Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Haematologica ; 104(7): 1332-1341, 2019 07.
Article in English | MEDLINE | ID: mdl-30630974

ABSTRACT

The nucleophosmin 1 gene (NPM1) is the most frequently mutated gene in acute myeloid leukemia. Notably, NPM1 mutations are always accompanied by additional mutations such as those in cohesin genes RAD21, SMC1A, SMC3, and STAG2 but not in the cohesin regulator, nipped B-like (NIPBL). In this work, we analyzed a cohort of adult patients with acute myeloid leukemia and NPM1 mutation and observed a specific reduction in the expression of NIPBL but not in other cohesin genes. In our zebrafish model, overexpression of the mutated form of NPM1 also induced downregulation of nipblb, the zebrafish ortholog of human NIPBL To investigate the hematopoietic phenotype and the interaction between mutated NPM1 and nipblb, we generated a zebrafish model with nipblb downregulation which showed an increased number of myeloid progenitors. This phenotype was due to hyper-activation of the canonical Wnt pathway: myeloid cells blocked in an undifferentiated state could be rescued when the Wnt pathway was inhibited by dkk1b mRNA injection or indomethacin administration. Our results reveal, for the first time, a role for NIPBL during zebrafish hematopoiesis and suggest that an interplay between NIPBL/NPM1 may regulate myeloid differentiation in zebrafish and humans through the canonical Wnt pathway and that dysregulation of these interactions may drive leukemic transformation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/pathology , Mutation , Nuclear Proteins/genetics , Adult , Animals , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nucleophosmin , Phenotype , Wnt Signaling Pathway , Zebrafish , Cohesins
2.
J Cell Physiol ; 233(2): 1455-1467, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28542953

ABSTRACT

During embryonic development, new arteries, and veins form from preexisting vessels in response to specific angiogenic signals. Angiogenic signaling is complex since not all endothelial cells exposed to angiogenic signals respond equally. Some cells will be selected to become tip cells and acquire migration and proliferation capacity necessary for vessel growth while others, the stalk cells become trailer cells that stay connected with pre-existing vessels and act as a linkage to new forming vessels. Additionally, stalk and tip cells have the capacity to interchange their roles. Stalk and tip cellular responses are mediated in part by the interactions of components of the Delta/Notch and Vegf signaling pathways. We have identified in zebrafish, that the transmembrane protein Tmem230a is a novel regulator of angiogenesis by its capacity to regulate the number of the endothelial cells in intersegmental vessels by co-operating with the Delta/Notch signaling pathway. Modulation of Tmem230a expression by itself is sufficient to rescue improper number of endothelial cells induced by aberrant expression or inhibition of the activity of genes associated with the Dll4/Notch pathway in zebrafish. Therefore, Tmem230a may have a modulatory role in vessel-network formation and growth. As the Tmem230 sequence is conserved in human, Tmem230 may represent a promising novel target for drug discovery and for disease therapy and regenerative medicine in promoting or restricting angiogenesis.


Subject(s)
Cell Proliferation , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Conserved Sequence , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Receptors, Notch/genetics , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Nat Mater ; 16(5): 587-596, 2017 05.
Article in English | MEDLINE | ID: mdl-28135264

ABSTRACT

Dynamics of epithelial monolayers has recently been interpreted in terms of a jamming or rigidity transition. How cells control such phase transitions is, however, unknown. Here we show that RAB5A, a key endocytic protein, is sufficient to induce large-scale, coordinated motility over tens of cells, and ballistic motion in otherwise kinetically arrested monolayers. This is linked to increased traction forces and to the extension of cell protrusions, which align with local velocity. Molecularly, impairing endocytosis, macropinocytosis or increasing fluid efflux abrogates RAB5A-induced collective motility. A simple model based on mechanical junctional tension and an active cell reorientation mechanism for the velocity of self-propelled cells identifies regimes of monolayer dynamics that explain endocytic reawakening of locomotion in terms of a combination of large-scale directed migration and local unjamming. These changes in multicellular dynamics enable collectives to migrate under physical constraints and may be exploited by tumours for interstitial dissemination.


