Skip to main content

Faculty of Health and Life Sciences

Professor Austin Smith FRS

Professor Austin Smith FRS

Director, Living Systems Institute

 austin.smith@exeter.ac.uk

 Living Systems Institute 

 

Living Systems Institute, University of Exeter, Stocker Road, Exeter, EX4 4QD


Overview

Pluripotent stem cell biology

Pluripotency is the capacity of single cells to generate all cell types of the animal. This cellular plasticity is the foundation of mammalian development. In the embryo pluripotency is short-lived, but in vitro pluripotent stem cells may be propagated without limit. Pluripotent stem cells may remain in a naive undifferentiated state while retaining the ability to differentiate into multiple cell lineages including the germline. Our aims are:

  1. to understand the conditions required to capture pluripotency from the early embryo;
  2. to define the regulatory network that confers broad developmental competence;
  3. to elucidate and take control of the mechanisms that direct alternative cell fates.

Collaboration with bioengineers, biophysicists, bioinformaticians and computational modellers is embedded in our research.  Our main interest is fundamental understanding. In addition, however, we aim to provide robust pluripotent stem cell platforms for biomedical applications and bioindustry.

Research Team

Group members

  • Francesca Carlisle, Experimental Officer
  • Tao Huang, Research Associate
  • Zhili Ren, Research Associate 
  • Somayyeh Tahajjodi, Research Assistant
  • Zhi (Klein) Zhang, PhD Student 
  • Jing Yen Yong, PhD Student
  • Ethan Sung, PhD Student

Enquiries are welcome from prospective PhD students and post-doctoral researchers, including compuational biologists, bioengineers and bioinformaticians. 

Principal Investigator Profile

Austin Smith studied Biochemistry at the University of Oxford where he became fascinated by pluripotency. He pursued this interest in PhD studies with Prof Martin Hooper at the University of Edinburgh and post-doctoral research in Oxford with Prof John Heath.  Austin then joined the Centre for Genome Research in Edinburgh as a Group Leader. In 1995 he became Director of Centre which he transformed into the Institute for Stem Cell Research. In 2006 Austin moved to the University of Cambridge and was founding Director of the Cambridge Stem Cell Institute until 2016. In 2019 he took up the post of Director of the Living Systems Institute at the University of Exeter.

Qualifications

Degrees

BA (Hons), Biochemistry, University of Oxford, 1982
PhD, Developmental Genetics, University of Edinburgh, 1986

Awards

  • 2000    Pfizer Academic Award, 'For pioneering work in the field of stem cell science'
  • 2002    Ellison-Cliffe Medal, Royal Society of Medicine
  • 2003    Fellow of the Royal Society of Edinburgh
  • 2003    Medical Research Council Professor
  • 2004    Member of the European Molecular Biology Organisation
  • 2006    Fellow of the Royal Society of London
  • 2010    Prix Louis-Jeantet de médicine
  • 2010    Member of Academia Europaea
  • 2016    McEwen Award for Innovation, International Society for Stem Cell Research

Career

  • Feb 1986-May 1986   Research Assistant, Dept. Zoology, University of Oxford
  • Jun 1986-Dec 1986    Postdoctoral Research Assistant, Dept. Zoology, University of Oxford
  • Jan 1987-May 1990    Postdoctoral Research Assistant, Dept. Biochemistry, Oxford
  • Jun 1990-Dec 1994    Group Leader, Centre for Genome Research, University of Edinburgh
  • Jan 1995-Dec 1995    Acting Director, Centre for Genome Research, University of Edinburgh
  • Jan 1996-Jul  2006     Reader, from 2001 Professor, University of Edinburgh
    •    1996-2005     Director, Institute for Stem Cell Research (formerly Centre for Genome Research)
    •    2004-2005     Director, MRC Centre Development in Stem Cell Biology
  • Aug 2005-Jul 2020     Professor, Department of Biochemistry & Cambridge Stem Cell Institute, University of Cambridge
  • Jan 2007-Mar 2016    Director, Wellcome Trust Centre for Stem Cell Research, later Wellcome-MRC Cambridge Stem Cell Institute
  • Nov 2019 - present     Director, Living Systems Institute, University of Exeter

Back to top


Research

Research interests

Pluripotent Stem Cell Biology
Our research focusses on understanding the developmental and molecular foundations of pluripotency in vivo and derivative pluripotent stem cell states in vitro. We seek to uncover the basic rules that govern pluripotent stem cell behaviour and apply this knowledge to control their generation, self-renewal and lineage commitment. To identify generic principles and expose species-specific adaptations we compare pluripotent cells from rodents, humans and other mammals. Our overarching goal is to recapitulate ex vivo the developmental trajectory from an emergent naïve population, through acquisition of multi-lineage competence, to lineage specification.

Research projects

Current research grants:

  • Medical Research Council Programme Grant – Human pluripotency
  • ERC Advanced Grant – Plasticity of the pluripotency network

Prospective PhD projects

  • I will be happy to consider potential PhD candidates in the field of pluripotent stem cell biology and early human development.
  • Wellcome GW4-CAT PhD Programme for Health Professionals -  I welcome expressions of interest; two clinical fellows and one MB PhD student have previously completed successful PhDs in my research group.

Publications

ORCID 0000-0002-3029-4682; 223 journal articles; H-index 103 (Scopus, Jun 2022)

Current Preprints

  • Branching topology of the human embryo transcriptome revealed by entropy sort feature weighting            www.biorxiv.org/content/biorxiv/early/2023/10/23/2023.10.12.562031.full.pdf

  • ERK signalling orchestrates metachronous transition from naïve to formative pluripotency  www.biorxiv.org/content/biorxiv/early/2023/07/20/2023.07.20.549835.full.pdf

  • Embryonic stem cells commit to differentiation by symmetric divisions following a variable lag period. bioRxiv, 2020.2006.2017.157578. 

Selected Recent Papers

  • Radley, A., Corujo-Simon, E., Nichols, J., Smith, A., and Dunn, S.-J. (2023). Entropy sorting of single-cell RNA sequencing data reveals the inner cell mass in the human pre-implantation embryo. Stem Cell Reports, doi: 10.1016/j.stemcr.2022.09.007
  • Guo, G., Stirparo, G.G., Strawbridge, S., Spindlow, D., Yang, J., Clarke, J., Dattani, A., Yanagida, A., Li, M.A., Myers, S., Özel, B.N., Nichols, J. and Smith, A. (2021). Human Naïve Epiblast Cells Possess Unrestricted Lineage Potential. Cell Stem Cell, doi: 10.1016/j.stem.2021.02.025
  • Kinoshita, M., Barber, M., Mansfield, W., Cui, Y., Spindlow, D., Stirparo, G.G., Dietmann, S., Nichols, J., and Smith, A. (2021). Capture of mouse and human stem cells with features of formative pluripotency. Cell Stem Cell, doi: 10.1016/j.stem.2020.11.005 
  • Stirparo, G.G., Smith, A., and Guo, G. (2021). Cancer-Related Mutations Are Not Enriched in Naive Human Pluripotent Stem Cells. Cell Stem Cell 28, 164-169 e162, doi: 10.1016/j.stem.2020.11.014
  • Kalkan, T., Bornelöv, S., Mulas, C., Diamanti, E., Lohoff, T., Ralser, M., Middelkamp, S., Lombard, P., Nichols, J. and Smith, A. (2019). Complementary Activity of ETV5, RBPJ, and TCF3 Drives Formative Transition from Naive Pluripotency. Cell Stem Cell, 24, 785-801.e7 doi: 10.1016/j.stem.2019.03.017
  • Rostovskaya, M., Stirparo, G. G. and Smith, A. (2019). Capacitation of human naïve pluripotent stem cells for multi-lineage differentiation. Development 146, doi: 10.1242/dev.172916

Review

  • Smith, A. (2017). Formative pluripotency: the executive phase in a developmental continuum. Development 144, 365-373. doi: 10.1242/dev.142679

Past Highlights

  • Smith, A. G., Heath, J. K., Donaldson, D. D., Wong, G. G., Moreau, J., Stahl, M. & Rogers, D. (1988). Inhibition of Pluripotential Embryonic Stem Cell Differentiation by Purified Polypeptides. Nature, 336, 688-690. doi:10.1038/336688a0
  • Nichols, J., Zevnik, B., Anastassiadis, K., Niwa, H., Klewe-Nebenius, D., Chambers, I., Schöler, H. & Smith, A. (1998). Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell, 95, 379-391. doi:10.1016/S0092-8674(00)81769-9
  • Chambers, I., Colby, D., Robertson, M., Nichols, J., Lee, S., Tweedie, S. & Smith, A.G. (2003) Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 113, 643-655. doi:10.1016/S0092-8674(03)00392-1.
  • Ying, Q.L., Wray, J., Nichols, J., Batlle-Morera, L., Doble, B., Woodgett, J., Cohen, P. & Smith, A.G. (2008) The ground state of embryonic stem cell self-renewal. Nature 453, 519-523 doi:10.1038/nature06968.
  • Takashima, Y. , Guo, G., Loos, R., Nichols, J., Ficz, G., Krueger, F., Oxley, D., Santos, F., Clarke, J., Mansfield, W., Reik, W., Bertone, P. & Smith, A. (2014) Resetting Transcription Factor Control Circuitry toward Ground-State Pluripotency in Human. Cell 158, 1254-1269. doi:10.1016/j.cell.2015.06.052.

