Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cancer Cell ; 41(7): 1242-1260.e6, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37267953

ABSTRACT

The accumulation of senescent cells in the tumor microenvironment can drive tumorigenesis in a paracrine manner through the senescence-associated secretory phenotype (SASP). Using a new p16-FDR mouse line, we show that macrophages and endothelial cells are the predominant senescent cell types in murine KRAS-driven lung tumors. Through single cell transcriptomics, we identify a population of tumor-associated macrophages that express a unique array of pro-tumorigenic SASP factors and surface proteins and are also present in normal aged lungs. Genetic or senolytic ablation of senescent cells, or macrophage depletion, result in a significant decrease in tumor burden and increased survival in KRAS-driven lung cancer models. Moreover, we reveal the presence of macrophages with senescent features in human lung pre-malignant lesions, but not in adenocarcinomas. Taken together, our results have uncovered the important role of senescent macrophages in the initiation and progression of lung cancer, highlighting potential therapeutic avenues and cancer preventative strategies.


Subject(s)
Cellular Senescence , Lung Neoplasms , Aged , Animals , Humans , Mice , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cellular Senescence/genetics , Endothelial Cells , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Macrophages/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Microenvironment
2.
Nat Commun ; 9(1): 3123, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30072790

ABSTRACT

The original version of this article contained an error in the spelling of Juan Pedro Martinez-Barbera, which was incorrectly given as Juan Pedro Martinez Barbera. This error has now been corrected in both the PDF and HTML versions of the Article.

3.
Nat Commun ; 9(1): 1763, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720666

ABSTRACT

Ensuring the fitness of the pluripotent cells that will contribute to future development is important both for the integrity of the germline and for proper embryogenesis. Consequently, it is becoming increasingly apparent that pluripotent cells can compare their fitness levels and signal the elimination of those cells that are less fit than their neighbours. In mammals the nature of the pathways that communicate fitness remain largely unknown. Here we identify that in the early mouse embryo and upon exit from naive pluripotency, the confrontation of cells with different fitness levels leads to an inhibition of mTOR signalling in the less fit cell type, causing its elimination. We show that during this process, p53 acts upstream of mTOR and is required to repress its activity. Finally, we demonstrate that during normal development around 35% of cells are eliminated by this pathway, highlighting the importance of this mechanism for embryonic development.


Subject(s)
Embryo, Mammalian/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Communication/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/genetics
4.
Pain ; 159(8): 1641-1651, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29697531

ABSTRACT

Voltage-gated potassium (Kv) channels are increasingly recognised as key regulators of nociceptive excitability. Kcns1 is one of the first potassium channels to be associated with neuronal hyperexcitability and mechanical sensitivity in the rat, as well as pain intensity and risk of developing chronic pain in humans. Here, we show that in mice, Kcns1 is predominantly expressed in the cell body and axons of myelinated sensory neurons positive for neurofilament-200, including Aδ-fiber nociceptors and low-threshold Aß mechanoreceptors. In the spinal cord, Kcns1 was detected in laminae III to V of the dorsal horn where most sensory A fibers terminate, as well as large motoneurons of the ventral horn. To investigate Kcns1 function specifically in the periphery, we generated transgenic mice in which the gene is deleted in all sensory neurons but retained in the central nervous system. Kcns1 ablation resulted in a modest increase in basal mechanical pain, with no change in thermal pain processing. After neuropathic injury, Kcns1 KO mice exhibited exaggerated mechanical pain responses and hypersensitivity to both noxious and innocuous cold, consistent with increased A-fiber activity. Interestingly, Kcns1 deletion also improved locomotor performance in the rotarod test, indicative of augmented proprioceptive signalling. Our results suggest that restoring Kcns1 function in the periphery may be of some use in ameliorating mechanical and cold pain in chronic states.