Subject(s)
Endocytosis , Epithelium/metabolism , Biomechanical Phenomena , Cell Line, Tumor , Cell Membrane/metabolism , Humans , rab5 GTP-Binding Proteins/metabolism
4.
Nature ; 473(7346): 234-8, 2011 05 12.
Article in English | MEDLINE | ID: mdl-21499261

ABSTRACT

Notch signalling is a key intercellular communication mechanism that is essential for cell specification and tissue patterning, and which coordinates critical steps of blood vessel growth. Although subtle alterations in Notch activity suffice to elicit profound differences in endothelial behaviour and blood vessel formation, little is known about the regulation and adaptation of endothelial Notch responses. Here we report that the NAD(+)-dependent deacetylase SIRT1 acts as an intrinsic negative modulator of Notch signalling in endothelial cells. We show that acetylation of the Notch1 intracellular domain (NICD) on conserved lysines controls the amplitude and duration of Notch responses by altering NICD protein turnover. SIRT1 associates with NICD and functions as a NICD deacetylase, which opposes the acetylation-induced NICD stabilization. Consequently, endothelial cells lacking SIRT1 activity are sensitized to Notch signalling, resulting in impaired growth, sprout elongation and enhanced Notch target gene expression in response to DLL4 stimulation, thereby promoting a non-sprouting, stalk-cell-like phenotype. In vivo, inactivation of Sirt1 in zebrafish and mice causes reduced vascular branching and density as a consequence of enhanced Notch signalling. Our findings identify reversible acetylation of the NICD as a molecular mechanism to adapt the dynamics of Notch signalling, and indicate that SIRT1 acts as rheostat to fine-tune endothelial Notch responses.


Subject(s)
Endothelial Cells/enzymology , Gene Expression Regulation , Receptors, Notch/metabolism , Signal Transduction/physiology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Acetylation , Animals , Endothelial Cells/cytology , Gene Knockout Techniques , Gene Silencing , HEK293 Cells , Humans , Mice , Mutation , Receptor, Notch1/metabolism , Zebrafish/embryology , Zebrafish/genetics
5.
Dev Biol ; 343(1-2): 94-103, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20423710

ABSTRACT

Segmentation is a key step in embryonic development. Acting in all germ layers, it is responsible for the generation of antero-posterior asymmetries. Hox genes, with their diverse expression in individual segments, are fundamental players in the determination of different segmental fates. In vertebrates, Hox gene products gain specificity for DNA sequences by interacting with Pbx, Prep and Meis homeodomain transcription factors. In this work we cloned and analysed prep1.2 in zebrafish. In-situ hybridization experiments show that prep1.2 is maternally and ubiquitously expressed up to early somitogenesis when its expression pattern becomes more restricted to the head and trunk mesenchyme. Experiments of loss of function with prep1.2 morpholinos change the shape of the hyoid and third pharyngeal cartilages while arches 4-7 and pectoral fins are absent, a phenotype strikingly similar to that caused by loss of retinoic acid (RA). In fact, we show that prep1.2 is positively regulated by RA and required for the normal expression of aldh1a2 at later stages, particularly in tissues involved in the development of the branchial arches and pectoral fins. Thus, prep1.2 and aldh1a2 are members of an indirect positive feedback loop required for pharyngeal endoderm and posterior branchial arches development. As the paralogue gene prep1.1 is more important in hindbrain patterning and neural crest chondrogenesis, we provide evidence of a functional specialization of prep genes in zebrafish head segmentation and morphogenesis.


Subject(s)
Branchial Region/embryology , Retinal Dehydrogenase/genetics , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Body Patterning , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Retinal Dehydrogenase/metabolism , Transcription Factors/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
6.
Sci Rep ; 11(1): 671, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436662

ABSTRACT

We developed a novel reporter transgenic zebrafish model called MITO-Luc/GFP zebrafish in which GFP and luciferase expression are under the control of the master regulator of proliferation NF-Y. In MITO-Luc/GFP zebrafish it is possible to visualize cell proliferation in vivo by fluorescence and bioluminescence. In this animal model, GFP and luciferase expression occur in early living embryos, becoming tissue specific in juvenile and adult zebrafish. By in vitro and ex vivo experiments we demonstrate that luciferase activity in adult animals occurs in intestine, kidney and gonads, where detectable proliferating cells are located. Further, by time lapse experiments in live embryos, we observed a wave of GFP positive cells following fin clip. In adult zebrafish, in addition to a bright bioluminescence signal on the regenerating tail, an early unexpected signal coming from the kidney occurs indicating not only a fin cell proliferation, but also a systemic response to tissue damage. Finally, we observed that luciferase activity was inhibited by anti-proliferative interventions, i.e. 5FU, cell cycle inhibitors and X-Rays. In conclusion, MITO-Luc/GFP zebrafish is a novel animal model that may be crucial to assess the spatial and temporal evolution of cell proliferation in vivo.