Back to top


Publications

Journal articles

Vallier L, Vitilio L, Durance C, Smith A (In Press). GMP-grade neural progenitor derivation and differentiation from clinical-grade human embryonic stem cells. Stem Cell Research and Therapy Abstract.
Richards D, Cockerell A, Wright L, Dattani A, Guo G, Smith A, Tsaneva K (2023). Biophysical models of early mammalian embryogenesis. Stem Cell Reports, 18, 26-46.
Miroshnikova YA, Shahbazi MN, Negrete J, Chalut KJ, Smith A (2023). Cell state transitions: catch them if you can. Development, 150(6). Abstract.  Author URL.
Radley A, Corujo-Simon E, Nichols J, Smith A, Dunn SJ (2023). Entropy sorting of single-cell RNA sequencing data reveals the inner cell mass in the human pre-implantation embryo. Stem Cell Reports, 18(1), 47-63. Abstract.
Agostinho de Sousa J, Wong C-W, Dunkel I, Owens T, Voigt P, Hodgson A, Baker D, Schulz EG, Reik W, Smith A, et al (2023). Epigenetic dynamics during capacitation of naïve human pluripotent stem cells. Science Advances, 9(39). Abstract.
Ludwig TE, Andrews PW, Barbaric I, Benvenisty N, Bhattacharyya A, Crook JM, Daheron LM, Draper JS, Healy LE, Huch M, et al (2023). ISSCR standards for the use of human stem cells in basic research. Stem Cell Reports, 18(9), 1744-1752. Abstract.
Huth M, Santini L, Galimberti E, Ramesmayer J, Titz-Teixeira F, Sehlke R, Oberhuemer M, Stummer S, Herzog V, Garmhausen M, et al (2022). NMD is required for timely cell fate transitions by fine-tuning gene expression and regulating translation. Genes and Development, 34(5), 348-367. Abstract.
Dattani A, Huang T, Liddle C, Smith A, Guo G (2022). Suppression of YAP safeguards human naïve pluripotency. Development, 149(24). Abstract.  Author URL.
Stirparo GG, Smith A, Guo G (2021). Cancer-Related Mutations Are Not Enriched in Naive Human Pluripotent Stem Cells. Cell Stem Cell, 28(1), 164-169.e2. Abstract.  Author URL.
Kinoshita M, Barber M, Mansfield W, Cui Y, Spindlow D, Stirparo GG, Dietmann S, Nichols J, Smith A (2021). Capture of Mouse and Human Stem Cells with Features of Formative Pluripotency. Cell Stem Cell, 28(3), 453-471.e8. Abstract.  Author URL.
Mulas C, Chaigne A, Smith A, Chalut KJ (2021). Cell state transitions: definitions and challenges. Development, 148(20). Abstract.
Lackner A, Sehlke R, Garmhausen M, Giuseppe Stirparo G, Huth M, Titz-Teixeira F, van der Lelij P, Ramesmayer J, Thomas HF, Ralser M, et al (2021). Cooperative genetic networks drive embryonic stem cell transition from naïve to formative pluripotency. EMBO J, 40(8). Abstract.  Author URL.
Kinoshita M, Li MA, Barber M, Mansfield W, Dietmann S, Smith A (2021). Disabling de novo DNA methylation in embryonic stem cells allows an illegitimate fate trajectory. Proceedings of the National Academy of Sciences, 118(38). Abstract.
Kinoshita M, Barber M, Mansfield W, Cui Y, Spindlow D, Stirparo GG, Dietmann S, Nichols J, Smith A (2021). Erratum: Capture of Mouse and Human Stem Cells with Features of Formative Pluripotency (Cell Stem Cell (2021) 28(3) (453–471.e8), (S1934590920305439), (10.1016/j.stem.2020.11.005)). Cell Stem Cell, 28(12). Abstract.
Guo G, Stirparo GG, Strawbridge SE, Spindlow D, Yang J, Clarke J, Dattani A, Yanagida A, Li MA, Myers S, et al (2021). Human naive epiblast cells possess unrestricted lineage potential. Cell Stem Cell, 28(6), 1040-1056.e6. Abstract.  Author URL.
Yanagida A, Spindlow D, Nichols J, Dattani A, Smith A, Guo G (2021). Naive stem cell blastocyst model captures human embryo lineage segregation. Cell Stem Cell, 28(6), 1016-1022.e4. Abstract.  Author URL.
Kinoshita M, Kobayashi T, Planells B, Klisch D, Spindlow D, Masaki H, Bornelöv S, Stirparo GG, Matsunari H, Uchikura A, et al (2021). Pluripotent stem cells related to embryonic disc exhibit common self-renewal requirements in diverse livestock species. Development, 148(23). Abstract.  Author URL.
Kinoshita M, Kobayashi T, Nagashima H, Alberio R, Smith A (2021). The people behind the papers - Masaki Kinoshita, Toshihiro Kobayashi, Hiroshi Nagashima, Ramiro Alberio and Austin Smith. Development (Cambridge), 148(23).
Kinoshita M, Li MA, Barber M, Mansfield W, Dietmann S, Smith A (2020). Disabling <i>de novo</i> DNA methylation in embryonic stem cells allows an illegitimate fate trajectory. Abstract.
Mulas C, Hodgson AC, Kohler TN, Agley CC, Humphreys P, Kleine-Brüggeney H, Hollfelder F, Smith A, Chalut KJ (2020). Microfluidic platform for 3D cell culture with live imaging and clone retrieval. Lab Chip, 20, 2580-2591. Abstract.
Mayer D, Stadler MB, Rittirsch M, Hess D, Lukonin I, Winzi M, Smith A, Buchholz F, Betschinger J (2020). Zfp281 orchestrates interconversion of pluripotent states by engaging Ehmt1 and Zic2. EMBO J, 39, e102591-e102591. Abstract.
Dunn S-J, Li MA, Carbognin E, Smith A, Martello G (2019). A common molecular logic determines embryonic stem cell self-renewal and reprogramming. EMBO J, 38 Abstract.
Rostovskaya M, Stirparo GG, Smith A (2019). Capacitation of human naïve pluripotent stem cells for multi-lineage differentiation. Development, 146 Abstract.
Kalkan T, Bornelöv S, Mulas C, Diamanti E, Lohoff T, Ralser M, Middelkamp S, Lombard P, Nichols J, Smith A, et al (2019). Complementary Activity of ETV5, RBPJ, and TCF3 Drives Formative Transition from Naive Pluripotency. Cell Stem Cell, 24, 785-801.e7. Abstract.
Mulas C, Kalkan T, von Meyenn F, Leitch HG, Nichols J, Smith A (2019). Correction: Defined conditions for propagation and manipulation of mouse embryonic stem cells (doi:10.1242/dev.173146). Development, 146
Mulas C, Kalkan T, von Meyenn F, Leitch HG, Nichols J, Smith A (2019). Defined conditions for propagation and manipulation of mouse embryonic stem cells. Development (Cambridge, England), 146(6). Abstract.
Huang M, Tailor J, Zhen Q, Gillmor AH, Miller ML, Weishaupt H, Chen J, Zheng T, Nash EK, McHenry LK, et al (2019). Engineering Genetic Predisposition in Human Neuroepithelial Stem Cells Recapitulates Medulloblastoma Tumorigenesis. Cell Stem Cell, 25, 433-446.e7. Abstract.
Ostermann L, Ladewig J, Müller F-J, Kesavan J, Tailor J, Smith A, Brüstle O, Koch P (2019). In Vitro Recapitulation of Developmental Transitions in Human Neural Stem Cells. Stem Cells, 37, 1429-1440. Abstract.
Kleine-Brüggeney H, van Vliet LD, Mulas C, Gielen F, Agley CC, Silva JCR, Smith A, Chalut K, Hollfelder F (2019). Long-Term Perfusion Culture of Monoclonal Embryonic Stem Cells in 3D Hydrogel Beads for Continuous Optical Analysis of Differentiation. Small, 15(5). Abstract.
Bredenkamp N, Stirparo GG, Nichols J, Smith A, Guo G (2019). The Cell-Surface Marker Sushi Containing Domain 2 Facilitates Establishment of Human Naive Pluripotent Stem Cells. Stem Cell Reports, 12(6), 1212-1222. Abstract.  Author URL.
Bredenkamp N, Yang J, Clarke J, Stirparo GG, von Meyenn F, Dietmann S, Baker D, Drummond R, Ren Y, Li D, et al (2019). Wnt Inhibition Facilitates RNA-Mediated Reprogramming of Human Somatic Cells to Naive Pluripotency. Stem Cell Reports, 13(6), 1083-1098. Abstract.  Author URL.
Guo G, von Meyenn F, Rostovskaya M, Clarke J, Dietmann S, Baker D, Sahakyan A, Myers S, Bertone P, Reik W, et al (2018). Epigenetic resetting of human pluripotency (vol 144, pg 2748, 2017). DEVELOPMENT, 145(8).  Author URL.
Zhang M, Leitch HG, Tang WWC, Festuccia N, Hall-Ponsele E, Nichols J, Surani MA, Smith A, Chambers I (2018). Esrrb Complementation Rescues Development of Nanog-Null Germ Cells. Cell Rep, 22, 332-339. Abstract.
Stirparo GG, Boroviak T, Guo G, Nichols J, Smith A, Bertone P (2018). Integrated analysis of single-cell embryo data yields a unified transcriptome signature for the human pre-implantation epiblast. Development (Cambridge), 145(3). Abstract.
Stirparo GG, Boroviak T, Guo G, Nichols J, Smith A, Bertone P (2018). Integrated analysis of single-cell embryo data yields a unified transcriptome signature for the human pre-implantation epiblast (vol 145, dev158501, 2018). DEVELOPMENT, 145(15).  Author URL.
Stirparo GG, Boroviak T, Guo G, Nichols J, Smith A, Bertone P (2018). Integrated analysis of single-cell embryo data yields a unified transcriptome signature for the human preimplantation epiblast. Development Abstract.
