, van der Burg M, Engelhardt KR, Stray-Pedersen A, Erichsen HC, Gennery AR, Trost M, Adams DJ, Anderson G, Lorenc A, Trynka G, and Hambleton S
- Subjects
- Humans, Animals, Mice, Male, Female, Infant, B-Lymphocytes immunology, Homeodomain Proteins genetics, Homeodomain Proteins immunology, T-Lymphocytes immunology, Child, Preschool, Mutation, Missense, Severe Combined Immunodeficiency genetics, Severe Combined Immunodeficiency immunology, V(D)J Recombination immunology, V(D)J Recombination genetics
- Abstract
Inborn errors of T cell development present a pediatric emergency in which timely curative therapy is informed by molecular diagnosis. In 11 affected patients across four consanguineous kindreds, we detected homozygosity for a single deleterious missense variant in the gene NudC domain-containing 3 ( NUDCD3 ) . Two infants had severe combined immunodeficiency with the complete absence of T and B cells (T
- B- SCID), whereas nine showed classical features of Omenn syndrome (OS). Restricted antigen receptor gene usage by residual T lymphocytes suggested impaired V(D)J recombination. Patient cells showed reduced expression of NUDCD3 protein and diminished ability to support RAG-mediated recombination in vitro, which was associated with pathologic sequestration of RAG1 in the nucleoli. Although impaired V(D)J recombination in a mouse model bearing the homologous variant led to milder immunologic abnormalities, NUDCD3 is absolutely required for healthy T and B cell development in humans.- Published
- 2024
- Full Text
- View/download PDF
30. Human skeletal muscle aging atlas.
- Author
-
Kedlian VR, Wang Y, Liu T, Chen X, Bolt L, Tudor C, Shen Z, Fasouli ES, Prigmore E, Kleshchevnikov V, Pett JP, Li T, Lawrence JEG, Perera S, Prete M, Huang N, Guo Q, Zeng X, Yang L, Polański K, Chipampe NJ, Dabrowska M, Li X, Bayraktar OA, Patel M, Kumasaka N, Mahbubani KT, Xiang AP, Meyer KB, Saeb-Parsy K, Teichmann SA, and Zhang H
- Subjects
- Humans, Animals, Mice, Adult, Aged, Sarcopenia pathology, Sarcopenia metabolism, Male, Neuromuscular Junction metabolism, Middle Aged, Female, Aging physiology, Muscle, Skeletal metabolism, Muscle, Skeletal physiology
- Abstract
Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
31. Transcriptional signals of transformation in human cancer.
- Author
-
Kildisiute G, Kalyva M, Elmentaite R, van Dongen S, Thevanesan C, Piapi A, Ambridge K, Prigmore E, Haniffa M, Teichmann SA, Straathof K, Cortés-Ciriano I, Behjati S, and Young MD
- Subjects
- Adult, Humans, Embryonic Development, Fetus, Gene Expression Profiling, Transcriptome, Liver Neoplasms genetics
- Abstract
Background: As normal cells transform into cancers, their cell state changes, which may drive cancer cells into a stem-like or more primordial, foetal, or embryonic cell state. The transcriptomic profile of this final state may encode information about cancer's origin and how cancers relate to their normal cell counterparts., Methods: Here, we used single-cell atlases to study cancer transformation in transcriptional terms. We utilised bulk transcriptomes across a wide spectrum of adult and childhood cancers, using a previously established method to interrogate their relationship to normal cell states. We extend and validate these findings using single-cell cancer transcriptomes and organ-specific atlases of colorectal and liver cancer., Results: Our bulk transcriptomic data reveals that adult cancers rarely return to an embryonic state, but that a foetal state is a near-universal feature of childhood cancers. This finding was confirmed with single-cell cancer transcriptomes., Conclusions: Our findings provide a nuanced picture of transformation in human cancer, indicating cancer-specific rather than universal patterns of transformation pervade adult epithelial cancers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
32. Single-cell multi-omics analysis of COVID-19 patients with pre-existing autoimmune diseases shows aberrant immune responses to infection.
- Author
-
Barmada A, Handfield LF, Godoy-Tena G, de la Calle-Fabregat C, Ciudad L, Arutyunyan A, Andrés-León E, Hoo R, Porter T, Oszlanczi A, Richardson L, Calero-Nieto FJ, Wilson NK, Marchese D, Sancho-Serra C, Carrillo J, Presas-Rodríguez S, Ramo-Tello C, Ruiz-Sanmartin A, Ferrer R, Ruiz-Rodriguez JC, Martínez-Gallo M, Munera-Campos M, Carrascosa JM, Göttgens B, Heyn H, Prigmore E, Casafont-Solé I, Solanich X, Sánchez-Cerrillo I, González-Álvaro I, Raimondo MG, Ramming A, Martin J, Martínez-Cáceres E, Ballestar E, Vento-Tormo R, and Rodríguez-Ubreva J
- Subjects
- Humans, SARS-CoV-2, Leukocytes, Mononuclear, Multiomics, Autoimmunity, Single-Cell Analysis, COVID-19, Autoimmune Diseases
- Abstract
In COVID-19, hyperinflammatory and dysregulated immune responses contribute to severity. Patients with pre-existing autoimmune conditions can therefore be at increased risk of severe COVID-19 and/or associated sequelae, yet SARS-CoV-2 infection in this group has been little studied. Here, we performed single-cell analysis of peripheral blood mononuclear cells from patients with three major autoimmune diseases (rheumatoid arthritis, psoriasis, or multiple sclerosis) during SARS-CoV-2 infection. We observed compositional differences between the autoimmune disease groups coupled with altered patterns of gene expression, transcription factor activity, and cell-cell communication that substantially shape the immune response under SARS-CoV-2 infection. While enrichment of HLA-DRlow CD14+ monocytes was observed in all three autoimmune disease groups, type-I interferon signaling as well as inflammatory T cell and monocyte responses varied widely between the three groups of patients. Our results reveal disturbed immune responses to SARS-CoV-2 in patients with pre-existing autoimmunity, highlighting important considerations for disease treatment and follow-up., (© 2023 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
