Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Sci Adv ; 10(25): eado1583, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905335

ABSTRACT

Neuroblastoma is a childhood developmental cancer; however, its embryonic origins remain poorly understood. Moreover, in-depth studies of early tumor-driving events are limited because of the lack of appropriate models. Herein, we analyzed RNA sequencing data obtained from human neuroblastoma samples and found that loss of expression of trunk neural crest-enriched gene MOXD1 associates with advanced disease and worse outcome. Further, by using single-cell RNA sequencing data of human neuroblastoma cells and fetal adrenal glands and creating in vivo models of zebrafish, chick, and mouse, we show that MOXD1 is a determinate of tumor development. In addition, we found that MOXD1 expression is highly conserved and restricted to mesenchymal neuroblastoma cells and Schwann cell precursors during healthy development. Our findings identify MOXD1 as a lineage-restricted tumor-suppressor gene in neuroblastoma, potentiating further stratification of these tumors and development of novel therapeutic interventions.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Neuroblastoma , Zebrafish , Animals , Humans , Mice , Cell Line, Tumor , Cell Lineage/genetics , Neural Crest/metabolism , Neural Crest/pathology , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Zebrafish/genetics
2.
Nat Commun ; 14(1): 1267, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36882421

ABSTRACT

The pediatric extra-cranial tumor neuroblastoma displays a low mutational burden while recurrent copy number alterations are present in most high-risk cases. Here, we identify SOX11 as a dependency transcription factor in adrenergic neuroblastoma based on recurrent chromosome 2p focal gains and amplifications, specific expression in the normal sympatho-adrenal lineage and adrenergic neuroblastoma, regulation by multiple adrenergic specific (super-)enhancers and strong dependency on high SOX11 expression in adrenergic neuroblastomas. SOX11 regulated direct targets include genes implicated in epigenetic control, cytoskeleton and neurodevelopment. Most notably, SOX11 controls chromatin regulatory complexes, including 10 SWI/SNF core components among which SMARCC1, SMARCA4/BRG1 and ARID1A. Additionally, the histone deacetylase HDAC2, PRC1 complex component CBX2, chromatin-modifying enzyme KDM1A/LSD1 and pioneer factor c-MYB are regulated by SOX11. Finally, SOX11 is identified as a core transcription factor of the core regulatory circuitry (CRC) in adrenergic high-risk neuroblastoma with a potential role as epigenetic master regulator upstream of the CRC.


Subject(s)
Neuroblastoma , Humans , Child , Neuroblastoma/genetics , Transcription Factors/genetics , Chromatin , Cell Nucleus , Chromosome Aberrations , Adrenergic Agents , DNA Helicases , Nuclear Proteins/genetics , SOXC Transcription Factors/genetics , Histone Demethylases
4.
Cancer Res ; 82(3): 484-496, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34853072

ABSTRACT

Cancer therapy frequently fails due to the emergence of resistance. Many tumors include phenotypically immature tumor cells, which have been implicated in therapy resistance. Neuroblastoma cells can adopt a lineage-committed adrenergic (ADRN) or an immature mesenchymal (MES) state. They differ in epigenetic landscape and transcription factors, and MES cells are more resistant to chemotherapy. Here we analyzed the response of MES cells to targeted drugs. Activating anaplastic lymphoma kinase (ALK) mutations are frequently found in neuroblastoma and ALK inhibitors (ALKi) are in clinical trials. ALKi treatment of ADRN neuroblastoma cells with a tumor-driving ALK mutation induced cell death. Conversely, MES cells did not express either mutant or wild-type ALK and were resistant to ALKi, and MES cells formed tumors that progressed under ALKi therapy. In assessing the role of MES cells in relapse development, TRAIL was identified to specifically induce apoptosis in MES cells and to suppress MES tumor growth. Addition of TRAIL to ALKi treatment of neuroblastoma xenografts delayed relapses in a subset of the animals, suggesting a role for MES cells in relapse formation. While ADRN cells resembled normal embryonal neuroblasts, MES cells resembled immature precursor cells, which also lacked ALK expression. Resistance to targeted drugs can therefore be an intrinsic property of immature cancer cells based on their resemblance to developmental precursors. SIGNIFICANCE: In neuroblastoma, mesenchymal tumor cells lack expression of the tumor-driving ALK oncogene and are resistant to ALKi, but dual treatment with ALKi and mesenchymal cell-targeting TRAIL delays tumor relapse.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Neuroblastoma/genetics , Cell Line, Tumor , Humans , Neuroblastoma/pathology
5.
Nat Cancer ; 2(1): 114-128, 2021 01.
Article in English | MEDLINE | ID: mdl-35121888