Subject(s)
Neuralgia/metabolism , Pain Threshold/physiology , Posterior Horn Cells/metabolism , Potassium Channels, Voltage-Gated/metabolism , Animals , Mice , Mice, Knockout , Motor Skills/physiology , Neuralgia/genetics , Physical Stimulation , Potassium Channels, Voltage-Gated/genetics , Proprioception/physiology
5.
Proc Natl Acad Sci U S A ; 113(5): E548-57, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26764381

ABSTRACT

Aberrant embryonic development of the hypothalamus and/or pituitary gland in humans results in congenital hypopituitarism (CH). Transcription factor 7-like 1 (TCF7L1), an important regulator of the WNT/ß-catenin signaling pathway, is expressed in the developing forebrain and pituitary gland, but its role during hypothalamo-pituitary (HP) axis formation or involvement in human CH remains elusive. Using a conditional genetic approach in the mouse, we first demonstrate that TCF7L1 is required in the prospective hypothalamus to maintain normal expression of the hypothalamic signals involved in the induction and subsequent expansion of Rathke's pouch progenitors. Next, we reveal that the function of TCF7L1 during HP axis development depends exclusively on the repressing activity of TCF7L1 and does not require its interaction with ß-catenin. Finally, we report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects. We demonstrate that these variants exhibit reduced repressing activity in vitro and in vivo relative to wild-type TCF7L1. Together, our data provide support for a conserved molecular function of TCF7L1 as a transcriptional repressor during HP axis development in mammals and identify variants in this transcription factor that are likely to contribute to the etiology of CH.


Subject(s)
Hypothalamo-Hypophyseal System , Transcription Factor 7-Like 1 Protein/physiology , Animals , Cohort Studies , Humans , Mice , Pituitary Gland/abnormalities , Pituitary Gland/metabolism , Pituitary Gland/physiopathology , Prosencephalon/abnormalities , Prosencephalon/metabolism
6.
Acta Neuropathol Commun ; 3: 74, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26610346

ABSTRACT

BACKGROUND: Tamoxifen (TAM) is an important cancer therapeutic and an experimental tool for effecting genetic recombination using the inducible Cre-Lox technique. Despite its widespread use in the clinic and laboratory, we know little about its effects on the nervous system. This is of significant concern because TAM, via unknown mechanisms, induces cognitive impairment in humans. A hallmark of cellular stress is induction of Activating Transcription Factor 3 (Atf3), and so to determine whether TAM induces cellular stress in the adult nervous system, we generated a knock-in mouse in which Atf3 promoter activity drives transcription of TAM-dependent Cre recombinase (Cre-ERT2); when crossed with tdtomato reporter mice, Atf3 induction results in robust and permanent genetic labeling of cells in which it is up-regulated even transiently. RESULTS: We found that granular neurons of the olfactory bulb and dentate gyrus, vascular cells and ependymal cells throughout the brain, and peripheral sensory neurons expressed tdtomato in response to TAM treatment. We also show that TAM induced Atf3 up-regulation through inhibition of cholesterol epoxide hydrolase (ChEH): reporter expression was mitigated by delivery in vitamin E-rich wheat germ oil (vitamin E depletes ChEH substrates), and was partially mimicked by a ChEH-specific inhibitor. CONCLUSIONS: This work demonstrates that TAM stresses cells of the adult central and peripheral nervous systems and highlights concerns about clinical and experimental use of TAM. We propose TAM administration in vitamin E-rich vehicles such as wheat germ oil as a simple remedy.


Subject(s)
Cholesterol/metabolism , Nervous System/cytology , Neurons/physiology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Up-Regulation/drug effects , Activating Transcription Factor 3/genetics , Animals , Calcitonin Gene-Related Peptide/metabolism , Dose-Response Relationship, Drug , Epoxide Hydrolases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Plant Lectins/genetics , Plant Lectins/metabolism , Plant Oils/pharmacology , Promoter Regions, Genetic , Vitamin E/pharmacology
7.
Cell Stem Cell ; 13(4): 433-45, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24094324