Dorn T, Kornherr J, Parrotta EI, Zawada D, Ayetey H, Santamaria G, Iop L, Mastantuono E, Sinnecker D, Goedel A, et al (2018). Interplay of cell-cell contacts and RhoA/MRTF-A signaling regulates cardiomyocyte identity. EMBO J, 37 Abstract.
Sutherland L, Ruhe M, Gattegno-Ho D, Mann K, Greaves J, Koscielniak M, Meek S, Lu Z, Waterfall M, Taylor R, et al (2018). LIF-dependent survival of embryonic stem cells is regulated by a novel palmitoylated Gab1 signalling protein. J Cell Sci, 131(18). Abstract.  Author URL.
Nett IR, Mulas C, Gatto L, Lilley KS, Smith A (2018). Negative feedback via RSK modulates Erk-dependent progression from naïve pluripotency. EMBO Rep, 19 Abstract.
Kinoshita M, Smith A (2018). Pluripotency Deconstructed. Dev Growth Differ, 60, 44-52. Abstract.
Boroviak T, Stirparo GG, Dietmann S, Hernando-Herraez I, Mohammed H, Reik W, Smith A, Sasaki E, Nichols J, Bertone P, et al (2018). Single cell transcriptome analysis of human, marmoset and mouse embryos reveals common and divergent features of preimplantation development. Development, 145 Abstract.
Greaves RB, Dietmann S, Smith A, Stepney S, Halley JD (2017). A conceptual and computational framework for modelling and understanding the non-equilibrium gene regulatory networks of mouse embryonic stem cells. PLoS Comput Biol, 13, e1005713-e1005713. Abstract.
Li MA, Amaral PP, Cheung P, Bergmann JH, Kinoshita M, Kalkan T, Ralser M, Robson S, von Meyenn F, Paramor M, et al (2017). A lncRNA fine tunes the dynamics of a cell state transition involving Lin28, let-7 and de novo DNA methylation. Elife, 6 Abstract.
Guo G, von Meyenn F, Rostovskaya M, Clarke J, Dietmann S, Baker D, Sahakyan A, Myers S, Bertone P, Reik W, et al (2017). Epigenetic resetting of human pluripotency. Development (Cambridge), 144(15), 2748-2763. Abstract.
Smith A (2017). Formative pluripotency: the executive phase in a developmental continuum. Development, 144, 365-373. Abstract.
Chen Y, Spitzer S, Agathou S, Karadottir RT, Smith A (2017). Gene Editing in Rat Embryonic Stem Cells to Produce in Vitro Models and in Vivo Reporters. Stem Cell Reports, 9, 1262-1274. Abstract.
Mulas C, Kalkan T, Smith A (2017). NODAL Secures Pluripotency upon Embryonic Stem Cell Progression from the Ground State. Stem Cell Reports, 9, 77-91. Abstract.
Ying Q-L, Smith A (2017). The Art of Capturing Pluripotency: Creating the Right Culture. Stem Cell Reports, 8, 1457-1464. Abstract.
Kalkan T, Olova N, Roode M, Mulas C, Lee HJ, Nett I, Marks H, Walker R, Stunnenberg HG, Lilley KS, et al (2017). Tracking the embryonic stem cell transition from ground state pluripotency. Development, 144, 1221-1234. Abstract.
Yordanov B, Dunn S-J, Kugler H, Smith A, Martello G, Emmott S (2016). A Method to Identify and Analyze Biological Programs through Automated Reasoning. NPJ Syst Biol Appl, 2 Abstract.
Morrison G, Scognamiglio R, Trumpp A, Smith A (2016). Convergence of cMyc and β-catenin on Tcf7l1 enables endoderm specification. EMBO J, 35, 356-368. Abstract.
Marks H, Kerstens HHD, Barakat TS, Splinter E, Dirks RAM, van Mierlo G, Joshi O, Wang S-Y, Babak T, Albers CA, et al (2016). Erratum to: Dynamics of gene silencing during X inactivation using allele-specific RNA-seq. Genome Biol, 17, 22-22.
Scognamiglio R, Cabezas-Wallscheid N, Thier MC, Altamura S, Reyes A, Prendergast Á, Baumgärtner D, Carnevalli LS, Atzberger A, Haas S, et al (2016). Myc Depletion Induces a Pluripotent Dormant State Mimicking Diapause. Cell, 164, 668-680. Abstract.
Guo G, von Meyenn F, Santos F, Chen Y, Reik W, Bertone P, Smith A, Nichols J (2016). Naive Pluripotent Stem Cells Derived Directly from Isolated Cells of the Human Inner Cell Mass. STEM CELL REPORTS, 6(4), 437-446.  Author URL.
Mulas C, Kalkan T, Smith A (2016). Nodal secures pluripotency upon embryonic stem cell progression from the ground state. Abstract.
Carbognin E, Betto RM, Soriano ME, Smith AG, Martello G (2016). Stat3 promotes mitochondrial transcription and oxidative respiration during maintenance and induction of naive pluripotency. EMBO J, 35, 618-634. Abstract.
Marks H, Kerstens HHD, Barakat TS, Splinter E, Dirks RAM, van Mierlo G, Joshi O, Wang S-Y, Babak T, Albers CA, et al (2015). Dynamics of gene silencing during X inactivation using allele-specific RNA-seq. Genome Biol, 16, 149-149. Abstract.
Boroviak T, Loos R, Lombard P, Okahara J, Behr R, Sasaki E, Nichols J, Smith A, Bertone P (2015). Lineage-Specific Profiling Delineates the Emergence and Progression of Naive Pluripotency in Mammalian Embryogenesis. Dev Cell, 35, 366-382. Abstract.
Pourquié O, Bruneau B, Keller G, Smith A (2015). Looking inwards: Opening a window onto human development. Development (Cambridge), 142, 1-2.
Pourquié O, Bruneau B, Keller G, Smith A (2015). Looking inwards: opening a window onto human development. Development, 142, 1-2.
Takashima Y, Guo G, Loos R, Nichols J, Ficz G, Krueger F, Oxley D, Santos F, Clarke J, Mansfield W, et al (2015). Resetting Transcription Factor Control Circuitry toward Ground-State Pluripotency in Human (vol 158, pg 1254, 2014). CELL, 162(2), 452-453.  Author URL.
Rostovskaya M, Bredenkamp N, Smith A (2015). Towards consistent generation of pancreatic lineage progenitors from human pluripotent stem cells. Philos Trans R Soc Lond B Biol Sci, 370, 20140365-20140365. Abstract.
Herberg M, Kalkan T, Glauche I, Smith A, Roeder I (2014). A model-based analysis of culture-dependent phenotypes of mESCs. PLoS One, 9, e92496-e92496. Abstract.
Dunn S-J, Martello G, Yordanov B, Emmott S, Smith AG (2014). Defining an essential transcription factor program for naïve pluripotency. Science, 344, 1156-1160. Abstract.
Mohsen-Kanson T, Hafner A-L, Wdziekonski B, Takashima Y, Villageois P, Carrière A, Svensson M, Bagnis C, Chignon-Sicard B, Svensson P-A, et al (2014). Differentiation of human induced pluripotent stem cells into brown and white adipocytes: role of Pax3. Stem Cells, 32, 1459-1467. Abstract.
Leeb M, Dietmann S, Paramor M, Niwa H, Smith A (2014). Genetic exploration of the exit from self-renewal using haploid embryonic stem cells. Cell Stem Cell, 14, 385-393. Abstract.
Kalkan T, Smith A (2014). Mapping the route from naive pluripotency to lineage specification. Philos Trans R Soc Lond B Biol Sci, 369 Abstract.
Yang S-H, Kalkan T, Morissroe C, Marks H, Stunnenberg H, Smith A, Sharrocks AD (2014). Otx2 and Oct4 drive early enhancer activation during embryonic stem cell transition from naive pluripotency. Cell Rep, 7, 1968-1981. Abstract.
Takashima Y, Guo G, Loos R, Nichols J, Ficz G, Krueger F, Oxley D, Santos F, Clarke J, Mansfield W, et al (2014). Resetting transcription factor control circuitry toward ground-state pluripotency in human. Cell, 158(6), 1254-1269. Abstract.
Boroviak T, Loos R, Bertone P, Smith A, Nichols J (2014). The ability of inner-cell-mass cells to self-renew as embryonic stem cells is acquired following epiblast specification. Nat Cell Biol, 16, 516-528. Abstract.
Martello G, Smith A (2014). The nature of embryonic stem cells. Annu Rev Cell Dev Biol, 30, 647-675. Abstract.
Danovi D, Folarin A, Gogolok S, Ender C, Elbatsh AMO, Engström PG, Stricker SH, Gagrica S, Georgian A, Yu D, et al (2013). A high-content small molecule screen identifies sensitivity of glioblastoma stem cells to inhibition of polo-like kinase 1. PLoS One, 8, e77053-e77053. Abstract.
McLaren D, Gorba T, Marguerie de Rotrou A, Pillai G, Chappell C, Stacey A, Lingard S, Falk A, Smith A, Koch P, et al (2013). Automated large-scale culture and medium-throughput chemical screen for modulators of proliferation and viability of human induced pluripotent stem cell-derived neuroepithelial-like stem cells. J Biomol Screen, 18, 258-268. Abstract.
Martello G, Sugimoto T, Diamanti E, Joshi A, Hannah R, Ohtsuka S, Göttgens B, Niwa H, Smith A (2013). Erratum: Esrrb is a pivotal target of the Gsk3/Tcf3 axis regulating embryonic stem cell self-renewal (Cell Stem Cell (2012) 11 (491-504)). Cell Stem Cell, 12, 630-630.
Betschinger J, Nichols J, Dietmann S, Corrin PD, Paddison PJ, Smith A (2013). Exit from pluripotency is gated by intracellular redistribution of the bHLH transcription factor Tfe3. Cell, 153, 335-347. Abstract.