ABSTRACT

Half of the children diagnosed with neuroblastoma (NB) have high-risk disease, disproportionately contributing to overall childhood cancer-related deaths. In addition to recurrent gene mutations, there is increasing evidence supporting the role of epigenetic deregulation in disease pathogenesis. Yet, comprehensive cis-regulatory network descriptions from NB are lacking. Here, using genome-wide H3K27ac profiles across 60 NBs, covering the different clinical and molecular subtypes, we identified four major super-enhancer-driven epigenetic subtypes and their underlying master regulatory networks. Three of these subtypes recapitulated known clinical groups; namely, MYCN-amplified, MYCN non-amplified high-risk and MYCN non-amplified low-risk NBs. The fourth subtype, exhibiting mesenchymal characteristics, shared cellular identity with multipotent Schwann cell precursors, was induced by RAS activation and was enriched in relapsed disease. Notably, CCND1, an essential gene in NB, was regulated by both mesenchymal and adrenergic regulatory networks converging on distinct super-enhancer modules. Overall, this study reveals subtype-specific super-enhancer regulation in NBs.


Subject(s)
Neuroblastoma , Child , Humans , Mutation , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Regulatory Sequences, Nucleic Acid
6.
Nat Commun ; 10(1): 1530, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948783

ABSTRACT

Transition between differentiation states in development occurs swift but the mechanisms leading to epigenetic and transcriptional reprogramming are poorly understood. The pediatric cancer neuroblastoma includes adrenergic (ADRN) and mesenchymal (MES) tumor cell types, which differ in phenotype, super-enhancers (SEs) and core regulatory circuitries. These cell types can spontaneously interconvert, but the mechanism remains largely unknown. Here, we unravel how a NOTCH3 intracellular domain reprogrammed the ADRN transcriptional landscape towards a MES state. A transcriptional feed-forward circuitry of NOTCH-family transcription factors amplifies the NOTCH signaling levels, explaining the swift transition between two semi-stable cellular states. This transition induces genome-wide remodeling of the H3K27ac landscape and a switch from ADRN SEs to MES SEs. Once established, the NOTCH feed-forward loop maintains the induced MES state. In vivo reprogramming of ADRN cells shows that MES and ADRN cells are equally oncogenic. Our results elucidate a swift transdifferentiation between two semi-stable epigenetic cellular states.


Subject(s)
Adrenergic Neurons/pathology , Cellular Reprogramming/genetics , Mesenchymal Stem Cells/pathology , Neuroblastoma/pathology , Receptor, Notch3/physiology , Adrenergic Neurons/metabolism , Cell Line, Tumor , Epigenesis, Genetic , Feedback, Physiological , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mesenchymal Stem Cells/metabolism , Neuroblastoma/metabolism , Receptor, Notch3/genetics , Receptor, Notch3/metabolism
7.
Article in English | MEDLINE | ID: mdl-34036221