33. A spatial human thymus cell atlas mapped to a continuous tissue axis.
- Author
-
Yayon N, Kedlian VR, Boehme L, Suo C, Wachter B, Beuschel RT, Amsalem O, Polanski K, Koplev S, Tuck E, Dann E, Van Hulle J, Perera S, Putteman T, Predeus AV, Dabrowska M, Richardson L, Tudor C, Kreins AY, Engelbert J, Stephenson E, Kleshchevnikov V, De Rita F, Crossland D, Bosticardo M, Pala F, Prigmore E, Chipampe NJ, Prete M, Fei L, To K, Barker RA, He X, Van Nieuwerburgh F, Bayraktar O, Patel M, Davies GE, Haniffa MA, Uhlmann V, Notarangelo LD, Germain RN, Radtke AJ, Marioni JC, Taghon T, and Teichmann SA
- Abstract
T cells develop from circulating precursors, which enter the thymus and migrate throughout specialised sub-compartments to support maturation and selection. This process starts already in early fetal development and is highly active until the involution of the thymus in adolescence. To map the micro-anatomical underpinnings of this process in pre- vs. post-natal states, we undertook a spatially resolved analysis and established a new quantitative morphological framework for the thymus, the Cortico-Medullary Axis. Using this axis in conjunction with the curation of a multimodal single-cell, spatial transcriptomics and high-resolution multiplex imaging atlas, we show that canonical thymocyte trajectories and thymic epithelial cells are highly organised and fully established by post-conception week 12, pinpoint TEC progenitor states, find that TEC subsets and peripheral tissue genes are associated with Hassall's Corpuscles and uncover divergence in the pace and drivers of medullary entry between CD4 vs. CD8 T cell lineages. These findings are complemented with a holistic toolkit for spatial analysis and annotation, providing a basis for a detailed understanding of T lymphocyte development., Competing Interests: Conflict of interest J.C.M has been an employee of Genentech, Inc. since September 2022. In the past three years, S.A.T. has consulted for or been a member of scientific advisory boards at Qiagen, Sanofi, GlaxoSmithKline, and ForeSite Labs. She is a consultant and equity holder for TransitionBio and EnsoCell. The remaining authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
34. Spatial multiomics map of trophoblast development in early pregnancy.
- Author
-
Arutyunyan A, Roberts K, Troulé K, Wong FCK, Sheridan MA, Kats I, Garcia-Alonso L, Velten B, Hoo R, Ruiz-Morales ER, Sancho-Serra C, Shilts J, Handfield LF, Marconato L, Tuck E, Gardner L, Mazzeo CI, Li Q, Kelava I, Wright GJ, Prigmore E, Teichmann SA, Bayraktar OA, Moffett A, Stegle O, Turco MY, and Vento-Tormo R
- Subjects
- Female, Humans, Pregnancy, Cell Movement, Placenta blood supply, Placenta cytology, Placenta physiology, Decidua blood supply, Decidua cytology, Maternal-Fetal Relations physiology, Single-Cell Analysis, Myometrium cytology, Myometrium physiology, Cell Differentiation, Organoids cytology, Organoids physiology, Stem Cells cytology, Transcriptome, Transcription Factors metabolism, Cell Communication, Multiomics, Pregnancy Trimester, First physiology, Trophoblasts cytology, Trophoblasts metabolism, Trophoblasts physiology
- Abstract
The relationship between the human placenta-the extraembryonic organ made by the fetus, and the decidua-the mucosal layer of the uterus, is essential to nurture and protect the fetus during pregnancy. Extravillous trophoblast cells (EVTs) derived from placental villi infiltrate the decidua, transforming the maternal arteries into high-conductance vessels
1 . Defects in trophoblast invasion and arterial transformation established during early pregnancy underlie common pregnancy disorders such as pre-eclampsia2 . Here we have generated a spatially resolved multiomics single-cell atlas of the entire human maternal-fetal interface including the myometrium, which enables us to resolve the full trajectory of trophoblast differentiation. We have used this cellular map to infer the possible transcription factors mediating EVT invasion and show that they are preserved in in vitro models of EVT differentiation from primary trophoblast organoids3,4 and trophoblast stem cells5 . We define the transcriptomes of the final cell states of trophoblast invasion: placental bed giant cells (fused multinucleated EVTs) and endovascular EVTs (which form plugs inside the maternal arteries). We predict the cell-cell communication events contributing to trophoblast invasion and placental bed giant cell formation, and model the dual role of interstitial EVTs and endovascular EVTs in mediating arterial transformation during early pregnancy. Together, our data provide a comprehensive analysis of postimplantation trophoblast differentiation that can be used to inform the design of experimental models of the human placenta in early pregnancy., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
35. Author Correction: Mapping the temporal and spatial dynamics of the human endometrium in vivo and in vitro.
- Author
-
Garcia-Alonso L, Handfield LF, Roberts K, Nikolakopoulou K, Fernando RC, Gardner L, Woodhams B, Arutyunyan A, Polanski K, Hoo R, Sancho-Serra C, Li T, Kwakwa K, Tuck E, Lorenzi V, Massalha H, Prete M, Kleshchevnikov V, Tarkowska A, Porter T, Mazzeo CI, van Dongen S, Dabrowska M, Vaskivskyi V, Mahbubani KT, Park JE, Jimenez-Linan M, Campos L, Kiselev VY, Lindskog C, Ayuk P, Prigmore E, Stratton MR, Saeb-Parsy K, Moffett A, Moore L, Bayraktar OA, Teichmann SA, Turco MY, and Vento-Tormo R
