Listening to scientific talks makes me nervous; it's so easy to conflate and confuse fast-flowing information. At the most recent meeting of the International Society for Stem Cell Research (ISSCR) this July in Barcelona, I frequently found myself wishing for the slower pace of reading the relevant source material. Others must also want such a list, I reasoned.

Here are some of the research papers, books and other sources that were discussed in talks given during the meeting. Unfortunately, we cannot list work that is still in the submission and preparation process, so there is no way to fully represent all work that was presented. I hope people will update this list by adding comments to the blog entry ISSCR reading list. I also hope that this list, necessarily incomplete, will be useful for the stem cell community.

This list would not have been possible without the cooperation of Michael Hagedorn at the ISSCR, who kindly agreed to contact the speakers; the speakers themselves; and especially the time and wisdom of Nature Publishing Group copy editor Nicole DeGennaro. On behalf of anyone who appreciates this list, I'd like to thank them.

Plenary I: Presidential Symposium on Brain Regeneration and Disease Repair

Supported by BD Biosciences

Fred H. Gage, Salk Institute for Biological Studies, USA

  • Coufal, N. G. et al. L1 retrotransposition in human neural progenitor cells. Nature 460, 1127–1131 (2009).

Elena Cattaneo, University of Milan, Italy

  • Zuccato, C. et al. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nature Genet. 35, 76–83 (2003).

  • Zuccato, C. & Cattaneo, E. Brain-derived neurotrophic factor in neurodegenerative diseases. Nature Rev. Neurol. 5, 311–322 (2009).

No response or work in submission

Marianne Bronner-Fraser, California Institute of Technology, USA

Etienne Hirsch, French National Institute for Health & Medical Research (INSERM) France

Nancy S. Wexler, Columbia University, The Hereditary Disease Foundation, USA

Plenary Session II: Signals Controlling Renewal & Differentiation

Supported by Black Family Stem Cell Institute

Elaine Fuchs, Rockefeller University, USA

  • Fuchs, E. Finding one's niche in the skin. Cell Stem Cell 4, 499–502 (2009).

  • Fuchs, E. The tortoise and the hair: slow-cycling cells in the stem cell race. Cell 137, 811–819 (2009).

  • Ezhkova, E. et al. Ezh2 Orchestrates Gene Expression for the Stepwise Differentiation of Tissue-Specific Stem Cells. Cell 136, 1122–1135 (2009).

  • Greco, V. et al. A two-step mechanism for stem cell activation during hair regeneration. Cell Stem Cell 4, 155–169 (2009).

  • Blanpain, C. & Fuchs, E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nature Rev. Mol. Cell Biol. 10, 207–217 (2009).

  • Nowak, J. A., Polak, L., Pasolli, H. A. & Fuchs, E. Hair follicle stem cells are specified and function in early skin morphogenesis. Cell Stem Cell 3, 33–43 (2008).

Yukiko Gutoh, University of Tokyo, Japan

  • Hirabayashi, Y. et al. Polycomb limits the neurogenic competence of neural precursor cells to promote astrogenic fate transition. Neuron (in the press).

Paolo Bianco, Sapienza University of Rome

  • Sacchetti, B. et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize the hematopoietic microenvironment. Cell 131, 324–336 (2007).

  • Bianco, P., Robey, P. & Simmons, P. Mesenchymal stem cells: revisiting history, concepts and assays. Cell Stem Cell 2, 313–319 (2008).

  • Dellavalle, A. et al. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nature Cell Biol. 9, 255–267 (2007).

No response or work in submission

Haifan Lin, Yale University School of Medicine, USA

Azim Surani, Wellcome Trust & Cancer Research UK Gurdon Institute, UK

Plenary Session III: Regulating Cell Type in the Endoderm

Supported by the New York Stem Cell Foundation (NYSCF)

Hans C. Clevers, Hubrecht Laboratory, Netherlands

Harry Heimberg, Diabetes Research Center, Vrije Universiteit Brussels, Belgium

  • Xu, X. et al. b cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 132, 197–207 (2008).

  • Collombat, P. et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into a and subsequently b cells. Cell 138, 449–462 (2009).

Douglas A. Melton, Harvard University, USA

  • Chen, S. et al. A small molecule that directs differentiation of human ESCs into the pancreatic lineage. Nature Chem. Biol. 5, 258–265 (2009).