ABSTRACT

Patients with neuroblastoma in molecular remission remain at considerable risk for disease recurrence. Studies have found that neuroblastoma tissue contains adrenergic (ADRN) and mesenchymal (MES) cells; the latter express low levels of commonly used markers for minimal residual disease (MRD). We identified MES-specific MRD markers and studied the dynamics of these markers during treatment. PATIENTS AND METHODS: Microarray data were used to identify genes differentially expressed between ADRN and MES cell lines. Candidate genes were then studied using real-time quantitative polymerase chain reaction in cell lines and control bone marrow and peripheral blood samples. After selecting a panel of markers, serial bone marrow, peripheral blood, and peripheral blood stem cell samples were obtained from patients with high-risk neuroblastoma and tested for marker expression; survival analyses were also performed. RESULTS: PRRX1, POSTN, and FMO3 mRNAs were used as a panel for specifically detecting MES mRNA in patient samples. MES mRNA was detected only rarely in peripheral blood; moreover, the presence of MES mRNA in peripheral blood stem cell samples was associated with low event-free survival and overall survival. Of note, during treatment, serial bone marrow samples obtained from 29 patients revealed a difference in dynamics between MES mRNA markers and ADRN mRNA markers. Furthermore, MES mRNA was detected in a higher percentage of patients with recurrent disease than in those who remained disease free (53% v 32%, respectively; P = .03). CONCLUSION: We propose that the markers POSTN and PRRX1, in combination with FMO3, be used for real-time quantitative polymerase chain reaction-based detection of MES neuroblastoma mRNA in patient samples because these markers have a unique pattern during treatment and are more prevalent in patients with poor outcome. Together with existing markers of MRD, these new markers should be investigated further in large prospective studies.

8.
Nat Commun ; 9(1): 4866, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451831

ABSTRACT

Chromosome 17q gains are almost invariably present in high-risk neuroblastoma cases. Here, we perform an integrative epigenomics search for dosage-sensitive transcription factors on 17q marked by H3K27ac defined super-enhancers and identify TBX2 as top candidate gene. We show that TBX2 is a constituent of the recently established core regulatory circuitry in neuroblastoma with features of a cell identity transcription factor, driving proliferation through activation of p21-DREAM repressed FOXM1 target genes. Combined MYCN/TBX2 knockdown enforces cell growth arrest suggesting that TBX2 enhances MYCN sustained activation of FOXM1 targets. Targeting transcriptional addiction by combined CDK7 and BET bromodomain inhibition shows synergistic effects on cell viability with strong repressive effects on CRC gene expression and p53 pathway response as well as several genes implicated in transcriptional regulation. In conclusion, we provide insight into the role of the TBX2 CRC gene in transcriptional dependency of neuroblastoma cells warranting clinical trials using BET and CDK7 inhibitors.


Subject(s)
Brain Neoplasms/genetics , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Kv Channel-Interacting Proteins/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Repressor Proteins/genetics , T-Box Domain Proteins/genetics , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA Copy Number Variations , Epigenesis, Genetic , Forkhead Box Protein M1/metabolism , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Kv Channel-Interacting Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Panobinostat/pharmacology , Phenylenediamines/pharmacology , Pyrimidines/pharmacology , Repressor Proteins/metabolism , Signal Transduction , T-Box Domain Proteins/metabolism , Triazoles/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase-Activating Kinase
9.
Nat Genet ; 49(8): 1261-1266, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28650485

ABSTRACT

Neuroblastoma and other pediatric tumors show a paucity of gene mutations, which has sparked an interest in their epigenetic regulation. Several tumor types include phenotypically divergent cells, resembling cells from different lineage development stages. It has been proposed that super-enhancer-associated transcription factor (TF) networks underlie lineage identity, but the role of these enhancers in intratumoral heterogeneity is unknown. Here we show that most neuroblastomas include two types of tumor cells with divergent gene expression profiles. Undifferentiated mesenchymal cells and committed adrenergic cells can interconvert and resemble cells from different lineage differentiation stages. ChIP-seq analysis of isogenic pairs of mesenchymal and adrenergic cells identified a distinct super-enhancer landscape and super-enhancer-associated TF network for each cell type. Expression of the mesenchymal TF PRRX1 could reprogram the super-enhancer and mRNA landscapes of adrenergic cells toward a mesenchymal state. Mesenchymal cells were more chemoresistant in vitro and were enriched in post-therapy and relapse tumors. Two super-enhancer-associated TF networks, which probably mediate lineage control in normal development, thus dominate epigenetic control of neuroblastoma and shape intratumoral heterogeneity.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Neuroblastoma/genetics , Neuroblastoma/pathology , AC133 Antigen/genetics , Adrenergic Neurons/cytology , Cell Line, Tumor , Cell Lineage , Homeodomain Proteins/genetics , Humans , Mesoderm/cytology , Transcription Factors/metabolism , Transcriptome
10.
Clin Cancer Res ; 19(13): 3485-94, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23649002

