Volume 84, Issue 2, Pages (January 1996)

Slides:



Advertisements
Similar presentations
Heterogeneity of Abnormal RUNX1 Leading to Clinicopathological Variations in Childhood B-Lymphoblastic Leukemia Xiayuan Liang, MD Department of Pathology.
Advertisements

Date of download: 6/22/2016 Copyright © 2016 American Medical Association. All rights reserved. From: Identification of a Novel TP53 Cancer Susceptibility.
Tumor spectrum analysis in p53-mutant mice
Volume 15, Issue 2, Pages (August 2001)
by Sergei Merkulov, Wan-Ming Zhang, Anton A. Komar, Alvin H
Vav‐1 gene‐targeting strategy.
Volume 93, Issue 3, Pages (May 1998)
Role of AP1/NFE2 binding sites in endogenous α-globin gene transcription by Melanie R. Loyd, Yasuhiro Okamoto, Mindy S. Randall, and Paul A. Ney Blood.
Volume 88, Issue 1, Pages (January 1997)
Volume 86, Issue 1, Pages (July 1996)
Globin Gene Expression Is Reprogrammed in Chimeras Generated by Injecting Adult Hematopoietic Stem Cells into Mouse Blastocysts  Hartmut Geiger, Stefanie.
Volume 93, Issue 5, Pages (May 1998)
Volume 6, Issue 4, Pages (April 1997)
Growth Disturbance in Fetal Liver Hematopoiesis of Mll-Mutant Mice
Volume 5, Issue 3, Pages (September 1996)
Volume 9, Issue 20, Pages S1-S2 (October 1999)
Analysis of the Human Ferrochelatase Promoter in Transgenic Mice
Brca1 Controls Homology-Directed DNA Repair
Sox17 Dependence Distinguishes the Transcriptional Regulation of Fetal from Adult Hematopoietic Stem Cells  Injune Kim, Thomas L. Saunders, Sean J. Morrison 
Volume 2, Issue 4, Pages (October 1998)
Volume 87, Issue 4, Pages (November 1996)
Volume 7, Issue 3, Pages (March 2001)
Volume 92, Issue 6, Pages (March 1998)
Figure 1. Generation of the S250F Aadc mutant mice
Volume 84, Issue 4, Pages (February 1996)
The Death Domain Kinase RIP Mediates the TNF-Induced NF-κB Signal
Volume 9, Issue 4, Pages (October 1998)
The Mouse Spo11 Gene Is Required for Meiotic Chromosome Synapsis
Volume 1, Issue 1, Pages (February 2002)
Volume 13, Issue 2, Pages (August 2000)
Volume 9, Issue 3, Pages (September 1998)
Requirement for p38α in Erythropoietin Expression
SOCS3 Is Essential in the Regulation of Fetal Liver Erythropoiesis
Chuxia Deng, Anthony Wynshaw-Boris, Fen Zhou, Ann Kuo, Philip Leder 
Georges Lacaud, Leif Carlsson, Gordon Keller  Immunity 
In Vivo Ablation of Surface Immunoglobulin on Mature B Cells by Inducible Gene Targeting Results in Rapid Cell Death  Kong-Peng Lam, Ralf Kühn, Klaus.
Volume 3, Issue 5, Pages (May 2003)
Volume 18, Issue 2, Pages (February 2003)
Volume 10, Issue 8, Pages (April 2000)
Yang Xu, Genhong Cheng, David Baltimore  Immunity 
Volume 19, Issue 1, Pages (July 1997)
Volume 105, Issue 5, Pages (June 2001)
Both E12 and E47 Allow Commitment to the B Cell Lineage
Small Evolutionarily Conserved RNA, Resembling C/D Box Small Nucleolar RNA, Is Transcribed from PWCR1, a Novel Imprinted Gene in the Prader-Willi Deletion.
Deletion of the Scl +19 enhancer increases the blood stem cell compartment without affecting the formation of mature blood lineages  Dominik Spensberger,
Volume 16, Issue 5, Pages (May 2002)
Volume 10, Issue 6, Pages (December 2002)
A Prematurely Expressed Igκ Transgene, but Not a VκJκ Gene Segment Targeted into the Igκ Locus, Can Rescue B Cell Development in λ5-Deficient Mice  Roberta.
Role of the INK4a Locus in Tumor Suppression and Cell Mortality
Volume 85, Issue 6, Pages (June 1996)
Volume 2, Issue 4, Pages (October 1998)
Targeted disruption of the Nijmegen breakage syndrome gene NBS1 leads to early embryonic lethality in mice  Jie Zhu, Simone Petersen, Lino Tessarollo,
Targeted Mutagenesis of the Endogenous Mouse Mis Gene Promoter
Β-globin Gene Switching and DNase I Sensitivity of the Endogenous β-globin Locus in Mice Do Not Require the Locus Control Region  M.A Bender, Michael.
Skeletal and CNS Defects in Presenilin-1-Deficient Mice
APOE Gene Targeting (A) Schematic representation of the endogenous APOE locus, the gene targeting vector and the targeted APOE locus. The exons of the.
Multiple Developmental Stage–Specific Enhancers Regulate CD8 Expression in Developing Thymocytes and in Thymus-Independent T Cells  Wilfried Ellmeier,
Volume 8, Issue 1, Pages (January 1998)
Joan E Durbin, Renée Hackenmiller, M.Celeste Simon, David E Levy  Cell 
Volume 10, Issue 4, Pages (April 1999)
Fig. 4. Targeted disruption of STK35 transcripts in mouse.
Securin is not required for cellular viability, but is required for normal growth of mouse embryonic fibroblasts  Junjie Mei, Xingxu Huang, Pumin Zhang 
Volume 9, Issue 3, Pages (September 1998)
Volume 10, Issue 8, Pages (April 2000)
The Death Domain Kinase RIP Mediates the TNF-Induced NF-κB Signal
Volume 6, Issue 4, Pages (April 1997)
Volume 86, Issue 1, Pages (July 1996)
Volume 4, Issue 2, Pages (February 1996)
Mutation of the Ca2+ Channel β Subunit Gene Cchb4 Is Associated with Ataxia and Seizures in the Lethargic (lh) Mouse  Daniel L Burgess, Julie M Jones,
Volume 21, Issue 6, Pages (December 2004)
Presentation transcript:

