Martin Zenke

Last updated
Martin Zenke
Portrait martin zenke.jpg
Born (1953-08-07) August 7, 1953 (age 70)
NationalityGerman
Alma mater
Scientific career
Fields stem cell research, cell biology, molecular biology, cancer biology, bioengineering
Institutions
Thesis Transcription of SV40 Chromatin (1982)

Martin Zenke born August 7, 1953, in Korbach is a German biochemist, cell biologist, professor for cell biology and scientist, who is conducting research on stem cells and biomedical engineering.

Contents

Biography

Martin Zenke grew up in Korbach/Waldeck, Germany and finished school at Alte Landesschule in Korbach in 1972. He studied chemistry/biochemistry and medicine at Philipps-University, Marburg/Lahn, Germany (1972-1978) and graduated in 1978 with a study on “The ribonucleotide reductase in synchronized cultures of Saccharomyces cerevisiae (baker’s yeast)”.

Professional career

In 1979 he moved to German Cancer Research Center (DKFZ), Heidelberg, Institute of Virology, Section DNA Tumor Viruses (Gerhard Sauer) for doctoral studies. [1] He received his PhD from Ruprecht-Karls-University, Heidelberg in 1982 on “Transcription of SV40 Chromatin”. [2]

From 1982 to 1985 Martin Zenke worked as postdoctoral fellow with Pierre Chambon at Université Louis Pasteur, Faculté de Médecine and Laboratoire de Genetique Moleculaire des Eucaryotes (LGME) in Strasbourg, France. [3] [4] [5] From 1985 to 1988 he was EMBL fellow and staff scientist [6] in the Differentiation Programme of European Molecular Biology Laboratory (EMBL), Heidelberg, Germany with Thomas Graf and Hartmut Beug. [7]

In 1988 he moved to the Research Institute of Molecular Pathology (IMP), Vienna, Austria to work as a Junior Scientist until 1995. [8] [9] [10] [11] [12] [13] In 1992 he received his senior lecture qualification in Molecular Genetics from the Faculty of Natural Sciences, University of Vienna, Vienna, Austria. From 1995 to 2003 Martin Zenke was a Research Group Leader at Max Delbrück Center for Molecular Medicine (MDC) in Berlin, Germany. [14] [15]

Since 2003 he is Professor of Cell Biology and Chairman, and the founding director of the Institute for Biomedical Engineering, Chair of Cell Biology at Rheinisch-Westfälische Technische Hochschule (RWTH) Medical School, Aachen, Germany. [16] [17] Since 2008 he is a member of the "Central Ethics Committee for Stem Cell Research", Federal Ministry of Education and Research (BMBF) and Federal Ministry of Health (BMG), Berlin, Germany. 2011-2014 he was the Managing Director of Helmholtz-Institute for Biomedical Engineering (3 years legislative period), RWTH Aachen University, Aachen, Germany.

Main research

1979-1986: SV40 Enhancer and SV40 Chromatin

In the 1980s Martin Zenke’s research focused on gene transcription and chromatin. [1] In 1986 he and his colleagues showed that transcriptional enhancers exhibit a modular structure and are composed of individual elements, which on their own are relatively weak but act in synergy, and thereby build up enhancer activity. [3] [4] [5] This is textbook knowledge nowadays but in the 1980s enhancers were initially thought to boost transcription by a unique and particular strong enhancer sequence and factor. Martin Zenke’s seminal work is depicted and referenced in Lewin’s Genes IX, [18] the standard molecular biology textbook.

1986-1998: The erbA Oncogene and Red Blood Cell Differentiation

In 1988 Martin Zenke started to work on retroviral oncogenes, in particular on the v-erbA and v-rel oncogenes. [7] [9] [10] [13] He found that the v-erbA oncogene is a loss of function version of the c-erbA/thyroid hormone receptor and acts as a dominant negative transcriptional repressor. [7] [9] [10] This was the first description of oncogenic activity by loss-of-function mutation. [19] [20] This discovery was surprising, since up to then oncogenic potential was believed to be solely due to activating mutations.

The erbA work led Martin Zenke to work on red blood cell differentiation, [11] [12] [14] focussing on the just discovered GATA transcription factors. [11] [12] He found that GATA-1 promotes red blood cell development [12] whereas GATA-2 blocks red blood cell development. [11] These findings were the first to suggest GATA-2 function in early blood cell development.