ABSTRACT

PURPOSE: Neuroblastoma is a childhood tumor of the peripheral sympathetic nervous system with an often lethal outcome due to metastatic disease. Migration and epithelial-mesenchymal transitions have been implicated in metastasis but they are hardly investigated in neuroblastoma. EXPERIMENTAL DESIGN: Cell migration of 16 neuroblastoma cell lines was quantified in Transwell migration assays. Gene expression profiling was used to derive a migration signature, which was applied to classify samples in a neuroblastoma tumor series. Differential expression of transcription factors was analyzed in the subsets. NOTCH3 was prioritized, and inducible transgene expression studies in cell lines were used to establish whether it functions as a master switch for motility. RESULTS: We identified a 36-gene expression signature that predicts cell migration. This signature was used to analyse expression profiles of 88 neuroblastoma tumors and identified a group with distant metastases and a poor prognosis. This group also expressed a known mesenchymal gene signature established in glioblastoma. Neuroblastomas recognized by the motility and mesenchymal signatures strongly expressed genes of the NOTCH pathway. Inducible expression of a NOTCH intracellular (NOTCH3-IC) transgene conferred a highly motile phenotype to neuroblastoma cells. NOTCH3-IC strongly induced expression of motility- and mesenchymal marker genes. Many of these genes were significantly coexpressed with NOTCH3 in neuroblastoma, as well as colon, kidney, ovary, and breast tumor series. CONCLUSION: The NOTCH3 transcription factor is a master regulator of motility in neuroblastoma. A subset of neuroblastoma with high expression of NOTCH3 and its downstream-regulated genes has mesenchymal characteristics, increased incidence of metastases, and a poor prognosis.


Subject(s)
Cell Movement/genetics , Neuroblastoma/genetics , Receptors, Notch/genetics , Transcription, Genetic , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Neuroblastoma/metabolism , Neuroblastoma/pathology , Receptor, Notch3 , Receptors, Notch/metabolism
11.
Proc Natl Acad Sci U S A ; 109(47): 19190-5, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23091029

ABSTRACT

Neuroblastoma is a pediatric tumor of the sympathetic nervous system. MYCN (V-myc myelocytomatosis viral-related oncogene, neuroblastoma derived [avian]) is amplified in 20% of neuroblastomas, and these tumors carry a poor prognosis. However, tumors without MYCN amplification also may have a poor outcome. Here, we identified downstream targets of MYCN by shRNA-mediated silencing MYCN in neuroblastoma cells. From these targets, 157 genes showed an expression profile correlating with MYCN mRNA levels in NB88, a series of 88 neuroblastoma tumors, and therefore represent in vivo relevant MYCN pathway genes. This 157-gene signature identified very poor prognosis tumors in NB88 and independent neuroblastoma cohorts and was more powerful than MYCN amplification or MYCN expression alone. Remarkably, this signature also identified poor outcome of a group of tumors without MYCN amplification. Most of these tumors have low MYCN mRNA levels but high nuclear MYCN protein levels, suggesting stabilization of MYCN at the protein level. One tumor has an MYC amplification and high MYC expression. Chip-on-chip analyses showed that most genes in this signature are directly regulated by MYCN. MYCN induces genes functioning in cell cycle and DNA repair while repressing neuronal differentiation genes. The functional MYCN-157 signature recognizes classical neuroblastoma with MYCN amplification, as well as a newly identified group marked by MYCN protein stabilization.


