1
|
Pulina MV, Sahr KE, Nowotschin S, Baron MH, Hadjantonakis AK. A conditional mutant allele for analysis of Mixl1 function in the mouse. Genesis 2014; 52:417-23. [PMID: 24596343 DOI: 10.1002/dvg.22768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 02/25/2014] [Accepted: 02/27/2014] [Indexed: 11/10/2022]
Abstract
Mixl1 is the only member of the Mix/Bix homeobox gene family identified in mammals. During mouse embryogenesis, Mixl1 is first expressed at embryonic day (E)5.5 in cells of the visceral endoderm (VE). At the time of gastrulation, Mixl1 expression is detected in the vicinity of the primitive streak. Mixl1 is expressed in cells located within the primitive streak, in nascent mesoderm cells exiting the primitive streak, and in posterior VE overlying the primitive streak. Genetic ablation of Mixl1 in mice has revealed its crucial role in mesoderm and endoderm cell specification and tissue morphogenesis during early embryonic development. However, the early lethality of the constitutive Mixl1(-/-) mutant precludes the study of its role at later stages of embryogenesis and in adult mice. To circumvent this limitation, we have generated a conditional Mixl1 allele (Mixl1(cKO) that permits temporal as well as spatial control of gene ablation. Animals homozygous for the Mixl1(cKO) conditional allele were viable and fertile. Mixl1(KO/KO) embryos generated by crossing of Mixl1(cKO/cKO) mice with Sox2-Cre or EIIa-Cre transgenic mice were embryonic lethal at early somite stages. By contrast to wild-type embryos, Mixl1(KO/KO) embryos contained no detectable Mixl1, validating the Mixl1(cKO) as a protein null after Cre-mediated excision. Mixl1(KO/KO) embryos resembled the previously reported Mixl1(-/-) mutant phenotype. Therefore, the Mixl1 cKO allele provides a tool for investigating the temporal and tissue-specific requirements for Mixl1 in the mouse.
Collapse
Affiliation(s)
- Maria V Pulina
- Developmental Biology Program, Sloan-Kettering Institute, New York, New York
| | | | | | | | | |
Collapse
|
2
|
Wolfe AD, Downs KM. Mixl1 localizes to putative axial stem cell reservoirs and their posterior descendants in the mouse embryo. Gene Expr Patterns 2014; 15:8-20. [PMID: 24632399 DOI: 10.1016/j.gep.2014.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 02/13/2014] [Accepted: 02/17/2014] [Indexed: 01/22/2023]
Abstract
Mixl1 is thought to play important roles in formation of mesoderm and endoderm. Previously, Mixl1 expression was reported in the posterior primitive streak and allantois, but the precise spatiotemporal whereabouts of Mixl1 protein throughout gastrulation have not been elucidated. To localize Mixl1 protein, immunohistochemistry was carried out at 2-4 h intervals on mouse gastrulae between primitive streak and 16-somite pair (s) stages (~E6.5-9.5). Mixl1 localized to the entire primitive streak early in gastrulation. However, by headfold stages (~E7.75-8.0), Mixl1 diminished within the mid-streak but remained concentrated at either end of the streak, and localized throughout midline posterior visceral endoderm. At the streak's anterior end, Mixl1 was confined to the posterior crown cells of Hensen's node, which contribute to dorsal hindgut endoderm, and the posterior notochord. In the posterior streak, Mixl1 localized to the Allantoic Core Domain (ACD), which is the source of most of the allantois and contributes to the posterior embryonic-extraembryonic interface. In addition, Mix1 co-localized with the early hematopoietic marker, Runx1, in the allantois and visceral yolk sac blood islands. During hindgut invagination (4-16s, ~E8.5-9.5), Mixl1 localized to the hindgut lip, becoming concentrated within the midline anastomosis of the splanchnopleure, which appears to create the ventral component of the hindgut and omphalomesenteric artery. Surrounding the distal hindgut, Mixl1 identified midline cells within tailbud mesoderm. Mixl1 was also found in the posterior notochord. These findings provide a critical systematic, and tissue-level understanding of embryonic Mixl1 localization, and support its role in regulation of crucial posterior axial mesendodermal stem cell niches during embryogenesis.
Collapse
Affiliation(s)
- Adam D Wolfe
- Department of Pediatrics, Division of Pediatric Hematology, Oncology & Bone Marrow Transplant, University of Wisconsin-Madison School of Medicine and Public Health, 1111 Highland Avenue, 4105 WIMR, Madison, WI 53705, United States
| | - Karen M Downs
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, United States
| |
Collapse
|
3
|
Pereira LA, Wong MS, Mei Lim S, Stanley EG, Elefanty AG. The Mix family of homeobox genes—Key regulators of mesendoderm formation during vertebrate development. Dev Biol 2012; 367:163-77. [DOI: 10.1016/j.ydbio.2012.04.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 04/24/2012] [Accepted: 04/30/2012] [Indexed: 10/28/2022]
|
4
|
Pereira LA, Wong MS, Lim SM, Sides A, Stanley EG, Elefanty AG. Brachyury and related Tbx proteins interact with the Mixl1 homeodomain protein and negatively regulate Mixl1 transcriptional activity. PLoS One 2011; 6:e28394. [PMID: 22164283 PMCID: PMC3229578 DOI: 10.1371/journal.pone.0028394] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 11/07/2011] [Indexed: 02/07/2023] Open
Abstract
Mixl1 is a homeodomain transcription factor required for mesoderm and endoderm patterning during mammalian embryogenesis. Despite its crucial function in development, co-factors that modulate the activity of Mixl1 remain poorly defined. Here we report that Mixl1 interacts physically and functionally with the T-box protein Brachyury and related members of the T-box family of transcription factors. Transcriptional and protein analyses demonstrated overlapping expression of Mixl1 and Brachyury during embryonic stem cell differentiation. In vitro protein interaction studies showed that the Mixl1 with Brachyury associated via their DNA-binding domains and gel shift assays revealed that the Brachyury T-box domain bound to Mixl1-DNA complexes. Furthermore, luciferase reporter experiments indicated that association of Mixl1 with Brachyury and related T-box factors inhibited the transactivating potential of Mixl1 on the Gsc and Pdgfrα promoters. Our results indicate that the activity of Mixl1 can be modulated by protein-protein interactions and that T-box factors can function as negative regulators of Mixl1 activity.