Martello G, Bertone P, Smith A (2013). Identification of the missing pluripotency mediator downstream of leukaemia inhibitory factor. EMBO J, 32, 2561-2574. Abstract.
Leitch HG, McEwen KR, Turp A, Encheva V, Carroll T, Grabole N, Mansfield W, Nashun B, Knezovich JG, Smith A, et al (2013). Naive pluripotency is associated with global DNA hypomethylation. Nat Struct Mol Biol, 20, 311-316. Abstract.
Smith A (2013). Nanog heterogeneity: tilting at windmills?. Cell Stem Cell, 13, 6-7. Abstract.
Brilli E, Reitano E, Conti L, Conforti P, Gulino R, Consalez GG, Cesana E, Smith A, Rossi F, Cattaneo E, et al (2013). Neural stem cells engrafted in the adult brain fuse with endogenous neurons. Stem Cells Dev, 22, 538-547. Abstract.
Leitch HG, Nichols J, Humphreys P, Mulas C, Martello G, Lee C, Jones K, Surani MA, Smith A (2013). Rebuilding pluripotency from primordial germ cells. Stem Cell Reports, 1, 66-78. Abstract.
Bianco P, Barker R, Brüstle O, Cattaneo E, Clevers H, Daley GQ, De Luca M, Goldstein L, Lindvall O, Mummery C, et al (2013). Regulation of stem cell therapies under attack in Europe: for whom the bell tolls. EMBO J, 32, 1489-1495. Abstract.
Chen Y, Blair K, Smith A (2013). Robust self-renewal of rat embryonic stem cells requires fine-tuning of glycogen synthase kinase-3 inhibition. Stem Cell Reports, 1, 209-217. Abstract.
Pourquié O, Bruneau B, Götz M, Keller G, Smith A (2013). Stem cells and regeneration: a special issue. Development, 140, 2445-2445.
Tailor J, Kittappa R, Leto K, Gates M, Borel M, Paulsen O, Spitzer S, Karadottir RT, Rossi F, Falk A, et al (2013). Stem cells expanded from the human embryonic hindbrain stably retain regional specification and high neurogenic potency. J Neurosci, 33, 12407-12422. Abstract.
Leitch HG, Smith A (2013). The mammalian germline as a pluripotency cycle. Development, 140, 2495-2501. Abstract.
Stricker SH, Feber A, Engström PG, Carén H, Kurian KM, Takashima Y, Watts C, Way M, Dirks P, Bertone P, et al (2013). Widespread resetting of DNA methylation in glioblastoma-initiating cells suppresses malignant cellular behavior in a lineage-dependent manner. Genes Dev, 27, 654-669. Abstract.
Yang S-H, Kalkan T, Morrisroe C, Smith A, Sharrocks AD (2012). A genome-wide RNAi screen reveals MAP kinase phosphatases as key ERK pathway regulators during embryonic stem cell differentiation. PLoS Genet, 8, e1003112-e1003112. Abstract.
Falk A, Koch P, Kesavan J, Takashima Y, Ladewig J, Alexander M, Wiskow O, Tailor J, Trotter M, Pollard S, et al (2012). Capture of neuroepithelial-like stem cells from pluripotent stem cells provides a versatile system for in vitro production of human neurons. PLoS One, 7, e29597-e29597. Abstract.
Blair K, Leitch HG, Mansfield W, Dumeau C-É, Humphreys P, Smith AG (2012). Culture parameters for stable expansion, genetic modification and germline transmission of rat pluripotent stem cells. Biol Open, 1, 58-65. Abstract.
Martello G, Sugimoto T, Diamanti E, Joshi A, Hannah R, Ohtsuka S, Göttgens B, Niwa H, Smith A (2012). Esrrb is a pivotal target of the Gsk3/Tcf3 axis regulating embryonic stem cell self-renewal. Cell Stem Cell, 11(4), 491-504. Abstract.
Leeb M, Walker R, Mansfield B, Nichols J, Smith A, Wutz A (2012). Germline potential of parthenogenetic haploid mouse embryonic stem cells. Development, 139, 3301-3305. Abstract.
Roode M, Blair K, Snell P, Elder K, Marchant S, Smith A, Nichols J (2012). Human hypoblast formation is not dependent on FGF signalling. Dev Biol, 361, 358-363. Abstract.
Scherf N, Herberg M, Thierbach K, Zerjatke T, Kalkan T, Humphreys P, Smith A, Glauche I, Roeder I (2012). Imaging, quantification and visualization of spatio-temporal patterning in mESC colonies under different culture conditions. Bioinformatics, 28, i556-i561. Abstract.
Juliandi B, Abematsu M, Sanosaka T, Tsujimura K, Smith A, Nakashima K (2012). Induction of superficial cortical layer neurons from mouse embryonic stem cells by valproic acid. Neurosci Res, 72, 23-31. Abstract.
Wray J, Kalkan T, Gomez-Lopez S, Eckardt D, Cook A, Kemler R, Smith A (2012). Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat Cell Biol, 14, 555-555.
van Oosten AL, Costa Y, Smith A, Silva JCR (2012). JAK/STAT3 signalling is sufficient and. dominant over antagonistic cues for the establishment of naive pluripotency. Nat Commun, 3, 817-817. Abstract.
Nichols J, Smith A (2012). Pluripotency in the embryo and in culture. Cold Spring Harb Perspect Biol, 4, a008128-a008128. Abstract.
Halley JD, Smith-Miles K, Winkler DA, Kalkan T, Huang S, Smith A (2012). Self-organizing circuitry and emergent computation in mouse embryonic stem cells. Stem Cell Res, 8, 324-333. Abstract.
Camnasio S, Delli Carri A, Lombardo A, Grad I, Mariotti C, Castucci A, Rozell B, Lo Riso P, Castiglioni V, Zuccato C, et al (2012). The first reported generation of several induced pluripotent stem cell lines from homozygous and heterozygous Huntington’s disease patients demonstrates mutation related enhanced lysosomal activity. Neurobiol Dis, 46, 41-51. Abstract.
Marks H, Kalkan T, Menafra R, Denissov S, Jones K, Hofemeister H, Nichols J, Kranz A, Stewart AF, Smith A, et al (2012). The transcriptional and epigenomic foundations of ground state pluripotency. Cell, 149, 590-604. Abstract.
Fujimoto Y, Abematsu M, Falk A, Tsujimura K, Sanosaka T, Juliandi B, Semi K, Namihira M, Komiya S, Smith A, et al (2012). Treatment of a mouse model of spinal cord injury by transplantation of human induced pluripotent stem cell-derived long-term self-renewing neuroepithelial-like stem cells. Stem Cells, 30, 1163-1173. Abstract.
Guo G, Huang Y, Humphreys P, Wang X, Smith A (2011). A <i>PiggyBac</i>-Based Recessive Screening Method to Identify Pluripotency Regulators. PLOS ONE, 6(4).  Author URL.
Wray J, Kalkan T, Gomez-Lopez S, Eckardt D, Cook A, Kemler R, Smith A (2011). Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat Cell Biol, 13, 838-845. Abstract.
Sun Y, Hu J, Zhou L, Pollard SM, Smith A (2011). Interplay between FGF2 and BMP controls the self-renewal, dormancy and differentiation of rat neural stem cells. J Cell Sci, 124, 1867-1877. Abstract.
Theunissen TW, van Oosten AL, Castelo-Branco G, Hall J, Smith A, Silva JCR (2011). Nanog overcomes reprogramming barriers and induces pluripotency in minimal conditions. Curr Biol, 21, 65-71. Abstract.
Gómez-López S, Wiskow O, Favaro R, Nicolis SK, Price DJ, Pollard SM, Smith A (2011). Sox2 and Pax6 maintain the proliferative and developmental potential of gliogenic neural stem cells in vitro. Glia, 59, 1588-1599. Abstract.
Blair K, Wray J, Smith A (2011). The liberation of embryonic stem cells. PLoS Genet, 7, e1002019-e1002019. Abstract.
Nichols J, Smith A (2011). The origin and identity of embryonic stem cells. Development, 138, 3-8. Abstract.
Smith A (2011). ’No’ to ban on stem-cell patents. Nature, 472, 418-418.
Guo G, Smith A (2010). A genome-wide screen in EpiSCs identifies Nr5a nuclear receptors as potent inducers of ground state pluripotency. DEVELOPMENT, 137(19), 3185-3192.  Author URL.
Leitch HG, Blair K, Mansfield W, Ayetey H, Humphreys P, Nichols J, Surani MA, Smith A (2010). Embryonic germ cells from mice and rats exhibit properties consistent with a generic pluripotent ground state. Development, 137, 2279-2287. Abstract.
Danovi D, Falk A, Humphreys P, Vickers R, Tinsley J, Smith AG, Pollard SM (2010). Imaging-based chemical screens using normal and glioma-derived neural stem cells. Biochem Soc Trans, 38, 1067-1071. Abstract.
Kawaguchi J, Nichols J, Gierl MS, Faial T, Smith A (2010). Isolation and propagation of enteric neural crest progenitor cells from mouse embryonic stem cells and embryos. Development, 137, 693-704. Abstract.
Moretti A, Bellin M, Jung CB, Thies T-M, Takashima Y, Bernshausen A, Schiemann M, Fischer S, Moosmang S, Smith AG, et al (2010). Mouse and human induced pluripotent stem cells as a source for multipotent Isl1+ cardiovascular progenitors. FASEB J, 24, 700-711. Abstract.
Smith A (2010). Pluripotent stem cells: private obsession and public expectation. EMBO Mol Med, 2, 113-116.