  • Maehr, R. Generation of pluripotent stem cells from patients with type 1 diabetes Proc. Natl Acad. Sci. USA advance online publication, doi: 10.1073/pnas.0906894106 (31 Aug 2009)

  • Further reading: Efficient ES cell differentiation: the right tweak at the right time

No response or in submission

Mark A. Krasnow, Howard Hughes Medical Institute Stanford University, USA

Sara Ferber, Endocrine Institute Sheba Medical Center, Israel

Concurrent Session I: Embryonic Stem Cells

Supported by Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA

Kathrin Plath, University of California, Los Angeles, USA

Miguel Ramalho-Santo, University of California, San Francisco, USA

  • Gaspar-Maia, A. et al. Chd1 regulates open chromatin and pluripotency of embryonic stem cells. Nature 460, 863–868 (2009).

  • Further reading: The architecture of pluripotency

Ayelet Lesman, Technion-Israel Institute of Technology, Israel

  • Caspi, O. et al. Tissue engineering of vascularized cardiac muscle from human embryonic stem cells. Circ. Res. 100, 263–272 (2007).

  • Lesman, A. et al. Transplantation of a tissue-engineered human vascularized cardiac muscle. Tissue Eng. Part A doi:10.1089/ten.TEA.2009.0130 (published online 30 July 2009).

No response or in submission

Nikola Baschuk, University of Cologne, Germany

Yi Sun, UCLA School of Medicine, USA

Stem Cells in Model Organisms

Victoria Prince, University of Chicago, USA

  • Kinkel, M. D. et al. Cyp26 enzymes function in endoderm to regulate pancreatic field size. Proc. Natl Acad. Sci. USA 106, 7864–7869 (2009).

No response or in submission

Parthive H. Patel, Fred Hutchinson Cancer Research Center, USA

Catherine M. Browne, Griffith University, Australia

Peter W. Reddien, Broad Institute MIT, USA

Cell Cycle & Timing Mechanisms

Andreas Trumpp, Deutsches Krebsforschungszentrum (DKFZ), Germany

  • Essers, M. A. G. et al. IFNa activates dormant haematopoietic stem cells in vivo. Nature 458, 904–908 (2009).

  • Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

Michael Sieweke, Universite de la Medterrane, CIML, France

  • Sarrazin, S. et al. MafB restricts M-CSF-dependent myeloid commitment divisions of hematopoietic stem cells. Cell 138, 300–313 (2009).

  • Further reading: The innate debate over HSCs

No response or in submission

Michael J. Edel, Centro de Medicina Regenerativa de Barcelona CMR[B], Spain

Paloma Garcia, Institute of Biomedical Research, UK

Yan Liu, Memorial Sloan-Kettering Cancer Center, USA

The Ethics of Egg Sharing

Donna Dickenson, University of London, UK

  • Baylis, F. & McLeod, C. The stem cell debate continues: the buying and selling of eggs for research. J. Med. Ethics 33, 726–731 (2007).

  • Dickenson, D. Body Shopping: Converting Body Parts to Profit (Oneworld, Oxford, 2009).

  • Dickenson, D. The threatened trade in human ova. Nature Rev. Genet. 5, 167 (2004).

  • Dickenson, D. & Alkorta, I. Ova donation for research: an international comparative perspective. Int. J. Fem. Approaches Bioeth. 1, 125–144 (2008).

  • Liu, Y. et al. Germline stem cells and neo-oogenesis in the adult human ovary. Dev. Biol. 306, 112–120 (2007).

  • Pennings, G. & Devroey, P. Subsidized in-vitro fertilization treatment and the effect on the number of egg sharers. Reprod. Biomed. Online 1, 8–10 (2006).

  • The Empire State Stem Cell Board. Statement of the empire state stem cell board on the compensation of oocyte donors. http://www.stemcell.ny.gov/docs/ESSCB_Statement_on_Compensation_of_Oocyte_Donors.pdf. (11 June 2009).

No response or in submission

Alison Murdoch, International Centre for Life, UK

Controlling Tissue Stem Cells

Eric Deneault, University of Montreal, Quebec, Canada

  • Deneault, E. et al. A functional screen to identify novel effectors of hematopoietic stem cell activity. Cell 137, 369–379 (2009).