Collapse
Affiliation(s)
- Lloyd A. Pereira
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre and the Pathology Department, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael S. Wong
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | - Sue Mei Lim
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | - Alexandra Sides
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | - Edouard G. Stanley
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
- * E-mail: (AGE); (EGS)
| | - Andrew G. Elefanty
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
- * E-mail: (AGE); (EGS)
| |
Collapse
|
5
|
Pereira LA, Wong MS, Mossman AK, Sourris K, Janes ME, Knezevic K, Hirst CE, Lim SM, Pimanda JE, Stanley EG, Elefanty AG. Pdgfrα and Flk1 are direct target genes of Mixl1 in differentiating embryonic stem cells. Stem Cell Res 2011; 8:165-79. [PMID: 22265737 DOI: 10.1016/j.scr.2011.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 09/28/2011] [Indexed: 11/25/2022] Open
Abstract
The Mixl1 homeodomain protein plays a key role in mesendoderm patterning during embryogenesis, but its target genes remain to be identified. We compared gene expression in differentiating heterozygous Mixl1(GFP/w) and homozygous null Mixl1(GFP/Hygro) mouse embryonic stem cells to identify potential downstream transcriptional targets of Mixl1. Candidate Mixl1 regulated genes whose expression was reduced in GFP+ cells isolated from differentiating Mixl1(GFP/Hygro) embryoid bodies included Pdgfrα and Flk1. Mixl1 bound to ATTA sequences located in the Pdgfrα and Flk1 promoters and chromatin immunoprecipitation assays confirmed Mixl1 occupancy of these promoters in vivo. Furthermore, Mixl1 transactivated the Pdgfrα and Flk1 promoters through ATTA sequences in a DNA binding dependent manner. These data support the hypothesis that Mixl1 directly regulates Pdgfrα and Flk1 gene expression and strengthens the position of Mixl1 as a key regulator of mesendoderm development during mammalian gastrulation.
Collapse
Affiliation(s)
- Lloyd A Pereira
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Zon LI. Derivation of adult stem cells during embryogenesis. HARVEY LECTURES 2010; 102:117-132. [PMID: 20166566 DOI: 10.1002/9780470593042.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Affiliation(s)
- Leonard I Zon
- Children's Hospital Boston, Howard Hughes Medical Institute, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Zhang H, Fraser ST, Papazoglu C, Hoatlin ME, Baron MH. Transcriptional activation by the Mixl1 homeodomain protein in differentiating mouse embryonic stem cells. Stem Cells 2010; 27:2884-95. [PMID: 19711456 DOI: 10.1002/stem.203] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Members of the Mix/Bix family of paired class homeobox genes play important roles in the development of vertebrate mesoderm and endoderm. The single Mix/Bix family member identified in the mouse, Mix-like 1 (Mixl1), is required for mesendoderm patterning during gastrulation and promotes mesoderm formation and hematopoiesis in embryonic stem cell (ESC)-derived embryoid bodies. Despite its crucial functions the transcriptional activity and targets of Mixl1 have not been well described. To investigate the molecular mechanisms of Mixl1-mediated transcriptional regulation, we have characterized the DNA-binding specificity and transcriptional properties of this homeodomain protein in differentiating ESCs. Mixl1 binds preferentially as a dimer to an 11-base pair (bp) Mixl1 binding sequence (MBS) that contains two inverted repeats separated by a 3-bp spacer. The MBS mediates transcriptional activation by Mixl1 in both NIH 3T3 cells and in a new application of an inducible ESC differentiation system. Consistent with our previous observation that early induction of Mixl1 expression in ESCs results in premature activation of Goosecoid (Gsc), we have found that Mixl1 occupies two variant MBSs within and activates transcription from the Gsc promoter in vitro and in vivo. These results strongly suggest that Gsc is a direct target gene of Mixl1 during embryogenesis. STEM CELLS 2009;27:2884-2895.