- Published
- 2023
- Full Text
- View/download PDF
36. A spatially resolved atlas of the human lung characterizes a gland-associated immune niche.
- Author
-
Madissoon E, Oliver AJ, Kleshchevnikov V, Wilbrey-Clark A, Polanski K, Richoz N, Ribeiro Orsi A, Mamanova L, Bolt L, Elmentaite R, Pett JP, Huang N, Xu C, He P, Dabrowska M, Pritchard S, Tuck L, Prigmore E, Perera S, Knights A, Oszlanczi A, Hunter A, Vieira SF, Patel M, Lindeboom RGH, Campos LS, Matsuo K, Nakayama T, Yoshida M, Worlock KB, Nikolić MZ, Georgakopoulos N, Mahbubani KT, Saeb-Parsy K, Bayraktar OA, Clatworthy MR, Stegle O, Kumasaka N, Teichmann SA, and Meyer KB
- Subjects
- Humans, Epithelial Cells metabolism, B-Lymphocytes, Immunoglobulin A metabolism, Respiratory Mucosa metabolism, Lung
- Abstract
Single-cell transcriptomics has allowed unprecedented resolution of cell types/states in the human lung, but their spatial context is less well defined. To (re)define tissue architecture of lung and airways, we profiled five proximal-to-distal locations of healthy human lungs in depth using multi-omic single cell/nuclei and spatial transcriptomics (queryable at lungcellatlas.org ). Using computational data integration and analysis, we extend beyond the suspension cell paradigm and discover macro and micro-anatomical tissue compartments including previously unannotated cell types in the epithelial, vascular, stromal and nerve bundle micro-environments. We identify and implicate peribronchial fibroblasts in lung disease. Importantly, we discover and validate a survival niche for IgA plasma cells in the airway submucosal glands (SMG). We show that gland epithelial cells recruit B cells and IgA plasma cells, and promote longevity and antibody secretion locally through expression of CCL28, APRIL and IL-6. This new 'gland-associated immune niche' has implications for respiratory health., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
37. Precise identification of cancer cells from allelic imbalances in single cell transcriptomes.
- Author
-
Trinh MK, Pacyna CN, Kildisiute G, Thevanesan C, Piapi A, Ambridge K, Anderson ND, Khabirova E, Prigmore E, Straathof K, Behjati S, and Young MD
- Subjects
- Allelic Imbalance genetics, Genotype, Humans, Polymorphism, Single Nucleotide, RNA, Messenger genetics, Neoplasms diagnosis, Neoplasms genetics, Transcriptome
- Abstract
A fundamental step of tumour single cell mRNA analysis is separating cancer and non-cancer cells. We show that the common approach to separation, using shifts in average expression, can lead to erroneous biological conclusions. By contrast, allelic imbalances representing copy number changes directly detect the cancer genotype and accurately separate cancer from non-cancer cells. Our findings provide a definitive approach to identifying cancer cells from single cell mRNA sequencing data., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
38. Single-cell roadmap of human gonadal development.
- Author
-
Garcia-Alonso L, Lorenzi V, Mazzeo CI, Alves-Lopes JP, Roberts K, Sancho-Serra C, Engelbert J, Marečková M, Gruhn WH, Botting RA, Li T, Crespo B, van Dongen S, Kiselev VY, Prigmore E, Herbert M, Moffett A, Chédotal A, Bayraktar OA, Surani A, Haniffa M, and Vento-Tormo R
- Subjects
- Animals, Chromatin genetics, Chromatin metabolism, Female, Granulosa Cells cytology, Granulosa Cells metabolism, Humans, Immunoglobulins, Macrophages metabolism, Male, Membrane Glycoproteins, Membrane Proteins, Mice, Microscopy, Fluorescence, PAX8 Transcription Factor, Pregnancy, Pregnancy Trimester, First, Pregnancy Trimester, Second, Receptors, Immunologic, Transcriptome, Cell Lineage, Germ Cells cytology, Germ Cells metabolism, Ovary cytology, Ovary embryology, Sex Differentiation genetics, Single-Cell Analysis, Testis cytology, Testis embryology
- Abstract
Gonadal development is a complex process that involves sex determination followed by divergent maturation into either testes or ovaries
1 . Historically, limited tissue accessibility, a lack of reliable in vitro models and critical differences between humans and mice have hampered our knowledge of human gonadogenesis, despite its importance in gonadal conditions and infertility. Here, we generated a comprehensive map of first- and second-trimester human gonads using a combination of single-cell and spatial transcriptomics, chromatin accessibility assays and fluorescent microscopy. We extracted human-specific regulatory programmes that control the development of germline and somatic cell lineages by profiling equivalent developmental stages in mice. In both species, we define the somatic cell states present at the time of sex specification, including the bipotent early supporting population that, in males, upregulates the testis-determining factor SRY and sPAX8s, a gonadal lineage located at the gonadal-mesonephric interface. In females, we resolve the cellular and molecular events that give rise to the first and second waves of granulosa cells that compartmentalize the developing ovary to modulate germ cell differentiation. In males, we identify human SIGLEC15+ and TREM2+ fetal testicular macrophages, which signal to somatic cells outside and inside the developing testis cords, respectively. This study provides a comprehensive spatiotemporal map of human and mouse gonadal differentiation, which can guide in vitro gonadogenesis., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