Irina M. Conboy, University of California, Berkeley, USA

  • Carlson, M. et al. Molecular aging and rejuvenation of human muscle stem cells. EMBO J. (in the press).

No response or in submission

Leif Carlsson, Umea Center for Molecular Medicine, Sweden

Jonas Frisen, Karolinska Institute, Sweden

Plenary Session IV: Hematopoietic Stem Cells in Differentiation & Disease

Meinrad Busslinger, Research Institute of Molecular Pathology, Austria

  • Cobaleda, C., Jochum, W. & Busslinger, M. Conversion of mature B cells into T cells by dedifferentiation to uncommitted progenitors. Nature 449, 473–477 (2007).

  • Cobaleda, C., Schebesta, A., Delogu, A. & Busslinger, M. Pax5: the guardian of B cell identity and function. Nature Immunol. 8, 463–470 (2007).

  • Schebesta, A. et al. Transcription factor Pax5 activates the chromatin of key genes involved in B cell signaling, adhesion, migration, and immune function. Immunity 27, 49–63 (2007).

  • Delogu, A. et al. Gene repression by Pax5 in B cells is essential for blood cell homeostasis and is reversed in plasma cells. Immunity 24 269–281 (2006).

Leonard I. Zon, Children's Hospital Boston, USA

  • Goessling, W. et al. Genetic interaction between PGE2 and wnt signaling regulates developmental specification of stem cells and regeneration. Cell 136, 1136–1146 (2009).

  • North, T. E. et al. Hematopoietic stem cell development is dependent on blood flow. Cell 137 736–748 (2009).

No response or in submission

Claudio Bordignon, Vito-Salute San Raffaele University, Italy

Ana Cumano, Institute Pasteur, France

Plenary Session V: Growth Control in Stem Cells and Cancer

Maria Blasco, Spanish National Cancer Research Centre, Spain

  • Marion, R. M. et al. Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 4 141–154 (2009).

  • Marion, R. M. et al. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 460 1149–1153 (2009).

  • Martinez, P. et al. Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice. Genes Dev. doi:10.1101/gad.543509 (published online 13 August 2009).

Jane E. Visvader, Walter & Eliza Hall Institute of Medical Research, Australia

  • Lim, E. et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nature Med. 15, 907–913 (2009).

No response or in submission

Eduard Batlle, Institute for Research in Biomedicine (IRB Barcelona), Spain

Thea Tlsty, University of California, San Francisco, USA

Yann Barrandon, EPFL-Laboratory of Cell Dynamics, Switzerland

Concurrent Session II: Differentiation of Stem Cells

No response or in submission

Hongkui Deng, Peking University, China

Christopher A. Fasano, Memorial Sloan-Kettering Cancer Center, USA

Xiaoying Bai, HHMI/Children's Hospital Boston, USA

Hideyuki Okano, Keio University, Japan Presented by Saturo Matsuda

Stem Cell Fate Choice

Deepak Srivastava, Gladstone Institute of Cardiovascular Disease, USA

  • Cordes, K. R. et al. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 460 705–710 (2009).

  • Further reading: Add microRNA to make smooth muscle

No response or in submission

Stuart Orkin, Harvard Medical School, USA

Laurraine M. Gereige, University of California, Los Angeles, USA

Jonathan R. Yeh, McGill University, Canada

Guoji Guo, Genome Institute of Singapore, Singapore

Stem Cell Technologies

Peter W. Zandstra, University of Toronto, Canada

  • Kirouac, D. C. et al. Cell–cell interaction networks regulate blood stem and progenitor cell fate. Mol. Syst. Biol. 5 293 (2009).

  • Discher, D. E., Mooney, D. J. & Zandstra, P. W. Growth factors, matrices, and forces combine and control stem cells. Science 324 1673–1677 (2009).

No response or in submission

Fernando Camargo, Children's Hospital and Harvard University, USA

Peter Christalla, University Medical Center Goettingen, Germany

Cato T. Laurencin, University of Connecticut Health Center, USA

Stem Cell Lineages, Asymmetry & Specification

Timm Schroeder, Helmholtz Center, Germany

  • Rieger, M. A., Hoppe, P. S., Smejkal, B. M., Eitelhuber, A. C. & Schroeder, T. Hematopoietic cytokines can instruct lineage choice. Science 325, 217–218 (2009).