Collapse
Affiliation(s)
- Hailan Zhang
- Departments of MedicineMount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | |
Collapse
|
8
|
Liu L, Tang XH, Scognamiglio T, Gudas LJ. Oral carcinogenesis induced by 4-nitroquinoline 1-oxide in lecithin:retinol acyltransferase gene knockout mice. J Nutr Biochem 2009; 21:975-82. [PMID: 19954945 DOI: 10.1016/j.jnutbio.2009.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 07/13/2009] [Accepted: 07/31/2009] [Indexed: 01/10/2023]
Abstract
Lecithin:retinol acyltransferase (LRAT) regulates retinol (vitamin A) metabolism by esterifying retinol. LRAT expression is decreased in cultured human squamous cell carcinoma cells of the head and neck relative to normal epithelial cells. We investigated whether the carcinogen 4-nitroquinoline 1-oxide (4-NQO) induced a higher incidence of oral cancer in LRAT knockout (LRAT(-/-)) than in wild-type (Wt) mice. We also investigated retinol deprivation during 4-NQO treatment in LRAT(-/-) mice as a model for rapid retinol deficiency. We observed higher levels of secreted frizzled-related protein (Sfrp) 2, an inhibitor of WNT signaling, in tongue tumors in LRAT(-/-) versus Wt. LRAT(-/-) embryonic stem cells also expressed higher Sfrp2 transcripts, indicating an interaction between retinol and WNT signaling. Cox-2, Cyclin D1, p21, Trop2 and RARβ2 were not differentially expressed in Wt versus LRAT(-/-) tongue tumors. Wt and LRAT(-/-) mice fed a retinol-sufficient diet showed the same oral tumor incidence after 4-NQO treatment. In contrast, tongue tumors developed in 60% of Wt mice and in 100% of LRAT(-/-) mice fed a retinol-deficient diet during 4-NQO treatment (P=.22); moreover, the bromodeoxyuridine labeling index was 21.0 ± 2.4% in LRAT(-/-) normal tongue epithelium as compared to 9.9 ± 0.8% in Wt normal tongue epithelium (P<.001). Thus, partial retinol deficiency during carcinogen treatment (achieved in LRAT(-/-)) resulted in more proliferating cells in tongue epithelia from LRAT(-/-) mice and, ultimately, a greater probability of carcinogenesis.
Collapse
Affiliation(s)
- Limin Liu
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | | | | | | |
Collapse
|
9
|
Zhu H, Doherty JR, Kuliyev E, Mead PE. CDK9/cyclin complexes modulate endoderm induction by direct interaction with Mix.3/mixer. Dev Dyn 2009; 238:1346-57. [PMID: 19347956 DOI: 10.1002/dvdy.21920] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Mix-related homeodomain proteins are involved in endoderm formation in the early vertebrate embryo. We used a yeast two-hybrid screen to identify proteins that interact with Mix.3/mixer to regulate endoderm induction. We demonstrate that cyclin-dependent kinase 9 (CDK9) interacts with the carboxyl terminal domain of Mix.3. CDK9 is the catalytic subunit of the PTEF-b transcription elongation complex that phosphorylates the C-terminal domain of RNA polymerase II to promote efficient elongation of nascent transcripts. Using whole embryo transcription reporter and animal pole explant assays, we show that Mix.3 activity is regulated by CDK9/cyclin complexes. Co-expression of cyclin T2 and cyclin K had different effects on Mix.3 transcriptional activity and endoderm induction. Our data suggest that binding of CDK9, and the recruitment of different cyclin partners, can modulate the endoderm-inducing activity of Mix.3 during embryonic development. Developmental Dynamics 238:1346-1357, 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Haiqing Zhu
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | |
Collapse
|
10
|
Lim SM, Pereira L, Wong MS, Hirst CE, Van Vranken BE, Pick M, Trounson A, Elefanty AG, Stanley EG. Enforced expression of Mixl1 during mouse ES cell differentiation suppresses hematopoietic mesoderm and promotes endoderm formation. Stem Cells 2009; 27:363-74. [PMID: 19038793 DOI: 10.1634/stemcells.2008-1008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The Mixl1 gene encodes a homeodomain transcription factor that is required for normal mesoderm and endoderm development in the mouse. We have examined the consequences of enforced Mixl1 expression during mouse embryonic stem cell (ESC) differentiation. We show that three independently derived ESC lines constitutively expressing Mixl1 (Mixl1(C) ESCs) differentiate into embryoid bodies (EBs) containing a higher proportion of E-cadherin (E-Cad)(+) cells. Our analysis also shows that this differentiation occurs at the expense of hematopoietic mesoderm differentiation, with Mixl1(C) ESCs expressing only low levels of Flk1 and failing to develop hemoglobinized cells. Immunohistochemistry and immunofluorescence studies revealed that Mixl1(C) EBs have extensive areas containing cells with an epithelial morphology that express E-Cad, FoxA2, and Sox17, consistent with enhanced endoderm formation. Luciferase reporter transfection experiments indicate that Mixl1 can transactivate the Gsc, Sox17, and E-Cad promoters, supporting the hypothesis that Mixl1 has a direct role in definitive endoderm formation. Taken together, these studies suggest that high levels of Mixl1 preferentially allocate cells to the endoderm during ESC differentiation.