39. Genetic and chemotherapeutic influences on germline hypermutation.
- Author
-
Kaplanis J, Ide B, Sanghvi R, Neville M, Danecek P, Coorens T, Prigmore E, Short P, Gallone G, McRae J, Carmichael J, Barnicoat A, Firth H, O'Brien P, Rahbari R, and Hurles M
- Subjects
- Age Factors, Humans, Male, Mutagenesis genetics, Mutation, Parents, Polymorphism, Single Nucleotide, Genetic Diseases, Inborn genetics, Germ Cells, Germ-Line Mutation genetics
- Abstract
Mutations in the germline generates all evolutionary genetic variation and is a cause of genetic disease. Parental age is the primary determinant of the number of new germline mutations in an individual's genome
1,2 . Here we analysed the genome-wide sequences of 21,879 families with rare genetic diseases and identified 12 individuals with a hypermutated genome with between two and seven times more de novo single-nucleotide variants than expected. In most families (9 out of 12), the excess mutations came from the father. Two families had genetic drivers of germline hypermutation, with fathers carrying damaging genetic variation in DNA-repair genes. For five of the families, paternal exposure to chemotherapeutic agents before conception was probably a key driver of hypermutation. Our results suggest that the germline is well protected from mutagenic effects, hypermutation is rare, the number of excess mutations is relatively modest and most individuals with a hypermutated genome will not have a genetic disease., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
40. Single-cell transcriptomics reveals a distinct developmental state of KMT2A-rearranged infant B-cell acute lymphoblastic leukemia.
- Author
-
Khabirova E, Jardine L, Coorens THH, Webb S, Treger TD, Engelbert J, Porter T, Prigmore E, Collord G, Piapi A, Teichmann SA, Inglott S, Williams O, Heidenreich O, Young MD, Straathof K, Bomken S, Bartram J, Haniffa M, and Behjati S
- Subjects
- Bone Marrow metabolism, Child, Gene Rearrangement genetics, Humans, Infant, Mutation genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Transcriptome genetics
- Abstract
KMT2A-rearranged infant ALL is an aggressive childhood leukemia with poor prognosis. Here, we investigated the developmental state of KMT2A-rearranged infant B-cell acute lymphoblastic leukemia (B-ALL) using bulk messenger RNA (mRNA) meta-analysis and examination of single lymphoblast transcriptomes against a developing bone marrow reference. KMT2A-rearranged infant B-ALL was uniquely dominated by an early lymphocyte precursor (ELP) state, whereas less adverse NUTM1-rearranged infant ALL demonstrated signals of later developing B cells, in line with most other childhood B-ALLs. We compared infant lymphoblasts with ELP cells and revealed that the cancer harbored hybrid myeloid-lymphoid features, including nonphysiological antigen combinations potentially targetable to achieve cancer specificity. We validated surface coexpression of exemplar combinations by flow cytometry. Through analysis of shared mutations in separate leukemias from a child with infant KMT2A-rearranged B-ALL relapsing as AML, we established that KMT2A rearrangement occurred in very early development, before hematopoietic specification, emphasizing that cell of origin cannot be inferred from the transcriptional state., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
41. Local and systemic responses to SARS-CoV-2 infection in children and adults.
- Author
-
Yoshida M, Worlock KB, Huang N, Lindeboom RGH, Butler CR, Kumasaka N, Dominguez Conde C, Mamanova L, Bolt L, Richardson L, Polanski K, Madissoon E, Barnes JL, Allen-Hyttinen J, Kilich E, Jones BC, de Wilton A, Wilbrey-Clark A, Sungnak W, Pett JP, Weller J, Prigmore E, Yung H, Mehta P, Saleh A, Saigal A, Chu V, Cohen JM, Cane C, Iordanidou A, Shibuya S, Reuschl AK, Herczeg IT, Argento AC, Wunderink RG, Smith SB, Poor TA, Gao CA, Dematte JE, Reynolds G, Haniffa M, Bowyer GS, Coates M, Clatworthy MR, Calero-Nieto FJ, Göttgens B, O'Callaghan C, Sebire NJ, Jolly C, De Coppi P, Smith CM, Misharin AV, Janes SM, Teichmann SA, Nikolić MZ, and Meyer KB
- Subjects
- Adult, Bronchi immunology, Bronchi virology, COVID-19 pathology, Chicago, Cohort Studies, Disease Progression, Epithelial Cells cytology, Epithelial Cells immunology, Epithelial Cells virology, Female, Humans, Immunity, Innate, London, Male, Nasal Mucosa immunology, Nasal Mucosa virology, SARS-CoV-2 growth & development, Single-Cell Analysis, Trachea virology, Young Adult, COVID-19 blood, COVID-19 immunology, Dendritic Cells immunology, Interferons immunology, Killer Cells, Natural immunology, SARS-CoV-2 immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
It is not fully understood why COVID-19 is typically milder in children
1-3 . Here, to examine the differences between children and adults in their response to SARS-CoV-2 infection, we analysed paediatric and adult patients with COVID-19 as well as healthy control individuals (total n = 93) using single-cell multi-omic profiling of matched nasal, tracheal, bronchial and blood samples. In the airways of healthy paediatric individuals, we observed cells that were already in an interferon-activated state, which after SARS-CoV-2 infection was further induced especially in airway immune cells. We postulate that higher paediatric innate interferon responses restrict viral replication and disease progression. The systemic response in children was characterized by increases in naive lymphocytes and a depletion of natural killer cells, whereas, in adults, cytotoxic T cells and interferon-stimulated subpopulations were significantly increased. We provide evidence that dendritic cells initiate interferon signalling in early infection, and identify epithelial cell states associated with COVID-19 and age. Our matching nasal and blood data show a strong interferon response in the airways with the induction of systemic interferon-stimulated populations, which were substantially reduced in paediatric patients. Together, we provide several mechanisms that explain the milder clinical syndrome observed in children., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