Yang J, van Oosten AL, Theunissen TW, Guo G, Silva JCR, Smith A (2010). Stat3 Activation is Limiting for Reprogramming to Ground State Pluripotency. CELL STEM CELL, 7(3), 319-328.  Author URL.
Wray J, Kalkan T, Smith AG (2010). The ground state of pluripotency. Biochem Soc Trans, 38, 1027-1032. Abstract.
Pollard S, Clarke ID, Smith A, Dirks P (2009). Brain Cancer Stem Cells: a Level Playing Field. CELL STEM CELL, 5, 468-469.
Sun Y, Kong W, Falk A, Hu J, Zhou L, Pollard S, Smith A (2009). CD133 (Prominin) negative human neural stem cells are clonogenic and tripotent. PLoS One, 4, e5498-e5498. Abstract.
Smith A (2009). Design principles of pluripotency. EMBO Mol Med, 1, 251-254.
Pollard SM, Yoshikawa K, Clarke ID, Danovi D, Stricker S, Russell R, Bayani J, Head R, Lee M, Bernstein M, et al (2009). Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell, 4, 568-580. Abstract.
Guo G, Yang J, Nichols J, Hall JS, Eyres I, Mansfield W, Smith A (2009). Klf4 reverts developmentally programmed restriction of ground state pluripotency. DEVELOPMENT, 136(7), 1063-1069.  Author URL.
Nichols J, Smith A (2009). Naive and primed pluripotent states. Cell Stem Cell, 4, 487-492. Abstract.
Silva J, Nichols J, Theunissen TW, Guo G, van Oosten AL, Barrandon O, Wray J, Yamanaka S, Chambers I, Smith A, et al (2009). Nanog is the Gateway to the Pluripotent Ground State. CELL, 138(4), 722-737.  Author URL.
Hall J, Guo G, Wray J, Eyres I, Nichols J, Grotewold L, Morfopoulou S, Humphreys P, Mansfield W, Walker R, et al (2009). Oct4 and LIF/Stat3 Additively Induce Kruppel Factors to Sustain Embryonic Stem Cell Self-Renewal. CELL STEM CELL, 5(6), 597-609.  Author URL.
Nichols J, Silva J, Roode M, Smith A (2009). Suppression of Erk signalling promotes ground state pluripotency in the mouse embryo. Development, 136, 3215-3222. Abstract.
Nichols J, Jones K, Phillips JM, Newland SA, Roode M, Mansfield W, Smith A, Cooke A (2009). Validated germline-competent embryonic stem cell lines from nonobese diabetic mice. Nat Med, 15, 814-818. Abstract.
Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying Q-L, Smith A, et al (2008). Capture of authentic embryonic stem cells from rat blastocysts. Cell, 135, 1287-1298. Abstract.
Silva J, Smith A (2008). Capturing pluripotency. Cell, 132, 532-536. Abstract.
Pollard SM, Wallbank R, Tomlinson S, Grotewold L, Smith A (2008). Fibroblast growth factor induces a neural stem cell phenotype in foetal forebrain progenitors and during embryonic stem cell differentiation. Mol Cell Neurosci, 38, 393-403. Abstract.
Sun Y, Pollard S, Conti L, Toselli M, Biella G, Parkin G, Willatt L, Falk A, Cattaneo E, Smith A, et al (2008). Long-term tripotent differentiation capacity of human neural stem (NS) cells in adherent culture. Mol Cell Neurosci, 38, 245-258. Abstract.
Batlle-Morera L, Smith A, Nichols J (2008). Parameters influencing derivation of embryonic stem cells from murine embryos. Genesis, 46, 758-767. Abstract.
Silva J, Barrandon O, Nichols J, Kawaguchi J, Theunissen TW, Smith A (2008). Promotion of reprogramming to ground state pluripotency by signal inhibition. PLoS Biol, 6, e253-e253. Abstract.
Ying Q-L, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A (2008). The ground state of embryonic stem cell self-renewal. Nature, 453, 519-523. Abstract.
Johnson CE, Crawford BE, Stavridis M, Ten Dam G, Wat AL, Rushton G, Ward CM, Wilson V, Van Kuppevelt TH, Esko JD, et al (2007). Erratum: Essential alterations of heparan sulfate during the differentiation of embryonic stem cells to sox1-enhanced green fluorescent protein-expressing neural progenitor cells (Stem Cells (2007) 25 (1913)-1923) DOI:10.1634/stemcells.2006-0445). Stem Cells, 25, 2389-2389.
Johnson CE, Crawford BE, Stavridis M, Ten Dam G, Wat AL, Rushton G, Ward CM, Wilson V, van Kuppevelt TH, Esko JD, et al (2007). Essential alterations of heparan sulfate during the differentiation of embryonic stem cells to Sox1-enhanced green fluorescent protein-expressing neural progenitor cells. Stem Cells, 25, 1913-1923. Abstract.
Kunath T, Saba-El-Leil MK, Almousailleakh M, Wray J, Meloche S, Smith A (2007). FGF stimulation of the Erk1/2 signalling cascade triggers transition of pluripotent embryonic stem cells from self-renewal to lineage commitment. Development, 134, 2895-2902. Abstract.
Chambers I, Silva J, Colby D, Nichols J, Nijmeijer B, Robertson M, Vrana J, Jones K, Grotewold L, Smith A, et al (2007). Nanog safeguards pluripotency and mediates germline development. Nature, 450, 1230-1234. Abstract.
Takashima Y, Era T, Nakao K, Kondo S, Kasuga M, Smith AG, Nishikawa S-I (2007). Neuroepithelial cells supply an initial transient wave of MSC differentiation. Cell, 129, 1377-1388. Abstract.
Glaser T, Pollard SM, Smith A, Brüstle O (2007). Tripotential differentiation of adherently expandable neural stem (NS) cells. PLoS One, 2, e298-e298. Abstract.
Smith A (2006). A glossary for stem-cell biology. Nature, 441, 1060-1060. Abstract.
Pollard SM, Conti L, Sun Y, Goffred D, Smith A (2006). Adherent neural stem (NS) cells from fetal and adult forebrain. CEREBRAL CORTEX, 16, I112-I120.
Pollard SM, Conti L, Sun Y, Goffredo D, Smith A (2006). Adherent neural stem (NS) cells from fetal and adult forebrain. Cereb Cortex, 16 Suppl 1, i112-i120. Abstract.
Smith AG (2006). Embryonic stem cells: potency and potential. EMBO Science & Society paper, Stem Cell Research - Status Prospectus Prerequisites
Pollard S, Conti L, Smith A (2006). Exploitation of adherent neural stem cells in basic and applied neurobiology. Regen Med, 1, 111-118. Abstract.
Silva J, Chambers I, Pollard S, Smith A (2006). Nanog promotes transfer of pluripotency after cell fusion. Nature, 441, 997-1001. Abstract.
Lowell S, Benchoua A, Heavey B, Smith AG (2006). Notch promotes neural lineage entry by pluripotent embryonic stem cells. PLoS Biol, 4, e121-e121. Abstract.
Blelloch R, Wang Z, Meissner A, Pollard S, Smith A, Jaenisch R (2006). Reprogramming efficiency following somatic cell nuclear transfer is influenced by the differentiation and methylation state of the donor nucleus. Stem Cells, 24, 2007-2013. Abstract.
Fouladi-Nashta AA, Jones CJP, Nijjar N, Mohamet L, Smith A, Chambers I, Kimber SJ (2005). Characterization of the uterine phenotype during the peri-implantation period for LIF-null, MF1 strain mice. Dev Biol, 281, 1-21. Abstract.
Wilmut I, West MD, Lanza RP, Gearhart JD, Smith A, Colman A, Trounson AO, Campbell KH (2005). Human embryonic stem cells. Science, 310, 1903-1903.
Conti L, Pollard SM, Gorba T, Reitano E, Toselli M, Biella G, Sun Y, Sanzone S, Ying Q-L, Cattaneo E, et al (2005). Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol, 3, e283-e283. Abstract.
Kawaguchi J, Mee PJ, Smith AG (2005). Osteogenic and chondrogenic differentiation of embryonic stem cells in response to specific growth factors. Bone, 36, 758-769. Abstract.
Kawaguchi J, Mee PJ, Smith AG (2005). Osteogenic and chondrogenic differentiation of embryonic stem cells in response to specific growth factors. Bone, 36(5), 758-769. Abstract.
Smith A (2005). The battlefield of pluripotency. Cell, 123, 757-760. Abstract.
Matveeva NM, Pristyazhnyuk IE, Temirova SA, Menzorov AG, Vasilkova A, Shilov AG, Smith A, Serov OL (2005). Unequal segregation of parental chromosomes in embryonic stem cell hybrids. Mol Reprod Dev, 71, 305-314. Abstract.
Smith AG (2004). ES cells: Research progress in UK. Cell Technology (Japan), 23, 1268-1272.
Sherwin JRA, Freeman TC, Stephens RJ, Kimber S, Smith AG, Chambers I, Smith SK, Sharkey AM (2004). Identification of genes regulated by leukemia-inhibitory factor in the mouse uterus at the time of implantation. Mol Endocrinol, 18, 2185-2195. Abstract.
Dacquin R, Mee PJ, Kawaguchi J, Olmsted-Davis EA, Gallagher JA, Nichols J, Lee K, Karsenty G, Smith A (2004). Knock-in of nuclear localised beta-galactosidase reveals that the tyrosine phosphatase Ptprv is specifically expressed in cells of the bone collar. Dev Dyn, 229, 826-834. Abstract.
Chambers I, Smith A (2004). Self-renewal of teratocarcinoma and embryonic stem cells. Oncogene, 23, 7150-7160. Abstract.
Zhao S, Nichols J, Smith AG, Li M (2004). SoxB transcription factors specify neuroectodermal lineage choice in ES cells. Mol Cell Neurosci, 27, 332-342. Abstract.