Subject(s)
Gene Amplification/genetics , Gene Expression Profiling , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Cell Differentiation/genetics , Cluster Analysis , DNA Repair/genetics , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Humans , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Neurons/pathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Treatment Outcome , Up-Regulation/genetics
12.
Nat Genet ; 44(11): 1199-206, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23042116

ABSTRACT

LIN28B regulates developmental processes by modulating microRNAs (miRNAs) of the let-7 family. A role for LIN28B in cancer has been proposed but has not been established in vivo. Here, we report that LIN28B showed genomic aberrations and extensive overexpression in high-risk neuroblastoma compared to several other tumor entities and normal tissues. High LIN28B expression was an independent risk factor for adverse outcome in neuroblastoma. LIN28B signaled through repression of the let-7 miRNAs and consequently resulted in elevated MYCN protein expression in neuroblastoma cells. LIN28B-let-7-MYCN signaling blocked differentiation of normal neuroblasts and neuroblastoma cells. These findings were fully recapitulated in a mouse model in which LIN28B expression in the sympathetic adrenergic lineage induced development of neuroblastomas marked by low let-7 miRNA levels and high MYCN protein expression. Interference with this pathway might offer therapeutic perspectives.


Subject(s)
DNA-Binding Proteins/genetics , MicroRNAs , Neuroblastoma , Nuclear Proteins , Oncogene Proteins , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Neuroblastoma/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , RNA-Binding Proteins , Signal Transduction
13.
Nature ; 483(7391): 589-93, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22367537

ABSTRACT

Neuroblastoma is a childhood tumour of the peripheral sympathetic nervous system. The pathogenesis has for a long time been quite enigmatic, as only very few gene defects were identified in this often lethal tumour. Frequently detected gene alterations are limited to MYCN amplification (20%) and ALK activations (7%). Here we present a whole-genome sequence analysis of 87 neuroblastoma of all stages. Few recurrent amino-acid-changing mutations were found. In contrast, analysis of structural defects identified a local shredding of chromosomes, known as chromothripsis, in 18% of high-stage neuroblastoma. These tumours are associated with a poor outcome. Structural alterations recurrently affected ODZ3, PTPRD and CSMD1, which are involved in neuronal growth cone stabilization. In addition, ATRX, TIAM1 and a series of regulators of the Rac/Rho pathway were mutated, further implicating defects in neuritogenesis in neuroblastoma. Most tumours with defects in these genes were aggressive high-stage neuroblastomas, but did not carry MYCN amplifications. The genomic landscape of neuroblastoma therefore reveals two novel molecular defects, chromothripsis and neuritogenesis gene alterations, which frequently occur in high-risk tumours.


Subject(s)
Chromosomes, Human/genetics , Neurites/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Aging/genetics , Cluster Analysis , DNA Helicases/genetics , DNA Mutational Analysis , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Growth Cones/metabolism , Growth Cones/pathology , Guanine Nucleotide Exchange Factors/genetics , Humans , Mutation , Neoplasm Staging , Neuroblastoma/diagnosis , Neuroblastoma/metabolism , Nuclear Proteins/genetics , Prognosis , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , X-linked Nuclear Protein , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
14.
Dev Cell ; 17(4): 516-26, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19853565

ABSTRACT

Hox and Cdx transcription factors regulate embryonic positional identities. Cdx mutant mice display posterior body truncations of the axial skeleton, neuraxis, and caudal urorectal structures. We show that trunk Hox genes stimulate axial extension, as they can largely rescue these Cdx mutant phenotypes. Conversely, posterior (paralog group 13) Hox genes can prematurely arrest posterior axial growth when precociously expressed. Our data suggest that the transition from trunk to tail Hox gene expression successively regulates the construction and termination of axial structures in the mouse embryo. Thus, Hox genes seem to differentially orchestrate posterior expansion of embryonic tissues during axial morphogenesis as an integral part of their function in specifying head-to-tail identity. In addition, we present evidence that Cdx and Hox transcription factors exert these effects by controlling Wnt signaling. Concomitant regulation of Cyp26a1 expression, restraining retinoic acid signaling away from the posterior growth zone, may likewise play a role in timing the trunk-tail transition.