42. Mapping the temporal and spatial dynamics of the human endometrium in vivo and in vitro.
- Author
-
Garcia-Alonso L, Handfield LF, Roberts K, Nikolakopoulou K, Fernando RC, Gardner L, Woodhams B, Arutyunyan A, Polanski K, Hoo R, Sancho-Serra C, Li T, Kwakwa K, Tuck E, Lorenzi V, Massalha H, Prete M, Kleshchevnikov V, Tarkowska A, Porter T, Mazzeo CI, van Dongen S, Dabrowska M, Vaskivskyi V, Mahbubani KT, Park JE, Jimenez-Linan M, Campos L, Kiselev VY, Lindskog C, Ayuk P, Prigmore E, Stratton MR, Saeb-Parsy K, Moffett A, Moore L, Bayraktar OA, Teichmann SA, Turco MY, and Vento-Tormo R
- Subjects
- Cell Differentiation, Cell Lineage, Cellular Microenvironment, Endometrial Neoplasms pathology, Endometrium embryology, Endometrium pathology, Female, Gonadal Steroid Hormones metabolism, Humans, In Vitro Techniques, Organoids, Receptors, Notch metabolism, Signal Transduction, Spatio-Temporal Analysis, Tissue Culture Techniques, Transcriptome, Uterus pathology, Wnt Proteins metabolism, Endometrium physiology, Menstrual Cycle
- Abstract
The endometrium, the mucosal lining of the uterus, undergoes dynamic changes throughout the menstrual cycle in response to ovarian hormones. We have generated dense single-cell and spatial reference maps of the human uterus and three-dimensional endometrial organoid cultures. We dissect the signaling pathways that determine cell fate of the epithelial lineages in the lumenal and glandular microenvironments. Our benchmark of the endometrial organoids reveals the pathways and cell states regulating differentiation of the secretory and ciliated lineages both in vivo and in vitro. In vitro downregulation of WNT or NOTCH pathways increases the differentiation efficiency along the secretory and ciliated lineages, respectively. We utilize our cellular maps to deconvolute bulk data from endometrial cancers and endometriotic lesions, illuminating the cell types dominating in each of these disorders. These mechanistic insights provide a platform for future development of treatments for common conditions including endometriosis and endometrial carcinoma., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
43. Detecting cryptic clinically relevant structural variation in exome-sequencing data increases diagnostic yield for developmental disorders.
- Author
-
Gardner EJ, Sifrim A, Lindsay SJ, Prigmore E, Rajan D, Danecek P, Gallone G, Eberhardt RY, Martin HC, Wright CF, FitzPatrick DR, Firth HV, and Hurles ME
- Subjects
- Child, Female, Humans, Male, Methyl-CpG-Binding Protein 2 genetics, Developmental Disabilities diagnosis, Developmental Disabilities genetics, Exome Sequencing methods
- Abstract
Structural variation (SV) describes a broad class of genetic variation greater than 50 bp in size. SVs can cause a wide range of genetic diseases and are prevalent in rare developmental disorders (DDs). Individuals presenting with DDs are often referred for diagnostic testing with chromosomal microarrays (CMAs) to identify large copy-number variants (CNVs) and/or with single-gene, gene-panel, or exome sequencing (ES) to identify single-nucleotide variants, small insertions/deletions, and CNVs. However, individuals with pathogenic SVs undetectable by conventional analysis often remain undiagnosed. Consequently, we have developed the tool InDelible, which interrogates short-read sequencing data for split-read clusters characteristic of SV breakpoints. We applied InDelible to 13,438 probands with severe DDs recruited as part of the Deciphering Developmental Disorders (DDD) study and discovered 63 rare, damaging variants in genes previously associated with DDs missed by standard SNV, indel, or CNV discovery approaches. Clinical review of these 63 variants determined that about half (30/63) were plausibly pathogenic. InDelible was particularly effective at ascertaining variants between 21 and 500 bp in size and increased the total number of potentially pathogenic variants identified by DDD in this size range by 42.9%. Of particular interest were seven confirmed de novo variants in MECP2, which represent 35.0% of all de novo protein-truncating variants in MECP2 among DDD study participants. InDelible provides a framework for the discovery of pathogenic SVs that are most likely missed by standard analytical workflows and has the potential to improve the diagnostic yield of ES across a broad range of genetic diseases., Competing Interests: Declaration of interests M.E.H. is a founder of, consultant to, director of, and holds shares in Congenica Ltd and is a consultant to the AZ Centre for Genomics Research. H.V.F. is a Section Editor for genetics for UpToDate. All other authors declare no conflict of interest., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