Johnson CE, Stavridis MP, Crawford BE, Wilson VA, Esko JD, Smith AG, Gallagher JT, Merry CLR (2004). The differentiation of ES cells into neuroectodermal precursors is associated with an increase in the levels and sulfation of heparan sulfate proteoglycans. INTERNATIONAL JOURNAL OF EXPERIMENTAL PATHOLOGY, 85, A65-A66.
Mee PJ, Shen MH, Smith AG, Brown WRA (2003). An unpaired mouse centromere passes consistently through male meiosis and does not significantly compromise spermatogenesis. Chromosoma, 112, 183-189. Abstract.
Ying QL, Nichols J, Chambers I, Smith A (2003). BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell, 115, 281-292. Abstract.
Ying Q-L, Stavridis M, Griffiths D, Li M, Smith A (2003). Conversion of embryonic stem cells into neuroectodermal precursors in adherent monoculture. Nat Biotechnol, 21, 183-186. Abstract.
Ying Q-L, Smith AG (2003). Defined conditions for neural commitment and differentiation. Methods Enzymol, 365, 327-341. Abstract.
Surani A, Smith A (2003). Differentiation and gene regulation programming, reprogramming and regeneration - Editorial overview. CURR OPIN GENET DEV, 13, 445-447.
Testa G, Zhang Y, Vintersten K, Benes V, Pijnappel WWMP, Chambers I, Smith AJH, Smith AG, Stewart AF (2003). Engineering the mouse genome with bacterial artificial chromosomes to create multipurpose alleles. Nat Biotechnol, 21, 443-447. Abstract.
Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A (2003). Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell, 113, 643-655. Abstract.
Buehr M, Smith A (2003). Genesis of embryonic stem cells. Philos Trans R Soc Lond B Biol Sci, 358, 1397-1402. Abstract.
Jones M, Rossant J, Robertson EJ, Smith A, Edwards RG (2003). Lineage allocation and asymmetries in the early mouse embryo - Discussion. PHILOSOPHICAL TRANSACTIONS OF THE ROYAL SOCIETY OF LONDON SERIES B-BIOLOGICAL SCIENCES, 358, 1349-1349.
Stavridis MP, Smith AG (2003). Neural differentiation of mouse embryonic stem cells. Biochem Soc Trans, 31, 45-49. Abstract.
Buehr M, Nichols J, Stenhouse F, Mountford P, Greenhalgh CJ, Kantachuvesiri S, Brooker G, Mullins J, Smith AG (2003). Rapid loss of Oct-4 and pluripotency in cultured rodent blastocysts and derivative cell lines. Biol Reprod, 68, 222-229. Abstract.
Aubert J, Stavridis MP, Tweedie S, O’Reilly M, Vierlinger K, Li M, Ghazal P, Pratt T, Mason JO, Roy D, et al (2003). Screening for mammalian neural genes via fluorescence-activated cell sorter purification of neural precursors from Sox1-gfp knock-in mice. Proc Natl Acad Sci U S A, 100 Suppl 1, 11836-11841. Abstract.
Medvinsky A, Smith A (2003). Stem cells: Fusion brings down barriers. Nature, 422, 823-825.
Ying Q-L, Nichols J, Evans EP, Smith AG (2002). Changing potency by spontaneous fusion. Nature, 416, 545-548. Abstract.
Aubert J, Dunstan H, Chambers I, Smith A (2002). Functional gene screening in embryonic stem cells implicates Wnt antagonism in neural differentiation. Nat Biotechnol, 20, 1240-1245. Abstract.
Andrews PW, Moore H, Smith A (2002). Human embryonic stem cells: prospects for human health - a 1-day international symposium held at the University of Sheffield. J Anat, 200, 221-223.
Billon N, Jolicoeur C, Ying QL, Smith A, Raff M (2002). Normal timing of oligodendrocyte development from genetically engineered, lineage-selectable mouse ES cells. J Cell Sci, 115, 3657-3665. Abstract.
Niwa H, Masui S, Chambers I, Smith AG, Miyazaki J-I (2002). Phenotypic complementation establishes requirements for specific POU domain and generic transactivation function of Oct-3/4 in embryonic stem cells. Mol Cell Biol, 22, 1526-1536. Abstract.
Burdon T, Smith A, Savatier P (2002). Signalling, cell cycle and pluripotency in embryonic stem cells. Trends Cell Biol, 12, 432-438. Abstract.
Lauer P, Metzner HJ, Zettlmeissl G, Li M, Smith AG, Lathe R, Dickneite G (2002). Targeted inactivation of the mouse locus encoding coagulation factor XIII-A: hemostatic abnormalities in mutant mice and characterization of the coagulation deficit. Thromb Haemost, 88, 967-974. Abstract.
Smith AG (2001). Embryo-derived stem cells: of mice and men. Annu Rev Cell Dev Biol, 17, 435-462. Abstract.
Nichols J, Chambers I, Taga T, Smith A (2001). Physiological rationale for responsiveness of mouse embryonic stem cells to gp130 cytokines. Development, 128, 2333-2339. Abstract.
Smith AG (2001). Self-renewal and differentiation of pluripotent embryonic stem cells. DEVELOPMENTAL BIOLOGY, 235, 289-290.
Shen MH, Mee PJ, Nichols J, Yang J, Brook F, Gardner RL, Smith AG, Brown WR (2000). A structurally defined mini-chromosome vector for the mouse germ line. Curr Biol, 10, 31-34. Abstract.
Gillespie CS, Sherman DL, Fleetwood-Walker SM, Cottrell DF, Tait S, Garry EM, Wallace VC, Ure J, Griffiths IR, Smith A, et al (2000). Peripheral demyelination and neuropathic pain behavior in periaxin-deficient mice. Neuron, 26, 523-531. Abstract.
Niwa H, Miyazaki J, Smith AG (2000). Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet, 24, 372-376. Abstract.
Wolf CR, Smith G, Smith AG, Brown K, Henderson CJ (1999). Adaptive responses to environmental chemicals. Biochem Soc Symp, 64, 129-139. Abstract.
Pagès G, Guérin S, Grall D, Bonino F, Smith A, Anjuere F, Auberger P, Pouysségur J (1999). Defective thymocyte maturation in p44 MAP kinase (Erk 1) knockout mice. Science, 286, 1374-1377. Abstract.
Aubert J, Dessolin S, Belmonte N, Li M, McKenzie FR, Staccini L, Villageois P, Barhanin B, Vernallis A, Smith AG, et al (1999). Leukemia inhibitory factor and its receptor promote adipocyte differentiation via the mitogen-activated protein kinase cascade. J Biol Chem, 274, 24965-24972. Abstract.
Svendsen CN, Smith AG (1999). New prospects for human stem-cell therapy in the nervous system. Trends Neurosci, 22, 357-364. Abstract.
Burdon T, Chambers I, Stracey C, Niwa H, Smith A (1999). Signaling mechanisms regulating self-renewal and differentiation of pluripotent embryonic stem cells. Cells Tissues Organs, 165, 131-143. Abstract.
Perarnau B, Saron MF, San Martin BR, Bervas N, Ong H, Soloski MJ, Smith AG, Ure JM, Gairin JE, Lemonnier FA, et al (1999). Single H2K(b), H2D(b) and double H2K(b)D(b) knockout mice: peripheral CD8(+) T cell repertoire and anti-lymphocytic choriomeningitis virus cytolytic responses. EUROPEAN JOURNAL OF IMMUNOLOGY, 29, 1243-1252.
Pérarnau B, Saron MF, Reina San Martin B, Bervas N, Ong H, Soloski MJ, Smith AG, Ure JM, Gairin JE, Lemonnier FA, et al (1999). Single H2Kb, H2Db and double H2KbDb knockout mice: peripheral CD8+ T cell repertoire and anti-lymphocytic choriomeningitis virus cytolytic responses. Eur J Immunol, 29, 1243-1252. Abstract.
Burdon T, Stracey C, Chambers I, Nichols J, Smith A (1999). Suppression of SHP-2 and ERK signalling promotes self-renewal of mouse embryonic stem cells. Dev Biol, 210, 30-43. Abstract.
Sen-Majumdar A, Sheehan K, Guo G, Allegre G, Simone J, Harvey M, Chow P, Moen R, Smith A (1998). A comparative study on the efficacy of CD8-positive cells in enhancing allogeneic bone marrow engraftment: Cell sorting vs microbead selection. Bone Marrow Transplantation, 22(5), 477-484. Abstract.
Smith A (1998). Cell therapy: in search of pluripotency. Curr Biol, 8, R802-R804. Abstract.
Loupart ML, Shen MH, Smith A (1998). Differential stability of a human mini-chromosome in mouse cell lines. Chromosoma, 107, 255-259. Abstract.
Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers I, Schöler H, Smith A (1998). Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell, 95, 379-391. Abstract.
Li M, Pevny L, Lovell-Badge R, Smith A (1998). Generation of purified neural precursors from embryonic stem cells by lineage selection. Curr Biol, 8, 971-974. Abstract.
Wastling JM, Knight P, Ure J, Wright S, Thornton EM, Scudamore CL, Mason J, Smith A, Miller HR (1998). Histochemical and ultrastructural modification of mucosal mast cell granules in parasitized mice lacking the beta-chymase, mouse mast cell protease-1. Am J Pathol, 153, 491-504. Abstract.
Henderson CJ, Smith AG, Ure J, Brown K, Bacon EJ, Wolf CR (1998). Increased skin tumorigenesis in mice lacking pi class glutathione S-transferases. Proc Natl Acad Sci U S A, 95, 5275-5280. Abstract.