ABSTRACT

Sox2(+) adult mouse pituitary cells can self-renew and terminally differentiate in vitro, but their physiological role in vivo and possible contribution to oncogenesis remain largely unknown. Using genetic lineage tracing, we show here that the Sox2(+) cell compartment of both the embryonic and adult pituitary contains stem/progenitor cells that are able to differentiate into all hormone-producing lineages and contribute to organ homeostasis during postnatal life. In addition, we show that targeted expression of oncogenic ß-catenin in Sox2(+) cells gives rise to pituitary tumors, but, unexpectedly, the tumor mass is not derived from the Sox2(+) mutation-sustaining cells, suggesting a paracrine role of Sox2(+) cells in pituitary oncogenesis. Our data therefore provide in vivo evidence of a role for Sox2(+) stem/progenitor cells in long-term physiological maintenance of the adult pituitary, and highlight an unexpected non-cell-autonomous role for these cells in the induction of pituitary tumors.


Subject(s)
Homeostasis , Pituitary Gland/cytology , Pituitary Neoplasms/pathology , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cells, Cultured , Mice , Mice, Transgenic , Mutation , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , SOXB1 Transcription Factors/genetics
8.
J Clin Invest ; 122(10): 3635-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945632

ABSTRACT

Sex-determining region Y (SRY) box 2 (SOX2) haploinsufficiency causes a form of hypopituitarism in humans that is characterized by gonadotrophin deficiency known as hypogonadotrophic hypogonadism. Here, we conditionally deleted Sox2 in mice to investigate the pathogenesis of hypogonadotrophic hypogonadism. First, we found that absence of SOX2 in the developing Rathke pouch of conditional embryos led to severe anterior lobe hypoplasia with drastically reduced expression of the pituitary-specific transcription factor POU class 1 homeobox 1 (POU1F1) as well as severe disruption of somatotroph and thyrotroph differentiation. In contrast, corticotrophs, rostral-tip POU1F1-independent thyrotrophs, and, interestingly, lactotrophs and gonadotrophs were less affected. Second, we identified a requirement for SOX2 in normal proliferation of periluminal progenitors; in its absence, insufficient precursors were available to produce all cell lineages of the anterior pituitary. Differentiated cells derived from precursors exiting cell cycle at early stages, including corticotrophs, rostral-tip thyrotrophs, and gonadotrophs, were generated, while hormone-producing cells originating from late-born precursors, such as somatotrophs and POU1F1-dependent thyrotrophs, were severely reduced. Finally, we found that 2 previously characterized patients with SOX2 haploinsufficiency and associated hypogonadotrophic hypogonadism had a measurable response to gonadotropin-releasing hormone (GnRH) stimulation, suggesting that it is not the absence of gonadotroph differentiation, but rather the deficient hypothalamic stimulation of gonadotrophs, that underlies typical hypogonadotrophic hypogonadism.


Subject(s)
Hypogonadism/genetics , Hypothalamo-Hypophyseal System/physiology , SOXB1 Transcription Factors/physiology , Animals , Cell Differentiation , Cell Lineage , Female , Gene Expression Regulation, Developmental , Gonadotropin-Releasing Hormone/therapeutic use , Heterozygote , Homeodomain Proteins/genetics , Humans , Hypogonadism/drug therapy , Hypogonadism/physiopathology , Mice , Mice, Knockout , Organogenesis/genetics , Organogenesis/physiology , Pituitary Gland, Anterior/abnormalities , Pituitary Gland, Anterior/embryology , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Repressor Proteins/genetics , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Somatotrophs/pathology , Thyrotrophs/pathology , Transcription Factor Pit-1/deficiency
9.
Development ; 138(22): 4931-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22007134