44. Blood and immune development in human fetal bone marrow and Down syndrome.
- Author
-
Jardine L, Webb S, Goh I, Quiroga Londoño M, Reynolds G, Mather M, Olabi B, Stephenson E, Botting RA, Horsfall D, Engelbert J, Maunder D, Mende N, Murnane C, Dann E, McGrath J, King H, Kucinski I, Queen R, Carey CD, Shrubsole C, Poyner E, Acres M, Jones C, Ness T, Coulthard R, Elliott N, O'Byrne S, Haltalli MLR, Lawrence JE, Lisgo S, Balogh P, Meyer KB, Prigmore E, Ambridge K, Jain MS, Efremova M, Pickard K, Creasey T, Bacardit J, Henderson D, Coxhead J, Filby A, Hussain R, Dixon D, McDonald D, Popescu DM, Kowalczyk MS, Li B, Ashenberg O, Tabaka M, Dionne D, Tickle TL, Slyper M, Rozenblatt-Rosen O, Regev A, Behjati S, Laurenti E, Wilson NK, Roy A, Göttgens B, Roberts I, Teichmann SA, and Haniffa M
- Subjects
- B-Lymphocytes cytology, Dendritic Cells cytology, Down Syndrome metabolism, Down Syndrome pathology, Endothelial Cells pathology, Eosinophils cytology, Erythroid Cells cytology, Granulocytes cytology, Humans, Immunity, Myeloid Cells cytology, Stromal Cells cytology, Bone Marrow, Bone Marrow Cells cytology, Down Syndrome blood, Down Syndrome immunology, Fetus cytology, Hematopoiesis, Immune System cytology
- Abstract
Haematopoiesis in the bone marrow (BM) maintains blood and immune cell production throughout postnatal life. Haematopoiesis first emerges in human BM at 11-12 weeks after conception
1,2 , yet almost nothing is known about how fetal BM (FBM) evolves to meet the highly specialized needs of the fetus and newborn. Here we detail the development of FBM, including stroma, using multi-omic assessment of mRNA and multiplexed protein epitope expression. We find that the full blood and immune cell repertoire is established in FBM in a short time window of 6-7 weeks early in the second trimester. FBM promotes rapid and extensive diversification of myeloid cells, with granulocytes, eosinophils and dendritic cell subsets emerging for the first time. The substantial expansion of B lymphocytes in FBM contrasts with fetal liver at the same gestational age. Haematopoietic progenitors from fetal liver, FBM and cord blood exhibit transcriptional and functional differences that contribute to tissue-specific identity and cellular diversification. Endothelial cell types form distinct vascular structures that we show are regionally compartmentalized within FBM. Finally, we reveal selective disruption of B lymphocyte, erythroid and myeloid development owing to a cell-intrinsic differentiation bias as well as extrinsic regulation through an altered microenvironment in Down syndrome (trisomy 21)., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
45. Single cell derived mRNA signals across human kidney tumors.
- Author
-
Young MD, Mitchell TJ, Custers L, Margaritis T, Morales-Rodriguez F, Kwakwa K, Khabirova E, Kildisiute G, Oliver TRW, de Krijger RR, van den Heuvel-Eibrink MM, Comitani F, Piapi A, Bugallo-Blanco E, Thevanesan C, Burke C, Prigmore E, Ambridge K, Roberts K, Braga FAV, Coorens THH, Del Valle I, Wilbrey-Clark A, Mamanova L, Stewart GD, Gnanapragasam VJ, Rampling D, Sebire N, Coleman N, Hook L, Warren A, Haniffa M, Kool M, Pfister SM, Achermann JC, He X, Barker RA, Shlien A, Bayraktar OA, Teichmann SA, Holstege FC, Meyer KB, Drost J, Straathof K, and Behjati S
- Subjects
- Adult, Algorithms, Child, Fetus metabolism, Gene Expression Regulation, Developmental, Humans, Kidney embryology, Kidney Neoplasms embryology, Kidney Neoplasms metabolism, Models, Genetic, Signal Transduction genetics, Kidney metabolism, Kidney Neoplasms genetics, RNA, Messenger genetics, RNA-Seq methods, Single-Cell Analysis methods, Transcriptome
- Abstract
Tumor cells may share some patterns of gene expression with their cell of origin, providing clues into the differentiation state and origin of cancer. Here, we study the differentiation state and cellular origin of 1300 childhood and adult kidney tumors. Using single cell mRNA reference maps of normal tissues, we quantify reference "cellular signals" in each tumor. Quantifying global differentiation, we find that childhood tumors exhibit fetal cellular signals, replacing the presumption of "fetalness" with a quantitative measure of immaturity. By contrast, in adult cancers our assessment refutes the suggestion of dedifferentiation towards a fetal state in most cases. We find an intimate connection between developmental mesenchymal populations and childhood renal tumors. We demonstrate the diagnostic potential of our approach with a case study of a cryptic renal tumor. Our findings provide a cellular definition of human renal tumors through an approach that is broadly applicable to human cancer.
- Published
- 2021
- Full Text
- View/download PDF
46. Single-cell multi-omics analysis of the immune response in COVID-19.
- Author
-
Stephenson E, Reynolds G, Botting RA, Calero-Nieto FJ, Morgan MD, Tuong ZK, Bach K, Sungnak W, Worlock KB, Yoshida M, Kumasaka N, Kania K, Engelbert J, Olabi B, Spegarova JS, Wilson NK, Mende N, Jardine L, Gardner LCS, Goh I, Horsfall D, McGrath J, Webb S, Mather MW, Lindeboom RGH, Dann E, Huang N, Polanski K, Prigmore E, Gothe F, Scott J, Payne RP, Baker KF, Hanrath AT, Schim van der Loeff ICD, Barr AS, Sanchez-Gonzalez A, Bergamaschi L, Mescia F, Barnes JL, Kilich E, de Wilton A, Saigal A, Saleh A, Janes SM, Smith CM, Gopee N, Wilson C, Coupland P, Coxhead JM, Kiselev VY, van Dongen S, Bacardit J, King HW, Rostron AJ, Simpson AJ, Hambleton S, Laurenti E, Lyons PA, Meyer KB, Nikolić MZ, Duncan CJA, Smith KGC, Teichmann SA, Clatworthy MR, Marioni JC, Göttgens B, and Haniffa M
- Subjects
- Cross-Sectional Studies, Humans, Monocytes immunology, Receptors, Antigen, B-Cell immunology, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology, COVID-19 immunology, Proteome, SARS-CoV-2 immunology, Single-Cell Analysis methods, Transcriptome
- Abstract
Analysis of human blood immune cells provides insights into the coordinated response to viral infections such as severe acute respiratory syndrome coronavirus 2, which causes coronavirus disease 2019 (COVID-19). We performed single-cell transcriptome, surface proteome and T and B lymphocyte antigen receptor analyses of over 780,000 peripheral blood mononuclear cells from a cross-sectional cohort of 130 patients with varying severities of COVID-19. We identified expansion of nonclassical monocytes expressing complement transcripts (CD16
+ C1QA/B/C+ ) that sequester platelets and were predicted to replenish the alveolar macrophage pool in COVID-19. Early, uncommitted CD34+ hematopoietic stem/progenitor cells were primed toward megakaryopoiesis, accompanied by expanded megakaryocyte-committed progenitors and increased platelet activation. Clonally expanded CD8+ T cells and an increased ratio of CD8+ effector T cells to effector memory T cells characterized severe disease, while circulating follicular helper T cells accompanied mild disease. We observed a relative loss of IgA2 in symptomatic disease despite an overall expansion of plasmablasts and plasma cells. Our study highlights the coordinated immune response that contributes to COVID-19 pathogenesis and reveals discrete cellular components that can be targeted for therapy.- Published