Collapse
Affiliation(s)
- Sue Mei Lim
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Middleton AM, King JR, Loose M. Bistability in a model of mesoderm and anterior mesendoderm specification in Xenopus laevis. J Theor Biol 2009; 260:41-55. [PMID: 19490918 DOI: 10.1016/j.jtbi.2009.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 04/10/2009] [Accepted: 05/16/2009] [Indexed: 11/20/2022]
Abstract
In this paper we develop a model of mesendoderm specification in Xenopus laevis based on an existing gene regulation network. The mesendoderm is a population of cells that may contribute to either the mesoderm or endoderm. The model that we develop encompasses the time evolution of transcription factor concentrations in a single cell and is shown to have stable steady states that correspond to mesoderm and anterior mesendodermal cell types, but not endoderm (except in cells where Goosecoid expression is inhibited). Both in vitro and in vivo versions of the model are developed and analysed, the former indicating how cell fate is determined in large part by the concentration of Activin administered to a cell, with the model results comparing favourably with current quantitative experimental data. A numerical investigation of the in vivo model suggests that cell fate is determined largely by a VegT and beta-Catenin pre-pattern, subsequently being reinforced by Nodal. We argue that this sensitivity of the model to a VegT and beta-Catenin pre-pattern indicates that a key VegT self-limiting mechanism (for which there is experimental evidence) is absent from the model. Furthermore, we find that the lack of a steady state corresponding to endoderm is entirely consistent with current in vivo data, and that the in vivo model corresponds to mesendoderm specification on the dorsal, but not the ventral, side of the embryo.
Collapse
|
12
|
ten Berge D, Koole W, Fuerer C, Fish M, Eroglu E, Nusse R. Wnt signaling mediates self-organization and axis formation in embryoid bodies. Cell Stem Cell 2009; 3:508-18. [PMID: 18983966 DOI: 10.1016/j.stem.2008.09.013] [Citation(s) in RCA: 366] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2007] [Revised: 08/25/2008] [Accepted: 09/18/2008] [Indexed: 12/18/2022]
Abstract
Embryonic stem cells (ESCs) form descendants of all three germ layers when differentiated as aggregates, termed embryoid bodies. In vivo, differentiation of cells depends on signals and morphogen gradients that provide instructive and positional cues, but do such gradients exist in embryoid bodies? We report here the establishment of anteroposterior polarity and the formation of a primitive streak-like region in the embryoid body, dependent on local activation of the Wnt pathway. In this region, cells undergo an epithelial-to-mesenchymal transition and differentiate into mesendodermal progenitors. Exogenous Wnt3a protein posteriorizes the embryoid body, resulting in predominantly mesendodermal differentiation. Conversely, inhibiting Wnt signaling promotes anterior character and results in neurectodermal differentiation. The activation of Wnt signaling and primitive streak formation requires external signals but is self-reinforcing after initiation. Our findings show that the Wnt pathway mediates the local execution of a gastrulation-like process in the embryoid body, which displays an unexpected degree of self-organization.
Collapse
Affiliation(s)
- Derk ten Berge
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Martinez-Ceballos E, Gudas LJ. Hoxa1 is required for the retinoic acid-induced differentiation of embryonic stem cells into neurons. J Neurosci Res 2008; 86:2809-19. [PMID: 18512762 DOI: 10.1002/jnr.21729] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability of embryonic stem (ES) cells to differentiate into different cell fates has been extensively evaluated, and several protocols exist for the generation of various types of cells from mouse and human ES cells. We used a differentiation protocol that involves embryoid body formation and all-trans-retinoic acid (RA, 5 microM) treatment (EB/5 microM RA) to test the ability of Hoxa1 null ES cells to adopt a neuronal fate. Hoxa1(-/-) ES cells, when treated in this EB/5 microM RA protocol, failed to differentiate along a neural lineage; Hoxa1(-/-) ES cells express severalfold lower levels of many neuronal differentiation markers, including nestin, beta-tubulin III, and MAP2, and conversely, higher levels of endodermal differentiation markers (i.e., Sox17, Col4a1) than wild type (Wt) cells. Reintroduction of exogenous Hoxa1, under the control of the metallothionein I promoter, into Hoxa1(-/-) ES cells restored their capacity to generate neurons. Moreover, overexpression of Sox17, a gene that regulates endodermal differentiation, in Wt ES cells resulted in endodermal differentiation and in a complete abolition of beta-tubulin III expression. Thus, Hoxa1 activity is essential for the neuronal differentiation of ES cells in the presence of all-trans-RA, and Hoxa1 may promote neural differentiation by inhibiting Sox17 expression. Pharmacological manipulation of Hoxa1 levels may provide a method for promoting neuronal differentiation for therapeutic uses. Furthermore, because mutations in the Hoxa1 gene can cause autism spectrum disorder in humans, these data also provide important mechanistic insights into the early developmental processes that may result in this disorder.