ABSTRACT

The Wnt/ß-catenin pathway plays an essential role during regionalisation of the vertebrate neural plate and its inhibition in the most anterior neural ectoderm is required for normal forebrain development. Hesx1 is a conserved vertebrate-specific transcription factor that is required for forebrain development in Xenopus, mice and humans. Mouse embryos deficient for Hesx1 exhibit a variable degree of forebrain defects, but the molecular mechanisms underlying these defects are not fully understood. Here, we show that injection of a hesx1 morpholino into a 'sensitised' zygotic headless (tcf3) mutant background leads to severe forebrain and eye defects, suggesting an interaction between Hesx1 and the Wnt pathway during zebrafish forebrain development. Consistent with a requirement for Wnt signalling repression, we highlight a synergistic gene dosage-dependent interaction between Hesx1 and Tcf3, a transcriptional repressor of Wnt target genes, to maintain anterior forebrain identity during mouse embryogenesis. In addition, we reveal that Tcf3 is essential within the neural ectoderm to maintain anterior character and that its interaction with Hesx1 ensures the repression of Wnt targets in the developing forebrain. By employing a conditional loss-of-function approach in mouse, we demonstrate that deletion of ß-catenin, and concomitant reduction of Wnt signalling in the developing anterior forebrain of Hesx1-deficient embryos, leads to a significant rescue of the forebrain defects. Finally, transcriptional profiling of anterior forebrain precursors from mouse embryos expressing eGFP from the Hesx1 locus provides molecular evidence supporting a novel function of Hesx1 in mediating repression of Wnt/ß-catenin target activation in the developing forebrain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Prosencephalon/embryology , Repressor Proteins/physiology , Wnt Signaling Pathway/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Down-Regulation/genetics , Embryo, Mammalian , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Prosencephalon/metabolism , Prosencephalon/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/genetics , beta Catenin/physiology
10.
J Clin Endocrinol Metab ; 96(10): E1709-18, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21832120

ABSTRACT

CONTEXT: Fibroblast growth factor (FGF) 8 is important for GnRH neuronal development with human mutations resulting in Kallmann syndrome. Murine data suggest a role for Fgf8 in hypothalamo-pituitary development; however, its role in the etiology of wider hypothalamo-pituitary dysfunction in humans is unknown. OBJECTIVE: The objective of this study was to screen for FGF8 mutations in patients with septo-optic dysplasia (n = 374) or holoprosencephaly (HPE)/midline clefts (n = 47). METHODS: FGF8 was analyzed by PCR and direct sequencing. Ethnically matched controls were then screened for mutated alleles (n = 480-686). Localization of Fgf8/FGF8 expression was analyzed by in situ hybridization in developing murine and human embryos. Finally, Fgf8 hypomorphic mice (Fgf8(loxPNeo/-)) were analyzed for the presence of forebrain and hypothalamo-pituitary defects. RESULTS: A homozygous p.R189H mutation was identified in a female patient of consanguineous parentage with semilobar HPE, diabetes insipidus, and TSH and ACTH insufficiency. Second, a heterozygous p.Q216E mutation was identified in a female patient with an absent corpus callosum, hypoplastic optic nerves, and Moebius syndrome. FGF8 was expressed in the ventral diencephalon and anterior commissural plate but not in Rathke's pouch, strongly suggesting early onset hypothalamic and corpus callosal defects in these patients. This was consolidated by significantly reduced vasopressin and oxytocin staining neurons in the hypothalamus of Fgf8 hypomorphic mice compared with controls along with variable hypothalamo-pituitary defects and HPE. CONCLUSION: We implicate FGF8 in the etiology of recessive HPE and potentially septo-optic dysplasia/Moebius syndrome for the first time to our knowledge. Furthermore, FGF8 is important for the development of the ventral diencephalon, hypothalamus, and pituitary.