1995-today: Stem Cells and Antigen Presenting Dendritic Cells

At the beginning of the 1990s, the studies on the v-rel oncogene [13] led Martin Zenke to work on antigen presenting dendritic cells (DC), a specific immune cell, which is important for immunity and immune tolerance. DC biology was poorly understood at that time and Martin Zenke was one of the first to apply gene expression profiling with DNA microarrays for gene mining. This work led to the discovery of the Id2 transcription factor in DC development. [15] The Id2 gene data sets received accession numbers 1 and 2 (E-MEXP-1 and E-MEXP-2) of the ArrayExpress database, [21] one of the two major genomic data repositories, which now contains several million entries.

The DC work is being followed mainly in the mouse system, [22] [23] to study gene circuitries of DC development and function using RNA-Seq, ChIP-seq, ATAC-seq, chromosome conformation capture (4C) and CRISPR/Cas9 gene editing, [24] and more recently in the human system using induced pluripotent stem cells (iPS cells).

2005-today: Pluripotent Stem Cells and Disease Modeling

Hematopoietic stem cells have been Martin Zenke’s prime interest for many years and in the 2005s he broadened his interest to also include pluripotent stem cells, such as embryonic stem cells (ES cells) and the more recently discovered induced pluripotent stem cells (iPS cells). [25] [26] [27]

A particular focus is on disease and patient specific iPS cells for disease modeling and compound screening. Emphasis is put on studying hematopoietic malignancies, [27] [28] thereby building on the close collaboration with preclinical and clinical partners. This focus also includes developing animal models of diseases and laboratory automation for cell production.

Martin Zenke also worked also on technology development: Automatic DNA sequencing, [29] and gene delivery into cells. [30] [31]

Publications

Related Research Articles

<span class="mw-page-title-main">Cellular differentiation</span> Developmental biology

Cellular differentiation is the process in which a stem cell changes from one type to a differentiated one. Usually, the cell changes to a more specialized type. Differentiation happens multiple times during the development of a multicellular organism as it changes from a simple zygote to a complex system of tissues and cell types. Differentiation continues in adulthood as adult stem cells divide and create fully differentiated daughter cells during tissue repair and during normal cell turnover. Some differentiation occurs in response to antigen exposure. Differentiation dramatically changes a cell's size, shape, membrane potential, metabolic activity, and responsiveness to signals. These changes are largely due to highly controlled modifications in gene expression and are the study of epigenetics. With a few exceptions, cellular differentiation almost never involves a change in the DNA sequence itself. However, metabolic composition does get altered quite dramatically where stem cells are characterized by abundant metabolites with highly unsaturated structures whose levels decrease upon differentiation. Thus, different cells can have very different physical characteristics despite having the same genome.

<span class="mw-page-title-main">Oct-4</span> Mammalian protein found in Homo sapiens

Oct-4, also known as POU5F1, is a protein that in humans is encoded by the POU5F1 gene. Oct-4 is a homeodomain transcription factor of the POU family. It is critically involved in the self-renewal of undifferentiated embryonic stem cells. As such, it is frequently used as a marker for undifferentiated cells. Oct-4 expression must be closely regulated; too much or too little will cause differentiation of the cells.

<span class="mw-page-title-main">GATA1</span> Protein-coding gene in humans

GATA-binding factor 1 or GATA-1 is the founding member of the GATA family of transcription factors. This protein is widely expressed throughout vertebrate species. In humans and mice, it is encoded by the GATA1 and Gata1 genes, respectively. These genes are located on the X chromosome in both species.

<span class="mw-page-title-main">Monoblast</span>

Monoblasts are the committed progenitor cells that differentiated from a committed macrophage or dendritic cell precursor (MDP) in the process of hematopoiesis. They are the first developmental stage in the monocyte series leading to a macrophage. Their myeloid cell fate is induced by the concentration of cytokines they are surrounded by during development. These cytokines induce the activation of transcription factors which push completion of the monoblast's myeloid cell fate. Monoblasts are normally found in bone marrow and do not appear in the normal peripheral blood. They mature into monocytes which, in turn, develop into macrophages. They then are seen as macrophages in the normal peripheral blood and many different tissues of the body. Macrophages can produce a variety of effector molecules that initiate local, systemic inflammatory responses. These monoblast differentiated cells are equipped to fight off foreign invaders using pattern recognition receptors to detect antigen as part of the innate immune response.