Mountford P, Nichols J, Zevnik B, O’Brien C, Smith A (1998). Maintenance of pluripotential embryonic stem cells by stem cell selection. Reprod Fertil Dev, 10, 527-533. Abstract.
Dani C, Chambers I, Johnstone S, Robertson M, Ebrahimi B, Saito M, Taga T, Li M, Burdon T, Nichols J, et al (1998). Paracrine induction of stem cell renewal by LIF-deficient cells: a new ES cell regulatory pathway. Dev Biol, 203, 149-162. Abstract.
Nichols J, Smith A, Buehr M (1998). Rat and mouse epiblasts differ in their capacity to generate extraembryonic endoderm. Reproduction, Fertility and Development, 10(7-8), 517-525. Abstract.
Nichols J, Smith A, Buehr M (1998). Rat and mouse epiblasts differ in their capacity to generate extraembryonic endoderm. REPRODUCTION FERTILITY AND DEVELOPMENT, 10, 517-525.
Niwa H, Burdon T, Chambers I, Smith A (1998). Self-renewal of pluripotent embryonic stem cells is mediated via activation of STAT3. Genes Dev, 12, 2048-2060. Abstract.
Livesey FJ, O’Brien JA, Li M, Smith AG, Murphy LJ, Hunt SP (1997). A Schwann cell mitogen accompanying regeneration of motor neurons. Nature, 390, 614-618. Abstract.
Dani C, Smith AG, Dessolin S, Leroy P, Staccini L, Villageois P, Darimont C, Ailhaud G (1997). Differentiation of embryonic stem cells into adipocytes in vitro. J Cell Sci, 110 ( Pt 11), 1279-1285. Abstract.
Lee K, Nichols J, Smith A (1997). Erratum: Identification of a developmentally regulated protein tyrosine phosphatase in embryonic stem cells that is a marker of pluripotential epiblast and early mesoderm (Mech. Dev. 59 (1996) 153-164). Mechanisms of Development, 61, 213-215.
Pascolo S, Bervas N, Ure JM, Smith AG, Lemonnier FA, Pérarnau B (1997). HLA-A2.1-restricted education and cytolytic activity of CD8(+) T lymphocytes from beta2 microglobulin (beta2m) HLA-A2.1 monochain transgenic H-2Db beta2m double knockout mice. J Exp Med, 185, 2043-2051. Abstract.
Shen MH, Yang J, Loupart ML, Smith A, Brown W (1997). Human mini-chromosomes in mouse embryonal stem cells. Hum Mol Genet, 6, 1375-1382. Abstract.
Ramage AD, Clark AJ, Smith AG, Mountford PS, Burt DW (1997). Improved EBV-based shuttle vector system: dicistronic mRNA couples the synthesis of the Epstein-Barr nuclear antigen-1 protein to neomycin resistance. Gene, 197, 83-89. Abstract.
Schrank B, Götz R, Gunnersen JM, Ure JM, Toyka KV, Smith AG, Sendtner M (1997). Inactivation of the survival motor neuron gene, a candidate gene for human spinal muscular atrophy, leads to massive cell death in early mouse embryos. Proc Natl Acad Sci U S A, 94, 9920-9925. Abstract.
Neophytou C, Vernallis AB, Smith A, Raff MC (1997). Müller-cell-derived leukaemia inhibitory factor arrests rod photoreceptor differentiation at a postmitotic pre-rod stage of development. Development, 124, 2345-2354. Abstract.
Chambers I, Cozens A, Broadbent J, Robertson M, Lee M, Li M, Smith A (1997). Structure of the mouse leukaemia inhibitory factor receptor gene: regulated expression of mRNA encoding a soluble receptor isoform from an alternative 5’ untranslated region. Biochem J, 328 ( Pt 3), 879-888. Abstract.
Nichols J, Davidson D, Taga T, Yoshida K, Chambers I, Smith A (1996). Complementary tissue-specific expression of LIF and LIF-receptor mRNAs in early mouse embryogenesis. Mech Dev, 57, 123-131. Abstract.
Lee K, Nichols J, Smith A (1996). Identification of a developmentally regulated protein tyrosine phosphatase in embryonic stem cells that is a marker of pluripotential epiblast and early mesoderm. Mech Dev, 59, 153-164. Abstract.
Wei XQ, Charles IG, Smith A, Ure J, Feng GJ, Huang FP, Xu D, Muller W, Moncada S, Liew FY, et al (1995). Altered immune responses in mice lacking inducible nitric oxide synthase. Nature, 375, 408-411. Abstract.
Li M, Sendtner M, Smith A (1995). Essential function of LIF receptor in motor neurons. Nature, 378, 724-727. Abstract.
Mountford PS, Smith AG (1995). Internal ribosome entry sites and dicistronic RNAs in mammalian transgenesis. Trends Genet, 11, 179-184. Abstract.
Nichols J, Chambers I, Smith A (1994). Derivation of germline competent embryonic stem cells with a combination of interleukin-6 and soluble interleukin-6 receptor. Exp Cell Res, 215, 237-239. Abstract.
Mountford P, Zevnik B, Düwel A, Nichols J, Li M, Dani C, Robertson M, Chambers I, Smith A (1994). Dicistronic targeting constructs: reporters and modifiers of mammalian gene expression. Proc Natl Acad Sci U S A, 91, 4303-4307. Abstract.
Yoshida K, Chambers I, Nichols J, Smith A, Saito M, Yasukawa K, Shoyab M, Taga T, Kishimoto T (1994). Maintenance of the pluripotential phenotype of embryonic stem cells through direct activation of gp130 signalling pathways. Mech Dev, 45, 163-171. Abstract.
Robertson M, Chambers I, Rathjen P, Nichols J, Smith A (1993). Expression of alternative forms of differentiation inhibiting activity (DIA/LIF) during murine embryogenesis and in neonatal and adult tissues. Dev Genet, 14, 165-173. Abstract.
Barnett MA, Buckle VJ, Evans EP, Porter AC, Rout D, Smith AG, Brown WR (1993). Telomere directed fragmentation of mammalian chromosomes. Nucleic Acids Res, 21, 27-36. Abstract.
Whyatt LM, Düwel A, Smith AG, Rathjen PD (1993). The responsiveness of embryonic stem cells to alpha and beta interferons provides the basis of an inducible expression system for analysis of developmental control genes. Mol Cell Biol, 13, 7971-7976. Abstract.
Ure JM, Fiering S, Smith AG (1992). A rapid and efficient method for freezing and recovering clones of embryonic stem cells. Trends Genet, 8, 6-6.
Smith AG, Nichols J, Robertson M, Rathjen PD (1992). Differentiation inhibiting activity (DIA/LIF) and mouse development. Dev Biol, 151, 339-351. Abstract.
Smith AG (1992). Introduction: Mammalian stem cell systems. Seminars in Cell Biology, 3, 383-384.
Smith AG (1992). Mouse embryo stem cells: their identification, propagation and manipulation. Semin Cell Biol, 3, 385-399. Abstract.
Smith AG (1991). Culture and differentiation of embryonic stem cells. Journal of Tissue Culture Methods, 13, 89-94. Abstract.
Smith AG, Rathjen P (1991). Embryonic stem cells, differentiation inhibiting activity, and the mouse embryo. Seminars in Cell Biology, 317-327.
Rathjen PD, Nichols J, Toth S, Edwards DR, Heath JK, Smith AG (1990). Developmentally programmed induction of differentiation inhibiting activity and the control of stem cell populations. Genes Dev, 4, 2308-2318. Abstract.
Rathjen PD, Toth S, Willis A, Heath JK, Smith AG (1990). Differentiation inhibiting activity is produced in matrix-associated and diffusible forms that are generated by alternate promoter usage. Cell, 62, 1105-1114. Abstract.
Nichols J, Evans EP, Smith AG (1990). Establishment of germ-line-competent embryonic stem (ES) cells using differentiation inhibiting activity. Development, 110, 1341-1348. Abstract.
Heath JK, Smith AG, Hsu LW, Rathjen P (1990). Growth and Differentiation factors in embryonic stem cell function. Journal of Cell Science Suplement, 12, 75-85.
Smith AG, Leary AG, Wong GG, Clark SC, Ogawa M (1990). Leukemia inhibitory factor LIF/Differentiation-Inhibiting activity (DIA)/human interleukin for DA cells (HILDA) augments proliferation of human hematopoietic cells. Blood, 75, 1960-1964.
Heath JK, Smith AG, Wills AJ, Edwards DR (1989). Growth and differentiation factors of embryonic stem cells. Cell to cell signalling in mammalian Development, 219-230.
Heath JK, Smith AG (1989). Growth factors in embryogenesis. Br Med Bull, 45, 319-336. Abstract.
Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, Stahl M, Rogers D (1988). Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature, 336, 688-690. Abstract.
Heath JK, Smith AG (1988). Regulatory factors of embryonic stem cells. J Cell Sci Suppl, 10, 257-266. Abstract.
Smith AG, Hooper ML (1987). Buffalo rat liver cells produce a diffusible activity which inhibits the differentiation of murine embryonal carcinoma and embryonic stem cells. Dev Biol, 121, 1-9. Abstract.
Smith TA, Smith AG, Hooper ML (1986). Selection of a mouse embryonal carcinoma clone resistant to the inhibition of metabolic cooperation by retinoic acid. Exp Cell Res, 165, 417-428. Abstract.