Subject(s)
Craniofacial Abnormalities/genetics , Fibroblast Growth Factor 8/genetics , Holoprosencephaly/genetics , Hypothalamic Diseases/genetics , Hypothalamo-Hypophyseal System/physiopathology , Mutation/physiology , Pituitary Diseases/genetics , Agenesis of Corpus Callosum/complications , Agenesis of Corpus Callosum/genetics , Arginine Vasopressin/metabolism , DNA Mutational Analysis , Female , Fibroblast Growth Factor 8/physiology , Human Growth Hormone/blood , Humans , Hydrocortisone/blood , Immunohistochemistry , In Situ Hybridization , Infant , Magnetic Resonance Imaging , Pituitary Gland/growth & development , Pituitary Gland/physiology , Prosencephalon/growth & development , Prosencephalon/physiology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Septo-Optic Dysplasia/genetics , Thyrotropin/blood
11.
Proc Natl Acad Sci U S A ; 108(28): 11482-7, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21636786

ABSTRACT

Wingless (Wnt)/ß-catenin signaling plays an essential role during normal development, is a critical regulator of stem cells, and has been associated with cancer in many tissues. Here we demonstrate that genetic expression of a degradation-resistant mutant form of ß-catenin in early Rathke's pouch (RP) progenitors leads to pituitary hyperplasia and severe disruption of the pituitary-specific transcription factor 1-lineage differentiation resulting in extreme growth retardation and hypopituitarism. Mutant mice mostly die perinatally, but those that survive weaning develop lethal pituitary tumors, which closely resemble human adamantinomatous craniopharyngioma, an epithelial tumor associated with mutations in the human ß-catenin gene. The tumorigenic effect of mutant ß-catenin is observed only when expressed in undifferentiated RP progenitors, but tumors do not form when committed or differentiated cells are targeted to express this protein. Analysis of affected pituitaries indicates that expression of mutant ß-catenin leads to a significant increase in the total numbers of pituitary progenitor/stem cells as well as in their proliferation potential. Our findings provide insights into the role of the Wnt pathway in normal pituitary development and demonstrate a causative role for mutated ß-catenin in an undifferentiated RP progenitor in the genesis of murine and human craniopharyngioma.


Subject(s)
Pituitary Gland/cytology , Pituitary Gland/metabolism , Pituitary Neoplasms/etiology , Pituitary Neoplasms/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Animals , Cell Differentiation , Craniopharyngioma/etiology , Craniopharyngioma/genetics , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Mutant Strains , Mutant Proteins/genetics , Mutant Proteins/metabolism , Pituitary Gland/growth & development , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
12.
Dis Model Mech ; 1(4-5): 241-54, 2008.
Article in English | MEDLINE | ID: mdl-19093031

ABSTRACT

A homozygous substitution of the highly conserved isoleucine at position 26 by threonine (I26T) in the transcriptional repressor HESX1 has been associated with anterior pituitary hypoplasia in a human patient, with no forebrain or eye defects. Two individuals carrying a homozygous substitution of the conserved arginine at position 160 by cysteine (R160C) manifest septo-optic dysplasia (SOD), a condition characterised by pituitary abnormalities associated with midline telencephalic structure defects and optic nerve hypoplasia. We have generated two knock-in mouse models containing either the I26T or R160C substitution in the genomic locus. Hesx1(I26T/I26T) embryos show pituitary defects comparable with Hesx1(-/-) mouse mutants, with frequent occurrence of ocular abnormalities, although the telencephalon develops normally. Hesx1(R160C/R160C) mutants display forebrain and pituitary defects that are identical to those observed in Hesx1(-/-) null mice. We also show that the expression pattern of HESX1 during early human development is very similar to that described in the mouse, suggesting that the function of HESX1 is conserved between the two species. Together, these results suggest that the I26T mutation yields a hypomorphic allele, whereas R160C produces a null allele and, consequently, a more severe phenotype in both mice and humans.