<span class="mw-page-title-main">V-erbA-related gene</span> Protein-coding gene in the species Homo sapiens

V-erbA-related protein 2 (EAR-2) also known as NR2F6 is a protein that in humans is encoded by the NR2F6 gene. V-erbA-related protein 2 is a member of the nuclear receptor family of intracellular transcription factors. It is named after its similarity to v-erbA, a helper of an oncoprotein called v-erbB in avian erythroblastosis virus.

<span class="mw-page-title-main">Induced pluripotent stem cell</span> Pluripotent stem cell generated directly from a somatic cell

Induced pluripotent stem cells are a type of pluripotent stem cell that can be generated directly from a somatic cell. The iPSC technology was pioneered by Shinya Yamanaka and Kazutoshi Takahashi in Kyoto, Japan, who together showed in 2006 that the introduction of four specific genes, collectively known as Yamanaka factors, encoding transcription factors could convert somatic cells into pluripotent stem cells. Shinya Yamanaka was awarded the 2012 Nobel Prize along with Sir John Gurdon "for the discovery that mature cells can be reprogrammed to become pluripotent."

<span class="mw-page-title-main">TP63</span> Protein-coding gene in the species Homo sapiens

Tumor protein p63, typically referred to as p63, also known as transformation-related protein 63 is a protein that in humans is encoded by the TP63 gene.

<span class="mw-page-title-main">GATA2</span> Protein-coding gene in the species Homo sapiens

GATA2 or GATA-binding factor 2 is a transcription factor, i.e. a nuclear protein which regulates the expression of genes. It regulates many genes that are critical for the embryonic development, self-renewal, maintenance, and functionality of blood-forming, lympathic system-forming, and other tissue-forming stem cells. GATA2 is encoded by the GATA2 gene, a gene which often suffers germline and somatic mutations which lead to a wide range of familial and sporadic diseases, respectively. The gene and its product are targets for the treatment of these diseases.

<span class="mw-page-title-main">SOX2</span> Transcription factor gene of the SOX family

SRY -box 2, also known as SOX2, is a transcription factor that is essential for maintaining self-renewal, or pluripotency, of undifferentiated embryonic stem cells. Sox2 has a critical role in maintenance of embryonic and neural stem cells.

<span class="mw-page-title-main">SPIB</span> Protein that controls transcription of a set of genes in eukaryotes

Transcription factor Spi-B is a protein that in humans is encoded by the SPIB gene.

<span class="mw-page-title-main">ZBTB32</span> Protein-coding gene in the species Homo sapiens

Zinc finger and BTB domain-containing protein 32 is a protein that in humans is encoded by the 1960 bp ZBTB32 gene. The 52 kDa protein is a transcriptional repressor and the gene is expressed in T and B cells upon activation, but also significantly in testis cells. It is a member of the Poxviruses and Zinc-finger (POZ) and Krüppel (POK) family of proteins, and was identified in multiple screens involving either immune cell tumorigenesis or immune cell development.

<span class="mw-page-title-main">FOXD3</span> Protein-coding gene in the species Homo sapiens

Forkhead box D3 also known as FOXD3 is a forkhead protein that in humans is encoded by the FOXD3 gene.

<span class="mw-page-title-main">Cell potency</span> Ability of a cell to differentiate into other cell types

Cell potency is a cell's ability to differentiate into other cell types. The more cell types a cell can differentiate into, the greater its potency. Potency is also described as the gene activation potential within a cell, which like a continuum, begins with totipotency to designate a cell with the most differentiation potential, pluripotency, multipotency, oligopotency, and finally unipotency.

<span class="mw-page-title-main">FOXA1</span> Protein-coding gene in the species Homo sapiens

Forkhead box protein A1 (FOXA1), also known as hepatocyte nuclear factor 3-alpha (HNF-3A), is a protein that in humans is encoded by the FOXA1 gene.

In the field of molecular biology, the Mi-2/NuRDcomplex, is a group of associated proteins with both ATP-dependent chromatin remodeling and histone deacetylase activities. As of 2007, Mi-2/NuRD was the only known protein complex that couples chromatin remodeling ATPase and chromatin deacetylation enzymatic functions.

Induced stem cells (iSC) are stem cells derived from somatic, reproductive, pluripotent or other cell types by deliberate epigenetic reprogramming. They are classified as either totipotent (iTC), pluripotent (iPSC) or progenitor or unipotent – (iUSC) according to their developmental potential and degree of dedifferentiation. Progenitors are obtained by so-called direct reprogramming or directed differentiation and are also called induced somatic stem cells.