  • Further reading: The innate debate over HSCs

No response or in submission

Gerald de Haan, UMCG, The Netherlands

Sylvia K. Nicolis, University of Milano Bicocca, Italy

Andrew W. Duncan, Oregon Health & Science University, USA

Cheryle A. Seguin, University of Western Ontario, Canada

Plenary Session VI: Cells that Build Bodies

Jeremy Brockes, University College London, UK

  • Kumar, A., Godwin, J. W., Gates, P. B., Garza-Garcia, A. A. & Brockes, J. P. Molecular basis for the nerve dependence of limb regeneration in an adult vertebrate. Science 318, 722–727 (2007).

Elly Tanaka, Center for Regenerative Therapies, Germany

  • Kragl, M. et al. Cells keep a memory of their tissue origin during axolotl limb regeneration. Nature 460, 60–65 (2009).

  • Alvarado, A. S. A cellular view of regeneration. Nature 460, 39–40 (2009).

  • Further reading: Regenerating limb tissue may not dedifferentiate

No response or in submission

Margaret Buckingham, Pasteur Institute, France

Paolo Macchiarini, Hospital Clinico de Barcelona, Spain

Olivier Pourquie, Stowers Institute for Medical Research, USA

Plenary Session VII: Stem Cells as Model Systems

Debbie Yelon, Skirball Institute of Biomolecular Medicine, USA

  • Thomas, N. A., Koudijs, M., van Eeden, F. J. M., Joyner A. L. & Yelon, D. Hedgehog signaling plays a cell-autonomous role in maximizing cardiac developmental potential. Development 135, 3789–3799 (2008).

  • de Pater, E. et al. Distinct phases of cardiomyocyte differentiation regulate growth of the zebrafish heart. Development 136, 1633–1641 (2009).

  • Waxman, J. S., Keegan, B. R., Roberts, R. W., Poss, K. D. & Yelon, D. Hoxb5b acts downstream of retinoic acid signaling in the forelimb field to restrict heart field potential in zebrafish. Dev. Cell 15, 923–934 (2008).

Shinya Yamanaka, Kyoto University, Japan

  • Yoshida, Y., Takahashi, K., Okita, K., Ichisaka, T. & Yamanaka, S. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell (in the press).

  • Hong, H. et al. Suppression of induced pluripotent stem cell generation by the p53–p21 pathway. Nature 460, 1132–1135 (2009).

  • Miura, K. et al. Variation in the safety of induced pluripotent stem cell lines. Nature Biotechnol. 27, 743–745 (2009).

  • Further reading: p53 prevents pluripotency; Cell origin and variation in induced pluripotent stem cell lines

Konrad Hochedlinger, Massachusetts General Hospital, USA

  • Utikal, J. et al. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature 460, 1145–1148 (2009).

  • Eminli, S. et al. Differentiation stage determines potential of hematopoietic cells for reprogramming into induced pluripotent stem cells Nature Genetics 41, 968–976 (2009)

  • Further reading: p53 prevents pluripotency

No response or in submission

Allan Spradling, HHMI/Carnegie Institute, USA

Concurrent Session III: Stem Cells and Cancer

Florence Guibal, INSERM, France

  • Guibal, F. C. et al. Identification of a myeloid committed progenitor as the cancer initiating cell in acute promyelocytic leukemia. Blood (in the press).

No response or in submission

Scott Armstrong, Children's Hospital Boston/Dana-Farber Cancer Institute, USA

David M. Langenau, Massachusetts General Hospital, USA 65

Emmanuelle Charafe-Jauffret, Centre de Recherche en Cancérologie de Marseille, France

Catherine A. O'Brien, University Health Network, Canada

Epigenetics & Reprogramming

Han Li, Spanish National Cancer Research Center, Spain

  • Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

No response or in submission

Huck-Hui Ng, National University of Singapore Genome Institute, Singapore

Jacob Hanna, Whitehead Institute for Biomedical Research, USA

Joel W. Blanchard, Harvard Stem Cell Institute, USA

Tuempong Wongtawan, University of Edinburgh, UK

Clinical Translation of Stem Cells

Supported by The International Society of Cellular Therapy (ISCT)

Angelo Lepore, The Johns Hopkins School of Medicine, USA

  • Lepore, A. C. et al. Focal transplantation–based astrocyte replacement is neuroprotective in a model of motor neuron disease. Nature Neurosci. 11, 1294–1301 (2008).