Chapters

Smith A (2004). Converting ES cell into neurons. In  (Ed) , 83-93.
Smith AG (2001). Embryonic Stem Cells. In Marshak DR, Gardner RL, Gottlieb DI (Eds.) Stem cell biology, Cold Spring Harbor Laboratory Pr, 205-230. Abstract.
Li M, Price D, Smith A (2001). Lineage selection and isolation of neural precursors from embryonic stem cells. In  (Ed) , 29-42.
Wolf CR, Campbell SJ, Clark AJ, Smith A, Bishop JO, Henderson CJ (1998). The use of transgenic animals to assess the role of metabolism in target organ toxicity. In  (Ed) , 443-453.
Smith AG, Hole N (1994). Embryonic Stem Cells and Haematopoiesis. In Freshney RI, Pragnell IB, Freshney MG (Eds.) Culture of hematopoietic cells, Wiley-Blackwell, 235-250. Abstract.

Conferences

Moretti A, Jung C, Bellin M, Nhan TM, Takashima Y, Bernshausen A, Schiemann M, Frei EM, Moosmang S, Smith AG, et al (2009). Induced pluripotent stem cells as a source for multipotent ISL1+cardiovascular progenitors.
Thornton K, Smith A, Merry CLR, Ulijn RV (2009). Phosphatase induced stiffness control in a self-assembled peptide hydrogel.
Wong KJ, Arvind DK, Sharwood-Smith N, Smith A (2005). Specknet-based responsive environments. Abstract.
Nichols J, Ying QL, Smith AG (2005). Understanding embryonic stem cells.
Chuang TT, Griffiths D, Evans N, Smith A, Schroeder J (2004). Regulation of neural stem cells by beta amyloid in vitro.
Mohamet L, Fouladi-Nashta A, Nijjar N, Smith AG, Kimber SJ (2004). Regulation of uterine gene expression by leukaemia inhibitory factor.
Nichols J, Tzouanacou E, Wilson V, Smith A (2003). The role of Oct-4 in early post-implantation development.
Nichols J, Tzouanacou E, Wilson V, Smith A (2003). The role of Oct-4 in early post-implantation development.
Heath JK, Smith AG, Hsu LW, Rathjen PD (1990). Growth and differentiation factors of pluripotential stem cells. Abstract.

Back to top


External Engagement and Impact

Awards and Honours

  • 2000    Pfizer Academic Award, 'For pioneering work in the field of stem cell science'
  • 2002    Ellison-Cliffe Medal, Royal Society of Medicine
  • 2003    Fellow of the Royal Society of Edinburgh
  • 2003    Medical Research Council Professor
  • 2004    Member of the European Molecular Biology Organisation
  • 2006    Fellow of the Royal Society of London
  • 2010    Prix Louis-Jeantet de médicine
  • 2010    Member of Academia Europaea
  • 2016    McEwen Award for Innovation, International Society for Stem Cell Research

Editorships and Editorial Boards

  • Development, Editor, 2006-2018
  • EMBO Molecular Medicine, Editorial Board, 2009-2015 
  • The EMBO Journal, Editorial Board, 2011- present
  • Stem Cell Reports, Editorial Board, 2013-present

Expert Groups

  • Royal Society Working Group on Stem Cell Research and Therapeutic Cloning, 2000
  • Academy of Medical Sciences Working Group on Interspecies Chimaeras, 2007
  • EMBL Council Working Group on Human Embryonic Stem Cells and Embryo Research, 2020-21

Funding Panels

  • The Wellcome Trust Molecular and Cell Panel (2000 – 2003)

Scientific Advisory Boards

  • RIKEN Center for Developmental Biology, Kobe, Japan (2000 – 2015); Chair from 2007-2015
  • University of Lund BMC Biomedical Centre, Lund, Sweden (2002-08)
  • Ontario Institute for Cancer Research, Cancer Stem Cell Program, Toronto, Canada (2008-13)
  • Global Center of Excellence (G-COE), Kyoto University, Japan (2009, 2012)
  • RIKEN Advisory Council (RAC), Tokyo, Japan (2009, 2014, 2017, 2019).
  • Stem Cell Institute for Regenerative Medicine, Erasmus University, Netherlands (2010)
  • REVIVE Stem Cell Network, Paris, France (2011- present)
  • Cell Fate and Cancer Programme, Centre for Genomic Regulation, Barcelona, Spain (2012)
  • Wellcome Trust Sanger Institute Cellular Genetics Programme, Hinxton, UK (2014)
  • Cell Therapy Catapult Academic Advisory Board, London, UK (2014-2018)
  • Izmir Biomedicine and Genome Center, Dokuz Eylul University, Turkey (2015-2018)

Back to top


Teaching

BIO3086  - Stem cells and Disease lectures

BIO4041 - Stem cell biology lecture

Back to top


Edit Profile