Volume 84, Issue 2, Pages 321-330 (January 1996) AML1, the Target of Multiple Chromosomal Translocations in Human Leukemia, Is Essential for Normal Fetal Liver Hematopoiesis  Tsukasa Okuda, Jan van Deursen, Scott W Hiebert, Gerard Grosveld, James R Downing  Cell  Volume 84, Issue 2, Pages 321-330 (January 1996) DOI: 10.1016/S0092-8674(00)80986-1

Figure 1 Targeted Disruption of Murine AML1 Gene by Homologous Recombination (A) Schematic representation of the AML1 cDNA and partial restriction map of the AML1 genomic locus with the targeting construct depicted below. The DNA-binding RHD is shown, as are SacI, SpeI, and XbaI (X) restriction sites. The arrows indicate the direction of transcription of the hygrR (hygr) and TK cassettes. The positions of AML1 exons 3 and 4 are shown as boxes, and their coding sequences are closed. Wild-type (14 kb) and targeted alleles (16 kb) could be distinguished by Southern blots of XbaI-digested DNA, hybridized with either the indicated 5′ or 3′ probes. (B) Southern blot analysis of ES cell clones and E11.5 embryos from an AML1+/− intercross. ES cell clones with homologous targeting of one AML1 allele contain a 16 kb restriction fragment in addition to the 14 kb fragment derived from the wild-type allele. Homozygous mutant embryos contain only the 16 kb restriction fragment. Cell 1996 84, 321-330DOI: (10.1016/S0092-8674(00)80986-1)

Figure 2 RT–PCR Analysis of mRNA from E11.5 Embryos Total embryo mRNA was isolated and amplified with AML1-specific primer that bracket the site of targeting in exon 4. RT–PCR amplification was also performed for the ubiquitously expressed HPRT mRNA as a control for the presence of amplifiable RNA. Cell 1996 84, 321-330DOI: (10.1016/S0092-8674(00)80986-1)

Figure 3 Phenotypes of E12.5 Control and AML1−/− Embryos (A) Appearance of a mutant mouse embryos (−/−) compared with their control littermate (+/−). The mutant embryo is equal in size compared with the control and appears to have developed normally. The mutant embryo is distinguished, however, by the presence of hemorrhage within the ventricle of the brain and vertebral canal. (B) Sagittal sections of the brains show a comparable level of development in the control and mutant embryos; however, in the AML1−/− embryo, hemorrhages can be seen within the ganglia of the cranial nerves with extension into the ventricles. (C) Sagittal section through the thoracic cavity demonstrates a hemorrhage within the pericardial space of the AML1−/− embryo. Cell 1996 84, 321-330DOI: (10.1016/S0092-8674(00)80986-1)

Figure 4 Histologic Analysis of the Hematopoietic System of a Control and Mutant Embryos at E11.5–E12.5. (A) Sections of livers from an E11.5 control (+/−) embryo contained numerous dark-staining hematopoietic elements. In contrast, sections of the AML1−/− mutant (−/−) embryo revealed the complete absence of hematopoietic precursors, with only rare primitive nucleated erythrocytes seen within vessels and sinusoids. (B) Touch preparation of liver from a control embryo contained numerous immature to midmature erythroid cells, along with scattered macrophages, immature myeloid cells, and rare megakaryocytes (data not shown). In contrast, liver touch preparation from AML1-deficient mice contained primarily hepatocytes, with only scattered primitive nucleated erythrocytes seen. (C) Cytocentrifuge preparations of peripheral blood from a viable E12.5 control embryo contained primitive nucleated erythrocytes, rare nonnucleated definitive erythrocytes, immature myeloid elements, and numerous platelets. Cytocentrifuge preparations from peripheral blood of a viable E12.5 AML1−/− embryo consisted exclusively of nucleated primitive erythrocytes with a complete absence of identifiable platelets. Cell 1996 84, 321-330DOI: (10.1016/S0092-8674(00)80986-1)

Figure 5 Embryoid Bodies Derived from Control and AML1-Deficient ES Cells (A) Hemoglobinized embryoid bodies were observed in cultures of both AML1+/− and AML1−/− ES cells. (B) Cytocentrifuge preparations of single embryoid bodies from control (+/−) and mutant (−/−) derived cultures. Cell 1996 84, 321-330DOI: (10.1016/S0092-8674(00)80986-1)

Figure 6 Contribution of AML1+/− and AML1−/− ES cells to Tissues in Chimeric Animals GPI analysis of chimeric animals derived from three AML1+/− (A–C) and three AML1−/− (D–F) ES cell clones. ES cells contain the GPI-1A isoform, whereas the host contains the GPI-1B isoform. Results from the following tissues are illustrated: kidney (K), thymus (T), liver (L), spleen (S), bone marrow (BM), peripheral blood (B), and skeletal muscle (SM). WT, wild type. Cell 1996 84, 321-330DOI: (10.1016/S0092-8674(00)80986-1)