Collapse
|
14
|
Izumi N, Era T, Akimaru H, Yasunaga M, Nishikawa SI. Dissecting the molecular hierarchy for mesendoderm differentiation through a combination of embryonic stem cell culture and RNA interference. Stem Cells 2007; 25:1664-74. [PMID: 17446562 DOI: 10.1634/stemcells.2006-0681] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although there is a criticism that embryonic stem (ES) cell differentiation does not always reflect the differentiation process involved in mouse development, it is a suitable model system to dissect the specific differentiation pathway. We established the culture conditions that selectively differentiated mouse ES cells into three germ layers containing mesendoderm, definitive endoderm (DE), visceral endoderm (VE), mesoderm, and neuroectoderm. However, the molecular mechanisms of differentiation under each specific condition still remain unclear. Here, in combination with the RNA interference-mediated gene knockdown (KD) method, we show that Eomesodermin (Eomes), Mixl1, Brachyury (T), and GATA6 are major molecular determinants in the differentiation of mesendoderm, DE, VE, and mesoderm. Eomes plays a pivotal role in an early stage of mesendoderm differentiation, whereas Mixl1 does the same in the later stage where mesendoderm differentiates into DE. Further analyses of quantitative reverse transcription polymerase chain reaction and overexpression of Mixl1 demonstrated that Mixl1 is genetically a downstream molecule of Eomes. In addition, both Eomes and Mixl1 act as negative regulators of T expression. This strategy also reveals that Eomes and T play cell-autonomous roles in platelet-derived growth factor receptor alpha (PDGFRalpha)+ vascular endothelial growth factor receptor 2 (VEGFR2)+ and PDGFRalpha+ mesoderm generations, respectively. Our results obtained from this study are fully consistent with previous knockout studies of those genes. The present study, therefore, demonstrates that the major molecular mechanism underlying in vitro ES cell differentiation largely recapitulates that in actual embryogenesis, and the combination of our culture system and RNAi-mediated gene KD is an useful tool to elucidate the molecular hierarchy in in vitro ES cell differentiation. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Naoki Izumi
- Laboratory for Stem Cell Biology, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | | | | | | | | |
Collapse
|
15
|
Drakos E, Rassidakis GZ, Leventaki V, Guo W, Medeiros LJ, Nagarajan L. Differential expression of the human MIXL1 gene product in non-Hodgkin and Hodgkin lymphomas. Hum Pathol 2007; 38:500-7. [PMID: 17303500 DOI: 10.1016/j.humpath.2006.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Revised: 09/20/2006] [Accepted: 09/28/2006] [Indexed: 11/16/2022]
Abstract
The Mix1 homeobox-like (MIXL1) gene encodes a paired class homeobox transcription factor that is involved in embryogenesis. Previous studies have shown that the MIXL1 gene product is expressed in B- and T-cell progenitors of normal bone marrow and, in some cell lines derived from hematopoietic neoplasms. The status of MIXL1 expression and subcellular localization in human lymphomas is unknown. Using a highly specific antibody, we assessed for MIXL1 expression in lymphoma cell lines of B- and T-cell lineage by reverse transcriptase-polymerase chain reaction, Western blot analysis, and immunohistochemistry. We also assessed for MIXL1 expression using immunohistochemical methods in 193 lymphoid tumors, including 140 B-cell non-Hodgkin lymphomas (NHL), 36 T-cell NHL, and 17 Hodgkin lymphomas (HL). MIXL1 was detected predominantly in the nuclear fraction of all cell lines tested and was predominantly nuclear in primary tumor specimens. Based on the distribution of the staining results (histogram), a 50% cutoff was selected for high versus low MIXL1 expression. High MIXL1 expression was detected more frequently in Burkitt lymphoma and diffuse large B-cell lymphoma compared with other types of B-cell NHL (P < .0001, chi(2) test). Most cases of T-cell NHL and all cases of HL also highly expressed MIXL1. Most plasma cell myelomas were negative for MIXL1, but rare cases had low MIXL1 expression. MIXL1 expression significantly correlated with proliferation index (Ki-67) in B-cell NHL (P < .0001). The frequent and high expression of MIXL1 in aggressive B-cell NHL, T-cell NHL, and HL suggests that MIXL1 may be involved in lymphomagenesis.
Collapse
Affiliation(s)
- Elias Drakos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Nagaoka M, Ise H, Harada I, Koshimizu U, Maruyama A, Akaike T. Embryonic undifferentiated cells show scattering activity on a surface coated with immobilized E-cadherin. J Cell Biochem 2007; 103:296-310. [PMID: 17559080 DOI: 10.1002/jcb.21406] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Rearrangement of cell-cell adhesion is a critical event in embryonic development and tissue formation. We investigated the regulatory function of E-cadherin, a key adhesion protein, in the developmental process by using E-cadherin/IgG Fc fusion protein as an adhesion matrix in cell culture. F9 embryonal carcinoma cells usually form colonies when cultured on gelatin or fibronectin matrices. However, F9 cells cultured on the E-cadherin/IgG Fc fusion protein matrix formed a scattered distribution, with a different cytoskeletal organization and E-cadherin-rich protrusions that were regulated by Rac1 activity. The same scattering activity was observed in P19 embryonal carcinoma cells. In contrast, three types of differentiated cells, NMuMG mammary gland cells, MDCK kidney epithelial cells, and mouse primary isolated hepatocytes, did not show the scattering activity observed in F9 and P19 cells. These results suggest that migratory behavior on an E-cadherin-immobilized surface is only observed in embryonic cells, and that the regulatory mechanisms underlying E-cadherin-mediated cell adhesion vary with the state of differentiation.
Collapse
Affiliation(s)
- Masato Nagaoka
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Guo W, Nagarajan L. Amino terminal tyrosine phosphorylation of human MIXL1. J Mol Signal 2006; 1:6. [PMID: 17224082 PMCID: PMC1769495 DOI: 10.1186/1750-2187-1-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Accepted: 12/05/2006] [Indexed: 03/08/2023] Open
Abstract
Seven members of the Mix family of paired-type homeoproteins regulate mesoderm/endoderm differentiation in amphibians. In mammals, the MIXL1 (Mix. 1 homeobox [Xenopus laevis]-like gene 1) gene is the sole representative of this family. Unlike the amphibian Mix genes that encode an open reading frame of >300 amino acids, mammalian MIXL1 encodes a smaller protein (~230aa). However, mammalian MIXL1 contains a unique proline-rich domain (PRD) with a potential to interact with signal transducing Src homolgy 3 (SH3) domains. Notably, human MIXL1 also contains a unique tyrosine residue Tyr20 that is amino-terminal to the PRD. Here we report that mammalian MIXL1 protein is phosphorylated at Tyr20 and the phosphorylation is dramatically reduced in the absence of PRD. Our findings are consistent with Tyr20 phosphorylation of MIXL1 being a potential regulatory mechanism that governs its activity.