Pioneer factors are transcription factors that can directly bind condensed chromatin. They can have positive and negative effects on transcription and are important in recruiting other transcription factors and histone modification enzymes as well as controlling DNA methylation. They were first discovered in 2002 as factors capable of binding to target sites on nucleosomal DNA in compacted chromatin and endowing competency for gene activity during hepatogenesis. Pioneer factors are involved in initiating cell differentiation and activation of cell-specific genes. This property is observed in histone fold-domain containing transcription factors and other transcription factors that use zinc finger(s) for DNA binding.

ATAC-seq is a technique used in molecular biology to assess genome-wide chromatin accessibility. In 2013, the technique was first described as an alternative advanced method for MNase-seq, FAIRE-Seq and DNase-Seq. ATAC-seq is a faster and more sensitive analysis of the epigenome than DNase-seq or MNase-seq.

<span class="mw-page-title-main">Insulated neighborhood</span>

In mammalian biology, insulated neighborhoods are chromosomal loop structures formed by the physical interaction of two DNA loci bound by the transcription factor CTCF and co-occupied by cohesin. Insulated neighborhoods are thought to be structural and functional units of gene control because their integrity is important for normal gene regulation. Current evidence suggests that these structures form the mechanistic underpinnings of higher-order chromosome structures, including topologically associating domains (TADs). Insulated neighborhoods are functionally important in understanding gene regulation in normal cells and dysregulated gene expression in disease.

<span class="mw-page-title-main">Thomas Graf (biologist)</span>

Thomas Graf is a biologist at the Centre for Genomic Regulation (CRG) in Barcelona, Spain. He is a pioneer in cell reprogramming, showing that blood cells can be transdifferentiated by transcription factors. He is also known for his early work on oncogenes carried by retroviruses and oncogene cooperation in leukemia formation.