Subject(s)
Disease Models, Animal , Homeodomain Proteins/genetics , Hypopituitarism/genetics , Mutation , Repressor Proteins/genetics , Septo-Optic Dysplasia/genetics , Animals , Body Patterning , Mice , Prosencephalon/abnormalities , Prosencephalon/embryology
13.
Dev Biol ; 324(2): 322-33, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18775421

ABSTRACT

Hesx1 has been shown to be essential for normal pituitary development. The homeobox gene Six3 is expressed in the developing pituitary gland during mouse development but its function in this tissue has been precluded by the fact that in the Six3-deficient embryos the pituitary gland is not induced. To gain insights into the function of Six3 during pituitary development we have generated Six3+/- ;Hesx1Cre/+ double heterozygous mice. Strikingly, these mice show marked dwarfism, which is first detectable around weaning, and die by the 5th-6th week of age. Thyroid and gonad development is also impaired in these animals. Analysis of Six3+/- ;Hesx1Cre/+ compound embryos indicates that hypopituitarism is the likely cause of these defects since pituitary development is severely impaired in these mutants. Similar to the Hesx1-deficient embryos, Rathke's pouch is initially expanded in Six3+/- ;Hesx1Cre/+ compound embryos due to an increase in cell proliferation. Subsequently, the anterior pituitary gland appears bifurcated, dysmorphic and occasionally ectopically misplaced in the nasopharyngeal cavity, but cell differentiation is unaffected. Our research has revealed a role for Six3 in normal pituitary development, which has likely been conserved during evolution as SIX3 is also expressed in the pituitary gland of the human embryo.


Subject(s)
Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pituitary Gland, Anterior/embryology , Animals , Cell Differentiation , Cell Proliferation , Dwarfism, Pituitary/genetics , Eye Proteins/genetics , Genes, Homeobox , Genotype , Gonads/abnormalities , Gonads/cytology , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mutation , Nerve Tissue Proteins/genetics , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Repressor Proteins , Thyroid Gland/abnormalities , Thyroid Gland/cytology , Homeobox Protein SIX3
14.
Biochim Biophys Acta ; 1783(1): 131-43, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17931718

ABSTRACT

Hesx1 is a highly conserved homeobox gene present in vertebrates, but absent from invertebrates. Gene targeting experiments in mice have shown that this transcriptional repressor is required for normal forebrain and pituitary development. In humans, mutations in HESX1 impairing either its repressing activity or DNA binding properties lead to a comparable phenotype to that observed in Hesx1 deficient mice. In an attempt to gain insights into the molecular function of HESX1, we have performed a yeast two-hybrid screen and identified DNA methyltransferase 1 (DNMT1) as a HESX1 binding protein. We show that Dnmt1 is co-expressed with Hesx1 within the anterior forebrain and in the developing Rathke's pouch. Mapping of the interacting regions indicates that the entire HESX1 protein is required to establish binding to a portion of the N-terminus of DNMT1 and its catalytic domain in the C-terminus. The HESX1-DNMT1 complexes can be immunoprecipitated in cells and co-localise in the nucleus. These results establish a link between HESX1 and DNMT1 and suggest a novel mechanism for the repressing properties of HESX1.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/genetics , Animals , Cell Line , Cricetinae , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Protein Binding , Repressor Proteins/genetics , Two-Hybrid System Techniques
15.
Development ; 134(8): 1499-508, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17360769

ABSTRACT

The homeobox gene Hesx1 is an essential repressor that is required within the anterior neural plate for normal forebrain development in mouse and humans. Combining genetic cell labelling and marker analyses, we demonstrate that the absence of Hesx1 leads to a posterior transformation of the anterior forebrain (AFB) during mouse development. Our data suggest that the mechanism underlying this transformation is the ectopic activation of Wnt/beta-catenin signalling within the Hesx1 expression domain in the AFB. When ectopically expressed in the developing mouse embryo, Hesx1 alone cannot alter the normal fate of posterior neural tissue. However, conditional expression of Hesx1 within the AFB can rescue the forebrain defects observed in the Hesx1 mutants. The results presented here provide new insights into the function of Hesx1 in forebrain formation.