Collapse
Affiliation(s)
- Wei Guo
- Department of Molecular Genetics, M.D. Anderson Cancer Center, University of Texas, Houston, Texas 77030, USA
| | - Lalitha Nagarajan
- Department of Molecular Genetics, M.D. Anderson Cancer Center, University of Texas, Houston, Texas 77030, USA
| |
Collapse
|
18
|
Glaser S, Metcalf D, Wu L, Hart AH, DiRago L, Mifsud S, D'Amico A, Dagger S, Campo C, Chan AC, Izon DJ, Robb L. Enforced expression of the homeobox gene Mixl1 impairs hematopoietic differentiation and results in acute myeloid leukemia. Proc Natl Acad Sci U S A 2006; 103:16460-5. [PMID: 17060613 PMCID: PMC1637604 DOI: 10.1073/pnas.0607776103] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mixl1, the sole murine homologue of the Xenopus Mix/Bix family of homeobox transcription factors, is essential for the patterning of axial mesendodermal structures during early embryogenesis. Gene targeting and overexpression studies have implicated Mixl1 as a regulator of hematopoiesis arising in differentiating embryonic stem cells. To assess the role of Mixl1 in the regulation of adult hematopoiesis, we overexpressed Mixl1 in murine bone marrow using a retroviral transduction/transplantation model. Enforced expression of Mixl1 profoundly perturbed hematopoietic lineage commitment and differentiation, giving rise to abnormal myeloid progenitors and impairing erythroid and lymphoid differentiation. Moreover, all mice reconstituted with Mixl1-transduced bone marrow developed fatal, transplantable acute myeloid leukemia with a mean latency period of 200 days. These observations establish a link between enforced Mixl1 expression and leukemogenesis in the mouse.
Collapse
Affiliation(s)
- Stefan Glaser
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | - Donald Metcalf
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
- To whom correspondence may be sent. E-mail:
or
| | - Li Wu
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | - Adam H. Hart
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | - Ladina DiRago
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | - Sandra Mifsud
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | - Angela D'Amico
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | - Samantha Dagger
- Telethon Institute for Child Health Research, 100 Roberts Road, Subiaco, Western Australia 6008, Australia; and
| | - Chiara Campo
- Telethon Institute for Child Health Research, 100 Roberts Road, Subiaco, Western Australia 6008, Australia; and
| | - Angela C. Chan
- Telethon Institute for Child Health Research, 100 Roberts Road, Subiaco, Western Australia 6008, Australia; and
| | - David J. Izon
- Telethon Institute for Child Health Research, 100 Roberts Road, Subiaco, Western Australia 6008, Australia; and
- St. Vincent's Institute, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Lorraine Robb
- *The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
- To whom correspondence may be sent. E-mail:
or
| |
Collapse
|
19
|
Mossman AK, Sourris K, Ng E, Stanley EG, Elefanty AG. Mixl1 and oct4 proteins are transiently co-expressed in differentiating mouse and human embryonic stem cells. Stem Cells Dev 2006; 14:656-63. [PMID: 16433620 DOI: 10.1089/scd.2005.14.656] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Embryonic stem cells (ESCs) have the capacity to form all the tissues in the body and hence, directed differentiation of ESCs along specific lineages represents a means to generate therapeutically useful cell types. It has been postulated that, during in vitro differentiation, ES cells sequentially pass through similar developmental stages as cells in the embryo. The availability of reagents that identify these stages would facilitate the monitoring and optimization of ESC differentiation. One key stage, the development of endodermal and mesodermal precursors in the early embryo, is marked by the transient expression of the transcription factor, Mixl1 and the stem cell gene, Oct4. In order to identify corresponding cells during ESC differentiation, we generated monoclonal antibodies to the Mixl1 protein that robustly detected both mouse and human proteins. Intracellular flow cytometry was used to show that approximately 90% of differentiating mouse ESCs transiently co-expressed Oct4 and Mixl1 proteins and that a subset of differentiating human ES cells also coexpressed MIXL1 and OCT4 proteins. These experiments have demonstrated for the first time by protein expression that both human and mouse ESCs passed through developmental stages during in vitro differentiation that corresponded to those observed in early mammalian development. Furthermore, these studies confirmed that anti-Mixl1 antibodies are a valuable reagent for monitoring ESC differentiation and will facilitate the efficient generation of clinically relevant cell types.