Subject(s)
Body Patterning/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Genes, Homeobox/physiology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , CDX2 Transcription Factor , Cytochrome P-450 Enzyme System/metabolism , Extremities/embryology , Gene Expression Profiling , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinoic Acid 4-Hydroxylase , Reverse Transcriptase Polymerase Chain Reaction , Skeleton , Tretinoin/pharmacology , Wnt Proteins/metabolism
15.
Development ; 133(3): 419-28, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16396910

ABSTRACT

Caudal related homeobox (Cdx) genes have so far been shown to be important for embryonic axial elongation and patterning in several vertebrate species. We have generated a targeted mutation of mouse Cdx4, the third member of this family of transcription factor encoding genes and the last one to be inactivated genetically. Cdx4-null embryos were born healthy and appeared morphologically normal. A subtle contribution of Cdx4 to anteroposterior (AP) vertebral patterning was revealed in Cdx1/Cdx4 and Cdx2/Cdx4 compound mutants. Neither Cdx4-null nor Cdx1/Cdx4 double mutants are impaired in their axial elongation, but a redundant contribution of Cdx4 in this function was unveiled when combined with a Cdx2 mutant allele. In addition, inactivation of Cdx4 combined with heterozygous loss of Cdx2 results in embryonic death around E10.5 and reveals a novel function of Cdx genes in placental ontogenesis. In a subset of Cdx2/Cdx4 compound mutants, the fully grown allantois failed to fuse with the chorion. The remaining majority of these mutants undergo successful chorio-allantois fusion but fail to properly extend their allantoic vascular network into the chorionic ectoderm and do not develop a functional placental labyrinth. We present evidence that Cdx4 plays a crucial role in the ontogenesis of the allantoic component of the placental labyrinth when one Cdx2 allele is inactivated. The axial patterning role of Cdx transcription factors thus extends posteriorly to the epiblast-derived extra-embryonic mesoderm and, consequent upon the evolution of placental mammals, is centrally involved in placental morphogenesis. The relative contribution of Cdx family members in the stepwise ontogenesis of a functional placenta is discussed, with Cdx2 playing an obligatory part, assisted by Cdx4. The possible participation of Cdx1 was not documented but cannot be ruled out until allelic combinations further decreasing Cdx dose have been analyzed. Cdx genes thus operate in a redundant way during placentogenesis, as they do during embryonic axial elongation and patterning, and independently from the previously reported early Cdx2-specific role in the trophectoderm at implantation.


Subject(s)
Allantois/anatomy & histology , Body Patterning , Embryo, Mammalian , Embryo, Nonmammalian , Homeodomain Proteins/genetics , Mutation , Animals , Biomarkers/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Fishes/embryology , Fishes/genetics , Fishes/metabolism , Gene Expression Regulation, Developmental , Gene Targeting , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Placenta/anatomy & histology
16.
Development ; 132(13): 2931-42, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15944185

ABSTRACT

The Hox genes confer positional information to the axial and paraxial tissues as they emerge gradually from the posterior aspect of the vertebrate embryo. Hox genes are sequentially activated in time and space, in a way that reflects their organisation into clusters in the genome. Although this co-linearity of expression of the Hox genes has been conserved during evolution, it is a phenomenon that is still not understood at the molecular level. This review aims to bring together recent findings that have advanced our understanding of the regulation of the Hox genes during mouse embryonic development. In particular, we highlight the integration of these transducers of anteroposterior positional information into the genetic network that drives tissue generation and patterning during axial elongation.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental/physiology , Genes, Homeobox , Animals , Mice
SELECTION OF CITATIONS
SEARCH DETAIL