No response or in submission

Edwin M. Horwitz, Children's Hospital of Philadelphia, USA

Jakub Tolar, University of Minnesota, USA

Mya S. Thu, City of Hope Medical Center, USA

Augusto Zani, UCL Institute of Child Health, UK

Systems Biology and “–omics” of Stem Cells

Justin Brumbaugh, University of Wisconsin, USA

  • Phanstiel, D. et al. Mass spectrometry identifies and quantifies 74 unique histone H4 isoforms in differentiating human embryonic stem cells. Proc. Natl Acad. Sci. USA 105, 4093–4098 (2008).

  • Brumbaugh, J., Phanstiel, D. & Coon, J. J. Unraveling the histone's potential: a proteomics perspective. Epigenetics 3, 254–257 (2008).

No response or in submission

Rong Lu, Stanford University, USA

Jiaqian Wu, Yale University, USA

Stem cell niche

Valentina Greco, Rockefeller University, USA

  • Greco, V. et al. A two-step mechanism for stem cell activation during hair regeneration. Cell Stem Cell 4, 155–169 (2009).

Trista E. North, Harvard Stem Cell Institute, USA

  • North, T. E. et al. Hematopoietic stem cell development is dependent on blood flow. Cell 137, 736–748 (2009).

No response or in submission

Hanna Mikkola, University of California, Los Angeles, USA

Shane R. Mayack, Joslin Diabetes Center, USA

Leanne Jones, Institute for Biological Studies, USA

Closing Plenary VIII: Reprogramming and Epigenetics

Supported by the Harvard Stem Cell Institute, MGH Center for Regenerative Medicine & The Stem Cell Program at Children's Hospital Boston

Richard A. Young, Massachusetts Institute of Technology, USA

  • Guenther, M., Levine, S. S., Boyer, L. A., Jaenisch, R. & Young, R. A. A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130, 77–88 (2007).

  • Cole, M. F., Johnstone, S. E., Newman, J. J., Kagey, M. H. & Young, R. A. Tcf3 is an integral component of the core regulatory circuitry of embryonic stem cells. Genes Dev. 22, 746–755 (2008).

  • Jaenisch, R. & Young, R. A. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell 132, 567–582 (2008).

  • Marson, A. et al. Wnt signaling promotes reprogramming of somatic cells to pluripotency. Cell Stem Cell 3, 132–135 (2008).

  • Marson, A. et al. Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 134, 521–533 (2008).

  • Seila, A. C. et al. Divergent transcription from active promoters. Science 322, 1849–1851 (2008).

Luciano Di Croce, Center for Genomic Regulation, Spain

  • Lee, M. G. et al. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science 318, 447–450 (2007).

  • Villa, R. et al. Role of the polycomb repressive complex 2 in acute promyelocytic leukemia. Cancer Cell 11, 513–525 (2007).

  • Di Croce, L. Chromatin modifying activity of leukaemia associated fusion proteins. Hum. Mol. Genet. 14, R77–R84 (2005).

  • Di Croce, L. et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 295, 1079–1082 (2002).

Juan Carlos Izpisùa Belmonte, Center of Regenerative Medicine in Barcelona, Spain

  • Aasen, T. et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nature Biotechnol. 26, 1276–1284 (2008).

  • Gonzalez, F. et al. Generation of mouse-induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector. Proc. Natl Acad. Sci. USA 106, 8918–8922 (2009).

  • Raya, A. et al. Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature 460, 53–59 (2009).

  • Kawamura, T. et al. The p53 pathway as a guardian against reprogramming. Nature (in the press).

Richard Gregory, Children's Hospital Boston, USA

  • Hagan, J. P., Piskounova, E. & Gregory, R. I. Lin28 recruits the TUTase Zcchc11 to inhibit let-7 maturation in mouse embryonic stem cells. Nature Struct. Mol. Biol. advance online publication, doi:10.1038/nsmb.1676 (27 August 2009).

No response or in submission

Janet Rossant, Hospital for Sick Children, Canada