Collapse
Affiliation(s)
- Anna K Mossman
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | |
Collapse
|
20
|
Willey S, Ayuso-Sacido A, Zhang H, Fraser ST, Sahr KE, Adlam MJ, Kyba M, Daley GQ, Keller G, Baron MH. Acceleration of mesoderm development and expansion of hematopoietic progenitors in differentiating ES cells by the mouse Mix-like homeodomain transcription factor. Blood 2006; 107:3122-30. [PMID: 16403910 PMCID: PMC1784910 DOI: 10.1182/blood-2005-10-4120] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cellular and molecular events underlying the formation and differentiation of mesoderm to derivatives such as blood are critical to our understanding of the development and function of many tissues and organ systems. How different mesodermal populations are set aside to form specific lineages is not well understood. Although previous genetic studies in the mouse embryo have pointed to a critical role for the homeobox gene Mix-like (mMix) in gastrulation, its function in mesoderm development remains unclear. Hematopoietic defects have been identified in differentiating embryonic stem cells in which mMix was genetically inactivated. Here we show that conditional induction of mMix in embryonic stem cell-derived embryoid bodies results in the early activation of mesodermal markers prior to expression of Brachyury/T and acceleration of the mesodermal developmental program. Strikingly, increased numbers of mesodermal, hemangioblastic, and hematopoietic progenitors form in response to premature activation of mMix. Differentiation to primitive (embryonic) and definitive (adult type) blood cells proceeds normally and without an apparent bias in the representation of different hematopoietic cell fates. Therefore, the mouse Mix gene functions early in the recruitment and/or expansion of mesodermal progenitors to the hemangioblastic and hematopoietic lineages.
Collapse
Affiliation(s)
- Stephen Willey
- Department of Medicine, Mt Sinai School of Medicine, New York, NY 10029-6574, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Baron MH. Early patterning of the mouse embryo: Implications for hematopoietic commitment and differentiation. Exp Hematol 2005; 33:1015-20. [PMID: 16140149 DOI: 10.1016/j.exphem.2005.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Prior to and during gastrulation, reciprocal interactions between embryonic and extraembryonic lineages are crucial for the correct patterning of the embryo. Several lines of investigation have underscored the importance of extraembryonic ectoderm and primitive endodermal in establishing the anterior-posterior axis of the embryo. Signals from these tissues help to position the primitive streak, from which mesoderm will emerge, within the epiblast (embryo proper). Molecules secreted by the visceral endoderm are required for activation of hematopoietic and endothelial cell development, but the pathways involved and their target tissue (e.g., posterior epiblast versus extraembryonic mesoderm) remain obscure. Recent evidence suggests that commitment of mesodermal progenitors to the hematopoietic and endothelial lineages begins earlier than previously anticipated, within or shortly after these cells emerge from the primitive streak.
Collapse
Affiliation(s)
- Margaret H Baron
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW A number of interesting surprises has emerged during the past year in the field of early hematopoietic development. This review highlights recent studies that have challenged the prevailing view of embryonic and fetal hematopoiesis in mammals, with a focus on the mouse as a model system. The authors apologize to the many colleagues whose work could not be cited because of space limitations. RECENT FINDINGS Advances in our understanding of the embryonic origins of hematopoiesis in mammals and in the regulation of primitive and definitive hematopoietic development are discussed. SUMMARY The ontological relation between primitive (embryonic) and definitive (fetal and adult) hematopoiesis still holds some mysteries for the biologist. Both technical and conceptual breakthroughs have refined our view of how blood cells form at different stages of development. What we learn from the embryo is not only of fundamental interest but may have future applications in the clinic.
Collapse
Affiliation(s)
- Margaret H Baron
- Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | |
Collapse
|
23
|
Shiraki N, Lai CJ, Hishikari Y, Kume S. TGF-β signaling potentiates differentiation of embryonic stem cells to Pdx-1 expressing endodermal cells. Genes Cells 2005; 10:503-16. [PMID: 15938710 DOI: 10.1111/j.1365-2443.2005.00854.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Embryonic stem (ES) cells have the capacity to differentiate to every cell type that constitutes fetal or adult tissues. To trace and quantitatively assess the differentiation of ES cells into gut endodermal cells, we used an ES cell line with the lacZ gene inserted into the pdx-1 locus. Targeted mutations of pdx-1 in mice demonstrate that pdx-1 is required for pancreatic and rostral duodenal development; therefore, pdx-1 serves as an excellent early gut regional specific marker. When these ES cells were differentiated by removal of leukemia inhibitory factor (LIF), only fractional cells turned into lacZ positive, which indicates pancreatic-duodenal differentiation. Co-cultivation of ES cells with pancreatic rudiments induced a significant increase in the proportion of lacZ positive cell numbers and this increase was further enhanced by forced expression of a chick putative endoderm inducer gene, cmix. Transforming growth factor (TGF)-beta2 mimicked the effects of pancreatic rudiments and this effect was enhanced by cmix expression. Expression analysis showed over-expression of cmix induced endodermal marker genes. These data indicate that one can make use of this knowledge on molecular events of embryonic development to drive ES cells to differentiate into pdx-1 expressing endodermal cells in vitro.