Subject(s)
Homeodomain Proteins/physiology , Prosencephalon/physiology , Animals , Axin Protein , Cell Differentiation , Cell Line , Cell Lineage , Cytoskeletal Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Repressor Proteins , Transcription Factors/metabolism , Wnt1 Protein/metabolism , beta Catenin/metabolism , Homeobox Protein SIX3
16.
Development ; 134(4): 789-99, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17229766

ABSTRACT

Planar-cell-polarity (PCP) signalling is necessary for initiation of neural tube closure in higher vertebrates. In mice with PCP gene mutations, a broad embryonic midline prevents the onset of neurulation through wide spacing of the neural folds. In order to evaluate the role of convergent extension in this defect, we vitally labelled the midline of loop-tail (Lp) embryos mutant for the PCP gene Vangl2. Injection of DiI into the node, and electroporation of a GFP expression vector into the midline neural plate, revealed defective convergent extension in both axial mesoderm and neuroepithelium, before the onset of neurulation. Chimeras containing both wild-type and Lp-mutant cells exhibited mainly wild-type cells in the midline neural plate and notochordal plate, consistent with a cell-autonomous disturbance of convergent extension. Inhibitor studies in whole-embryo culture demonstrated a requirement for signalling via RhoA-Rho kinase, but not jun N-terminal kinase, in convergent extension and the onset of neural tube closure. These findings identify a cell-autonomous defect of convergent extension, requiring PCP signalling via RhoA-Rho kinase, during the development of severe neural tube defects in the mouse.


Subject(s)
Cell Polarity/physiology , Central Nervous System/growth & development , Nerve Tissue Proteins/genetics , Neural Tube Defects/genetics , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Animals , Body Patterning , Central Nervous System/embryology , Embryonic Development , Mice , Mutation
17.
Dev Biol ; 290(1): 44-56, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16364283

ABSTRACT

Little is known about the mechanism by which embryonic liver, lung, and pancreas progenitor cells emerge from the endodermal epithelium to initiate organogenesis. Understanding this process and its genetic control provides insight into ontogeny, developmental abnormalities, and tissue regeneration. We find that shortly after hepatic endoderm cells are specified, they undergo a transition from a columnar, gut morphology to a pseudostratified morphology, with concomitant "interkinetic nuclear migration" (INM) during cell division. INM is a hallmark of pseudostratified epithelia and the process used by neural progenitors to emerge from the neural epithelium. We find that the transition of the hepatic endoderm, but not the neural epithelium, to a pseudostratified epithelium is dependent upon the cell-autonomous activity of the homeobox gene Hex. In the absence of Hex, hepatic endoderm cells survive but maintain a columnar, simple epithelial phenotype and ectopically express Shh and other genes characteristic of the midgut epithelium. Thus, Hex promotes endoderm organogenesis by promoting the transition to a pseudostratified epithelium, which in turn allows hepatoblasts to emerge into the stromal environment and continue differentiating.


Subject(s)
Endoderm/cytology , Homeodomain Proteins/physiology , Liver/embryology , Organogenesis , Stromal Cells/cytology , Transcription Factors/physiology , Animals , Body Patterning , Cell Differentiation/physiology , Cell Proliferation , Endoderm/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins , Homeodomain Proteins/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Stromal Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics
18.
Genesis ; 43(3): 129-35, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16267821

ABSTRACT

We generated a ROSA26-eGFP-DTA mouse line by introducing an eGFP-DTA (enhanced green fluorescent protein -- diphtheria toxin fragment A) cassette into the ROSA26 locus by homologous recombination in ES cells. This mouse expresses eGFP ubiquitously, but DTA expression is prevented by the presence of eGFP, a Neo cassette, and a strong transcriptional stop sequence. Mice carrying this construct are normal and fertile, indicating the absence of DTA expression. However, upon Cre-mediated excision of the floxed region DTA expression is activated, resulting in the specific ablation of Cre-expressing cells. As an example of this approach, we ablated Nkx2.5 and Wnt1-expressing cells by using the Nkx2.5-Cre and Wnt1-Cre mouse lines, respectively. We observed loss of the precise tissues in which Nkx2.5 and Wnt1 are expressed. Apart from being a general GFP reporter, the ROSA26-GFP-DTA mouse line should provide a useful resource for genetic ablation of specific groups of cells.