References

  1. 1 2 Zenke, M; Sauer, G (Aug 11, 1982). "Spliced and unspliced virus specific RNA sequences are associated with purified simian virus 40 chromatin". Nucleic Acids Research. 10 (15): 4543–50. doi:10.1093/nar/10.15.4543. PMC   321110 . PMID   6290985.
  2. Zenke, Martin (1982). Transkription von SV40 Chromatin (Thesis) (in German). Frankfurt, Signatur: H 90b/9811: Heidelberg, Univ., Diss., 1982. p. 118.{{cite thesis}}: CS1 maint: location (link)
  3. 1 2 Zenke, M; Grundström, T; Matthes, H; Wintzerith, M; Schatz, C; Wildeman, A; Chambon, P (February 1986). "Multiple sequence motifs are involved in SV40 enhancer function". The EMBO Journal. 5 (2): 387–97. doi:10.1002/j.1460-2075.1986.tb04224.x. PMC   1166744 . PMID   3011406.
  4. 1 2 Wildeman, A G; Zenke, M; Schatz, C; Wintzerith, M; Grundström, T; Matthes, H; Takahashi, K; Chambon, P (June 1986). "Specific protein binding to the simian virus 40 enhancer in vitro". Molecular and Cellular Biology. 6 (6): 2098–2105. doi:10.1128/mcb.6.6.2098. PMC   367750 . PMID   3023918.
  5. 1 2 Davidson, I; Fromental, C; Augereau, P; Wildeman, A; Zenke, M; Chambon, P (Oct 9–15, 1986). "Cell-type specific protein binding to the enhancer of simian virus 40 in nuclear extracts". Nature. 323 (6088): 544–8. Bibcode:1986Natur.323..544D. doi:10.1038/323544a0. PMID   3020434. S2CID   4327015.
  6. Zenke, Martin. "EMBL Alumni" . Retrieved 26 April 2012.
  7. 1 2 3 Zenke, M; Kahn, P; Disela, C; Vennström, B; Leutz, A; Keegan, K; Hayman, MJ; Choi, HR; Yew, N; Engel, JD (Jan 15, 1988). "v-erbA specifically suppresses transcription of the avian erythrocyte anion transporter (band 3) gene". Cell. 52 (1): 107–19. doi:10.1016/0092-8674(88)90535-1. PMID   2830979. S2CID   29464344.
  8. Research Institute of Molecular Pathology, former groups
  9. 1 2 3 Zenke, M; Muñoz, A; Sap, J; Vennström, B; Beug, H (Jun 15, 1990). "v-erbA oncogene activation entails the loss of hormone-dependent regulator activity of c-erbA". Cell. 6. 61 (6): 1035–49. doi:10.1016/0092-8674(90)90068-P. PMID   1972036. S2CID   36806100.
  10. 1 2 3 Disela, C; Glineur, C; Bugge, T; Sap, J; Stengl, G; Dodgson, J; Stunnenberg, H; Beug, H; Zenke, M (November 1991). "v-erbA overexpression is required to extinguish c-erbA function in erythroid cell differentiation and regulation of the erbA target gene CAII". Genes & Development. 5 (11): 2033–2047. doi: 10.1101/gad.5.11.2033 . PMID   1682217.
  11. 1 2 3 4 Briegel, K; Lim, K C; Plank, C; Beug, H; Engel, J D; Zenke, M (June 1993). "Ectopic expression of a conditional GATA-2/estrogen receptor chimera arrests erythroid differentiation in a hormone-dependent manner". Genes & Development. 7 (6): 1097–1109. doi: 10.1101/gad.7.6.1097 . PMID   8504932.
  12. 1 2 3 4 Briegel, K.; Bartunek, P.; Stengl, G.; Lim, K.C.; Beug, H.; Engel, J.D.; Zenke, M. (1 December 1996). "Regulation and function of transcription factor GATA-1 during red blood cell differentiation". Development. 122 (12): 3839–3850. doi:10.1242/dev.122.12.3839. PMID   9012505.
  13. 1 2 3 Boehmelt, G; Madruga, J; Dörfler, P; Briegel, K; Schwarz, H; Enrietto, PJ; Zenke, M (Jan 27, 1995). "Dendritic cell progenitor is transformed by a conditional v-Rel estrogen receptor fusion protein v-RelER". Cell. 80 (2): 341–52. doi: 10.1016/0092-8674(95)90417-4 . PMID   7834754.
  14. 1 2 Panzenböck, Birgit; Bartunek, Petr; Mapara, Markus Y.; Zenke, Martin (15 November 1998). "Growth and Differentiation of Human Stem Cell Factor/Erythropoietin-Dependent Erythroid Progenitor Cells In Vitro". Blood. 92 (10): 3658–3668. doi:10.1182/blood.V92.10.3658. PMID   9808559.
  15. 1 2 Hacker, C; Kirsch, RD; Ju, XS; Hieronymus, T; Gust, TC; Kuhl, C; Jorgas, T; Kurz, SM; Rose-John, S; Yokota, Y; Zenke, M (April 2003). "Transcriptional profiling identifies Id2 function in dendritic cell development". Nature Immunology. 4 (4): 380–6. doi:10.1038/ni903. PMID   12598895. S2CID   35329390.
  16. Zenke, Martin. "Dr. Martin Zenke nimmt Ruf auf C4-Professur in Aachen an". public relations department of the Max Delbrück Center for Molecular Medicine. Retrieved 26 April 2012.
  17. Zenke, Martin. "currently appointed professors RWTH 2003". RWTH Aachen. Retrieved 26 April 2012.