- 2021
- Full Text
- View/download PDF
47. Contribution of retrotransposition to developmental disorders.
- Author
-
Gardner EJ, Prigmore E, Gallone G, Danecek P, Samocha KE, Handsaker J, Gerety SS, Ironfield H, Short PJ, Sifrim A, Singh T, Chandler KE, Clement E, Lachlan KL, Prescott K, Rosser E, FitzPatrick DR, Firth HV, and Hurles ME
- Subjects
- Humans, Mutation Rate, Retroelements genetics, Developmental Disabilities genetics, Genetic Variation, Retroelements physiology
- Abstract
Mobile genetic Elements (MEs) are segments of DNA which can copy themselves and other transcribed sequences through the process of retrotransposition (RT). In humans several disorders have been attributed to RT, but the role of RT in severe developmental disorders (DD) has not yet been explored. Here we identify RT-derived events in 9738 exome sequenced trios with DD-affected probands. We ascertain 9 de novo MEs, 4 of which are likely causative of the patient's symptoms (0.04%), as well as 2 de novo gene retroduplications. Beyond identifying likely diagnostic RT events, we estimate genome-wide germline ME mutation rate and selective constraint and demonstrate that coding RT events have signatures of purifying selection equivalent to those of truncating mutations. Overall, our analysis represents a comprehensive interrogation of the impact of retrotransposition on protein coding genes and a framework for future evolutionary and disease studies.
- Published
- 2019
- Full Text
- View/download PDF
48. Exome-wide assessment of the functional impact and pathogenicity of multinucleotide mutations.
- Author
-
Kaplanis J, Akawi N, Gallone G, McRae JF, Prigmore E, Wright CF, Fitzpatrick DR, Firth HV, Barrett JC, and Hurles ME
- Subjects
- Child, DNA Mutational Analysis, Humans, Mutation Rate, Mutation, Missense, Nucleotides, Polymorphism, Single Nucleotide, Developmental Disabilities genetics, Exome, Mutation
- Abstract
Approximately 2% of de novo single-nucleotide variants (SNVs) appear as part of clustered mutations that create multinucleotide variants (MNVs). MNVs are an important source of genomic variability as they are more likely to alter an encoded protein than a SNV, which has important implications in disease as well as evolution. Previous studies of MNVs have focused on their mutational origins and have not systematically evaluated their functional impact and contribution to disease. We identified 69,940 MNVs and 91 de novo MNVs in 6688 exome-sequenced parent-offspring trios from the Deciphering Developmental Disorders Study comprising families with severe developmental disorders. We replicated the previously described MNV mutational signatures associated with DNA polymerase zeta, an error-prone translesion polymerase, and the APOBEC family of DNA deaminases. We estimate the simultaneous MNV germline mutation rate to be 1.78 × 10
-10 mutations per base pair per generation. We found that most MNVs within a single codon create a missense change that could not have been created by a SNV. MNV-induced missense changes were, on average, more physicochemically divergent, were more depleted in highly constrained genes (pLI ≥ 0.9), and were under stronger purifying selection compared with SNV-induced missense changes. We found that de novo MNVs were significantly enriched in genes previously associated with developmental disorders in affected children. This shows that MNVs can be more damaging than SNVs even when both induce missense changes, and are an important variant type to consider in relation to human disease., (© 2019 Kaplanis et al.; Published by Cold Spring Harbor Laboratory Press.)- Published
- 2019
- Full Text
- View/download PDF
49. Molecular autopsy by trio exome sequencing (ES) and postmortem examination in fetuses and neonates with prenatally identified structural anomalies.
- Author
-
Quinlan-Jones E, Lord J, Williams D, Hamilton S, Marton T, Eberhardt RY, Rinck G, Prigmore E, Keelagher R, McMullan DJ, Maher ER, Hurles ME, and Kilby MD
- Subjects
- Autopsy methods, Cohort Studies, Congenital Abnormalities diagnosis, Exome genetics, Female, Fetal Diseases diagnosis, Fetus diagnostic imaging, Humans, Infant, Newborn, Male, Pregnancy, Exome Sequencing methods, Congenital Abnormalities genetics, Fetal Diseases genetics, Prenatal Diagnosis methods
- Abstract
Purpose: To determine the diagnostic yield of combined exome sequencing (ES) and autopsy in fetuses/neonates with prenatally identified structural anomalies resulting in termination of pregnancy, intrauterine, neonatal, or early infant death., Methods: ES was undertaken in 27 proband/parent trios following full autopsy. Candidate pathogenic variants were classified by a multidisciplinary clinical review panel using American College of Medical Genetics and Genomics (ACMG) guidelines., Results: A genetic diagnosis was established in ten cases (37%). Pathogenic/likely pathogenic variants were identified in nine different genes including four de novo autosomal dominant, three homozygous autosomal recessive, two compound heterozygous autosomal recessive, and one X-linked. KMT2D variants (associated with Kabuki syndrome postnatally) occurred in two cases. Pathogenic variants were identified in 5/13 (38%) cases with multisystem anomalies, in 2/4 (50%) cases with fetal akinesia deformation sequence, and in 1/4 (25%) cases each with cardiac and brain anomalies and hydrops fetalis. No pathogenic variants were detected in fetuses with genitourinary (1), skeletal (1), or abdominal (1) abnormalities., Conclusion: This cohort demonstrates the clinical utility of molecular autopsy with ES to identify an underlying genetic cause in structurally abnormal fetuses/neonates. These molecular findings provided parents with an explanation of the developmental abnormality, delineated the recurrence risks, and assisted the management of subsequent pregnancies.