Subject(s)
Diphtheria Toxin/genetics , Gene Expression Regulation, Developmental , Peptide Fragments/genetics , Animals , Brain/embryology , Cell Death , Diphtheria Toxin/metabolism , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart/embryology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hybridization, Genetic , In Situ Hybridization , Mice , Mice, Knockout , Mice, Transgenic , Peptide Fragments/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
19.
Dev Biol ; 264(2): 537-49, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14651936

ABSTRACT

The relation between the role of the organizer at the gastrula stage and the activity of earlier signals in the specification, maintenance, and regionalization of the developing brain anlage is still controversial. Mouse embryos homozygous for null mutation in the cripto gene die at about 9.0 days postcoitum (d.p.c.) and fail to gastrulate and to form the node (the primary organizer). Here, we study the presence and the distribution of anterior neural plate molecular domains in cripto null mutants. We demonstrate that, in cripto(-/-) embryos, the main prosencephalic and mesencephalic regions are present and that they assume the correct topological organization. The identity of the anterior neural domains is maintained in mutant embryos at 8.5 d.p.c., as well as in mutant explants dissected at 8.5 d.p.c. and cultured in vitro for 24 h. Our data imply the existence of a stable neural regionalization of anterior character inside the cripto(-/-) embryos, despite the failure in both the gastrulation process and node formation. These results suggest that, in mouse embryos, the specification of the anterior neural identities can be maintained without an absolute requirement for the embryonic mesoderm and the node.


Subject(s)
Epidermal Growth Factor , Gastrula/physiology , Membrane Glycoproteins , Neoplasm Proteins/physiology , Nervous System/embryology , Organizers, Embryonic/physiology , Animals , Ectoderm/physiology , Female , Mesencephalon/embryology , Mesoderm/physiology , Mice , Mice, Inbred C57BL , Prosencephalon/embryology
20.
J Neurosci ; 23(10): 4199-207, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12764108

ABSTRACT

Midbrain dopaminergic and hindbrain serotonergic neurons play an important role in the modulation of behavior and are involved in a series of neuropsychiatric disorders. Despite the importance of these cells, little is known about the molecular mechanisms governing their development. During embryogenesis, midbrain dopaminergic neurons are specified rostral to the midbrain-hindbrain organizer (MHO), and hindbrain serotonergic neurons are specified caudal to it. We report that in transgenic mice in which Otx2 and accordingly the MHO are shifted caudally, the midbrain dopaminergic neuronal population expands to the ectopically positioned MHO and is enlarged. Complementary, the extension of the hindbrain serotonergic cell group is decreased. These changes are preserved in adulthood, and the additional, ectopic dopaminergic neurons project to the striatum, which is a proper dopaminergic target area. In addition, in mutants in which Otx2 and the MHO are shifted rostrally, dopaminergic and serotonergic neurons are relocated at the newly positioned MHO. However, in these mice, the size ratio between these two cell populations is changed in favor of the serotonergic cell population. To investigate whether the position of the MHO during embryogenesis is also of functional relevance for adult behavior, we tested mice with a caudally shifted MHO and report that these mutants show a higher locomotor activity. Together, we provide evidence that the position of the MHO determines the location and size of midbrain dopaminergic and hindbrain serotonergic cell populations in vivo. In addition, our data suggest that the position of the MHO during embryogenesis can modulate adult locomotor activity.


Subject(s)
Dopamine , Mesencephalon/anatomy & histology , Mesencephalon/physiology , Neurons/cytology , Neurons/physiology , Organizers, Embryonic/anatomy & histology , Organizers, Embryonic/physiology , Rhombencephalon/anatomy & histology , Rhombencephalon/physiology , Serotonin , Animals , Behavior, Animal/physiology , Brain Mapping/methods , Dopamine/physiology , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Male , Mesencephalon/cytology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Organizers, Embryonic/cytology , Otx Transcription Factors , Rhombencephalon/cytology , Serotonin/physiology , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...