  18. Lewin, B. (2008). Genes IX. ISBN   978-0763752224.
  19. Weinberg, Robert A. (22 November 1991). "Tumor Suppressor Genes". Science. 254 (5035): 1138–1146. Bibcode:1991Sci...254.1138W. doi:10.1126/science.1659741. PMID   1659741.
  20. Lewin, Benjamin (January 1991). "Oncogenic conversion by regulatory changes in transcription factors". Cell. 64 (2): 303–312. doi:10.1016/0092-8674(91)90640-k. PMID   1988149. S2CID   26966778.
  21. "ArrayExpress < EMBL-EBI". www.ebi.ac.uk.
  22. Seré, Kristin; Baek, Jea-Hyun; Ober-Blöbaum, Julia; Müller-Newen, Gerhard; Tacke, Frank; Yokota, Yoshifumi; Zenke, Martin; Hieronymus, Thomas (November 2012). "Two Distinct Types of Langerhans Cells Populate the Skin during Steady State and Inflammation". Immunity. 37 (5): 905–916. doi: 10.1016/j.immuni.2012.07.019 . PMID   23159228.
  23. Romani, Nikolaus; Tripp, Christoph; Stoitzner, Patrizia (November 2012). "Langerhans Cells Come in Waves". Immunity. 37 (5): 766–768. doi:10.1016/j.immuni.2012.10.013. PMC   4285563 . PMID   23159223.
  24. Lin, Qiong; Chauvistré, Heike; Costa, Ivan G.; Gusmao, Eduardo G.; Mitzka, Saskia; Hänzelmann, Sonja; Baying, Bianka; Klisch, Theresa; Moriggl, Richard; Hennuy, Benoit; Smeets, Hubert; Hoffmann, Kurt; Benes, Vladimir; Seré, Kristin; Zenke, Martin (17 October 2015). "Epigenetic program and transcription factor circuitry of dendritic cell development". Nucleic Acids Research. 43 (20): 9680–9693. doi:10.1093/nar/gkv1056. PMC   4787753 . PMID   26476451.
  25. Ruau, David; Ensenat‐Waser, Roberto; Dinger, Timo C.; Vallabhapurapu, Duttu S.; Rolletschek, Alexandra; Hacker, Christine; Hieronymus, Thomas; Wobus, Anna M.; Müller, Albrecht M.; Zenke, Martin (April 2008). "Pluripotency Associated Genes Are Reactivated by Chromatin‐Modifying Agents in Neurosphere Cells". Stem Cells. 26 (4): 920–926. doi: 10.1634/stemcells.2007-0649 . PMID   18203677. S2CID   41497590.
  26. Kim, Jeong Beom; Zaehres, Holm; Wu, Guangming; Gentile, Luca; Ko, Kinarm; Sebastiano, Vittorio; Araúzo-Bravo, Marcos J.; Ruau, David; Han, Dong Wook; Zenke, Martin; Schöler, Hans R. (July 2008). "Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors". Nature. 454 (7204): 646–650. Bibcode:2008Natur.454..646K. doi:10.1038/nature07061. PMID   18594515. S2CID   4318637.
  27. 1 2 Toledo, Marcelo A. S.; Gatz, Malrun; Sontag, Stephanie; Gleixner, Karoline V.; Eisenwort, Gregor; Feldberg, Kristina; Hamouda, Ahmed E. I.; Kluge, Frederick; Guareschi, Riccardo; Rossetti, Giulia; Sechi, Antonio S.; Dufva, Olli M. J.; Mustjoki, Satu M.; Maurer, Angela; Schüler, Herdit M.; Goetzke, Roman; Braunschweig, Till; Kaiser, Anne; Panse, Jens; Jawhar, Mohamad; Reiter, Andreas; Hilberg, Frank; Ettmayer, Peter; Wagner, Wolfgang; Koschmieder, Steffen; Brümmendorf, Tim H.; Valent, Peter; Chatain, Nicolas; Zenke, Martin (15 April 2021). "Nintedanib targets KIT D816V neoplastic cells derived from induced pluripotent stem cells of systemic mastocytosis". Blood. 137 (15): 2070–2084. doi: 10.1182/blood.2019004509 . PMID   33512435. S2CID   231758973.
  28. Dorrance, Adrienne (15 April 2021). ""Mast"ering drug discovery with iPSCs". Blood. 137 (15): 1993–1994. doi: 10.1182/blood.2020010456 . PMID   33856443. S2CID   233243904.
  29. Ansorge, Wilhelm; Sproat, Brian; Stegemann, Josef; Schwager, Christian; Zenke, Martin (1987). "Automated DNA sequencing: ultrasensitive detection of fluorescent bands during electrophoresis". Nucleic Acids Research. 15 (11): 4593–4602. doi:10.1093/nar/15.11.4593. PMC   340882 . PMID   3588303.
  30. Zenke, M.; Steinlein, P.; Wagner, E.; Cotten, M.; Beug, H.; Birnstiel, M. L. (1 May 1990). "Receptor-mediated endocytosis of transferrin-polycation conjugates: an efficient way to introduce DNA into hematopoietic cells". Proceedings of the National Academy of Sciences. 87 (10): 3655–3659. Bibcode:1990PNAS...87.3655Z. doi: 10.1073/pnas.87.10.3655 . PMC   53961 . PMID   2339110.
  31. Diebold, Sandra S.; Kursa, Margaretha; Wagner, Ernst; Cotten, Matt; Zenke, Martin (July 1999). "Mannose Polyethylenimine Conjugates for Targeted DNA Delivery into Dendritic Cells". Journal of Biological Chemistry. 274 (27): 19087–19094. doi: 10.1074/jbc.274.27.19087 . PMID   10383411.