- Published
- 2019
- Full Text
- View/download PDF
50. Prenatal exome sequencing analysis in fetal structural anomalies detected by ultrasonography (PAGE): a cohort study.
- Author
-
Lord J, McMullan DJ, Eberhardt RY, Rinck G, Hamilton SJ, Quinlan-Jones E, Prigmore E, Keelagher R, Best SK, Carey GK, Mellis R, Robart S, Berry IR, Chandler KE, Cilliers D, Cresswell L, Edwards SL, Gardiner C, Henderson A, Holden ST, Homfray T, Lester T, Lewis RA, Newbury-Ecob R, Prescott K, Quarrell OW, Ramsden SC, Roberts E, Tapon D, Tooley MJ, Vasudevan PC, Weber AP, Wellesley DG, Westwood P, White H, Parker M, Williams D, Jenkins L, Scott RH, Kilby MD, Chitty LS, Hurles ME, and Maher ER
- Subjects
- Abnormal Karyotype embryology, Abortion, Eugenic statistics & numerical data, Abortion, Spontaneous epidemiology, Congenital Abnormalities diagnosis, Congenital Abnormalities epidemiology, DNA Copy Number Variations genetics, Female, Fetus diagnostic imaging, Humans, Infant, Newborn, Live Birth epidemiology, Male, Nuchal Translucency Measurement, Parents, Perinatal Death etiology, Pregnancy, Prospective Studies, Stillbirth epidemiology, Exome Sequencing methods, Abnormal Karyotype statistics & numerical data, Congenital Abnormalities genetics, Fetal Development genetics, Fetus abnormalities, Exome Sequencing statistics & numerical data
- Abstract
Background: Fetal structural anomalies, which are detected by ultrasonography, have a range of genetic causes, including chromosomal aneuploidy, copy number variations (CNVs; which are detectable by chromosomal microarrays), and pathogenic sequence variants in developmental genes. Testing for aneuploidy and CNVs is routine during the investigation of fetal structural anomalies, but there is little information on the clinical usefulness of genome-wide next-generation sequencing in the prenatal setting. We therefore aimed to evaluate the proportion of fetuses with structural abnormalities that had identifiable variants in genes associated with developmental disorders when assessed with whole-exome sequencing (WES)., Methods: In this prospective cohort study, two groups in Birmingham and London recruited patients from 34 fetal medicine units in England and Scotland. We used whole-exome sequencing (WES) to evaluate the presence of genetic variants in developmental disorder genes (diagnostic genetic variants) in a cohort of fetuses with structural anomalies and samples from their parents, after exclusion of aneuploidy and large CNVs. Women were eligible for inclusion if they were undergoing invasive testing for identified nuchal translucency or structural anomalies in their fetus, as detected by ultrasound after 11 weeks of gestation. The partners of these women also had to consent to participate. Sequencing results were interpreted with a targeted virtual gene panel for developmental disorders that comprised 1628 genes. Genetic results related to fetal structural anomaly phenotypes were then validated and reported postnatally. The primary endpoint, which was assessed in all fetuses, was the detection of diagnostic genetic variants considered to have caused the fetal developmental anomaly., Findings: The cohort was recruited between Oct 22, 2014, and June 29, 2017, and clinical data were collected until March 31, 2018. After exclusion of fetuses with aneuploidy and CNVs, 610 fetuses with structural anomalies and 1202 matched parental samples (analysed as 596 fetus-parental trios, including two sets of twins, and 14 fetus-parent dyads) were analysed by WES. After bioinformatic filtering and prioritisation according to allele frequency and effect on protein and inheritance pattern, 321 genetic variants (representing 255 potential diagnoses) were selected as potentially pathogenic genetic variants (diagnostic genetic variants), and these variants were reviewed by a multidisciplinary clinical review panel. A diagnostic genetic variant was identified in 52 (8·5%; 95% CI 6·4-11·0) of 610 fetuses assessed and an additional 24 (3·9%) fetuses had a variant of uncertain significance that had potential clinical usefulness. Detection of diagnostic genetic variants enabled us to distinguish between syndromic and non-syndromic fetal anomalies (eg, congenital heart disease only vs a syndrome with congenital heart disease and learning disability). Diagnostic genetic variants were present in 22 (15·4%) of 143 fetuses with multisystem anomalies (ie, more than one fetal structural anomaly), nine (11·1%) of 81 fetuses with cardiac anomalies, and ten (15·4%) of 65 fetuses with skeletal anomalies; these phenotypes were most commonly associated with diagnostic variants. However, diagnostic genetic variants were least common in fetuses with isolated increased nuchal translucency (≥4·0 mm) in the first trimester (in three [3·2%] of 93 fetuses)., Interpretation: WES facilitates genetic diagnosis of fetal structural anomalies, which enables more accurate predictions of fetal prognosis and risk of recurrence in future pregnancies. However, the overall detection of diagnostic genetic variants in a prospectively ascertained cohort with a broad range of fetal structural anomalies is lower than that suggested by previous smaller-scale studies of fewer phenotypes. WES improved the identification of genetic disorders in fetuses with structural abnormalities; however, before clinical implementation, careful consideration should be given to case selection to maximise clinical usefulness., Funding: UK Department of Health and Social Care and The Wellcome Trust., (Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.