Collapse
Affiliation(s)
- Nobuaki Shiraki
- Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | | | | | | |
Collapse
|
24
|
Kubo A, Chen V, Kennedy M, Zahradka E, Daley GQ, Keller G. The homeobox gene HEX regulates proliferation and differentiation of hemangioblasts and endothelial cells during ES cell differentiation. Blood 2005; 105:4590-7. [PMID: 15728128 PMCID: PMC1895005 DOI: 10.1182/blood-2004-10-4137] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this report we have investigated the role of the homeobox gene Hex in the development and differentiation of the blast colony-forming cell (BL-CFC), a progenitor with hemangioblast characteristics generated in embryonic stem (ES) cell-derived embryoid bodies (EBs). Molecular analysis showed that Hex is expressed in mesoderm, in populations that contain BL-CFCs, and in blast cell colonies, the progeny of the BL-CFCs. Hex(-/-) EBs displayed a defect in macrophage development but generated higher numbers of BL-CFCs than did wild-type EBs. In addition to differences in these progenitor populations, we also found that endothelial cells from the Hex(-/-) EBs showed enhanced proliferative potential compared with those from wild-type EBs. Forced expression of Hex at the onset of ES cell differentiation resulted in reduced EB cellularity, fetal liver kinase-1 (Flk-1) expression, and BL-CFC development. Taken together, these findings demonstrate that Hex functions at multiple stages of development within the differentiating EBs and uncover a novel role for this transcription factor as a negative regulator of the hemangioblast and the endothelial lineage.
Collapse
Affiliation(s)
- Atsushi Kubo
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
25
|
Ng ES, Azzola L, Sourris K, Robb L, Stanley EG, Elefanty AG. The primitive streak gene Mixl1 is required for efficient haematopoiesis and BMP4-induced ventral mesoderm patterning in differentiating ES cells. Development 2005; 132:873-84. [PMID: 15673572 DOI: 10.1242/dev.01657] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The homeobox gene Mixl1 is expressed in the primitive streak of the gastrulating embryo, and marks cells destined to form mesoderm and endoderm. The role of Mixl1 in development of haematopoietic mesoderm was investigated by analysing the differentiation of ES cells in which GFP was targeted to one (Mixl1(GFP/w)) or both (Mixl1(GFP/GFP)) alleles of the Mixl1 locus. In either case, GFP was transiently expressed, with over 80% of cells in day 4 embryoid bodies (EBs) being GFP(+). Up to 45% of Mixl1(GFP/w) day 4 EB cells co-expressed GFP and the haemangioblast marker FLK1, and this doubly-positive population was enriched for blast colony forming cells (BL-CFCs). Mixl1-null ES cells, however, displayed a haematopoietic defect characterised by reduced and delayed Flk1 expression and a decrease in the frequency of haematopoietic CFCs. These data indicated that Mixl1 was required for efficient differentiation of cells from the primitive streak stage to blood. Differentiation of ES cells under serum-free conditions demonstrated that induction of Mixl1- and Flk1-expressing haematopoietic mesoderm required medium supplemented with BMP4 or activin A. In conclusion, this study has revealed an important role for Mixl1 in haematopoietic development and demonstrates the utility of the Mixl1(GFP/w) ES cells for evaluating growth factors influencing mesendodermal differentiation.
Collapse
Affiliation(s)
- Elizabeth S Ng
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, VIC 3800, Australia
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
The intensity of research on pancreatic development has increased markedly in the past 5 years, primarily for two reasons: we now know that the insulin-producing beta-cells normally arise from an endodermally derived, pancreas-specified precursor cell, and successful transplants of islet cells have been performed, relieving patients with type I diabetes of symptoms for extended periods after transplantation. Combining in vitro beta-cell formation from a pancreatic biopsy of a diabetic patient or from other stem-cell sources followed by endocrine cell transplantation may be the most beneficial route for a future diabetes therapy. However, to achieve this, a thorough understanding of the genetic components regulating the development of beta-cells is required. The following review discusses our current understanding of the transcription factor networks necessary for pancreatic development and how several genetic interactions coming into play at the earliest stages of endodermal development gradually help to build the pancreatic organ. Developmental Dynamics 229:176-200, 2004.
Collapse
Affiliation(s)
- Jan Jensen
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver, Colorado, USA.
| |
Collapse
|
27
|
Abstract
Hematopoiesis and vasculogenesis in the mammalian embryo begin in the blood islands of the yolk sac and continue, somewhat later, within the embryo proper. A subset of the first endothelial and hematopoietic cells of the yolk sac arise in close spatial and temporal association, apparently from a common mesodermal progenitor, the "hemangioblast." The mechanisms that control formation of hemangioblast and embryonic hematopoietic and endothelial (angioblastic) stem/progenitor cells are still not well understood. Formation of these cell types from nascent mesoderm requires signals from an adjacent outer layer of primitive (visceral) endoderm. Indian hedgehog (Ihh), a member of the hedgehog family of extracellular morphogens, is secreted by visceral endoderm and alone is sufficient to induce hematopoiesis and vasculogenesis in explanted embryos. While gene targeting studies in mice support a role for hedgehog signaling in these processes in vivo, they also suggest that additional molecules (perhaps, for example, Wnt proteins) are required for induction and patterning of hematopoietic and vascular mesoderm. Indian hedgehog likely functions through upregulation of genes encoding other signaling molecules, such as bone morphogenetic protein (Bmp)-4, in the target tissue. This review will focus on hematopoietic and vascular development in the early mouse embryo and will discuss potential implications of recent studies for stem cell transplantation in humans.
Collapse
Affiliation(s)
- Margaret H Baron
- Department of Medicine, Molecular, Brookdale Department of Cell and Developmental Biology, Ruttenberg Cancer Center, Mount Sinai School of Medicine, 1425 Madison Avenue 11-70B, Box 1079, New York, NY 10029, USA.
| |
Collapse
|