Subject(s)
Animals, Genetically Modified/growth & development , Cell Proliferation , Evolution, Molecular , Green Fluorescent Proteins/metabolism , Luciferases/metabolism , Spatio-Temporal Analysis , Zebrafish/growth & development , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Green Fluorescent Proteins/genetics , Luciferases/genetics , Regeneration , Zebrafish/genetics , Zebrafish/metabolism
7.
Nat Commun ; 12(1): 4872, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381052

ABSTRACT

The Netrin-1 receptor UNC5B is an axon guidance regulator that is also expressed in endothelial cells (ECs), where it finely controls developmental and tumor angiogenesis. In the absence of Netrin-1, UNC5B induces apoptosis that is blocked upon Netrin-1 binding. Here, we identify an UNC5B splicing isoform (called UNC5B-Δ8) expressed exclusively by ECs and generated through exon skipping by NOVA2, an alternative splicing factor regulating vascular development. We show that UNC5B-Δ8 is a constitutively pro-apoptotic splicing isoform insensitive to Netrin-1 and required for specific blood vessel development in an apoptosis-dependent manner. Like NOVA2, UNC5B-Δ8 is aberrantly expressed in colon cancer vasculature where its expression correlates with tumor angiogenesis and poor patient outcome. Collectively, our data identify a mechanism controlling UNC5B's necessary apoptotic function in ECs and suggest that the NOVA2/UNC5B circuit represents a post-transcriptional pathway regulating angiogenesis.


Subject(s)
Apoptosis , Blood Vessels/growth & development , Netrin Receptors/metabolism , RNA Isoforms/metabolism , Alternative Splicing , Animals , Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Endothelial Cells , Humans , Morphogenesis , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Netrin Receptors/genetics , Netrin-1/metabolism , Neuro-Oncological Ventral Antigen , RNA Isoforms/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Survival Analysis , Zebrafish
8.
Front Cell Dev Biol ; 8: 844, 2020.
Article in English | MEDLINE | ID: mdl-33015043

ABSTRACT

Histone deacetylase 8 (HDAC8), a class I HDAC that modifies non-histone proteins such as p53, is highly expressed in different hematological neoplasms including a subtype of acute myeloid leukemia (AML) bearing inversion of chromosome 16 [inv(16)]. To investigate HDAC8 contribution to hematopoietic stem cell maintenance and myeloid leukemic transformation, we generated a zebrafish model with Hdac8 overexpression and observed an increase in hematopoietic stem/progenitor cells, a phenotype that could be reverted using a specific HDAC8 inhibitor, PCI-34051 (PCI). In addition, we demonstrated that AML cell lines respond differently to PCI treatment: HDAC8 inhibition elicits cytotoxic effect with cell cycle arrest followed by apoptosis in THP-1 cells, and cytostatic effect in HL60 cells that lack p53. A combination of cytarabine, a standard anti-AML chemotherapeutic, with PCI resulted in a synergistic effect in all the cell lines tested. We, then, searched for a mechanism behind cell cycle arrest caused by HDAC8 inhibition in the absence of functional p53 and demonstrated an involvement of the canonical WNT signaling in zebrafish and in cell lines. Together, we provide the evidence for the role of HDAC8 in hematopoietic stem cell differentiation in zebrafish and AML cell lines, suggesting HDAC8 inhibition as a therapeutic target in hematological malignancies. Accordingly, we demonstrated the utility of a highly specific HDAC8 inhibition as a therapeutic strategy in combination with standard chemotherapy.

9.
Nat Commun ; 11(1): 3516, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665580

ABSTRACT

It is unclear whether the establishment of apical-basal cell polarity during the generation of epithelial lumens requires molecules acting at the plasma membrane/actin interface. Here, we show that the I-BAR-containing IRSp53 protein controls lumen formation and the positioning of the polarity determinants aPKC and podocalyxin. Molecularly, IRSp53 acts by regulating the localization and activity of the small GTPase RAB35, and by interacting with the actin capping protein EPS8. Using correlative light and electron microscopy, we further show that IRSp53 ensures the shape and continuity of the opposing plasma membrane of two daughter cells, leading to the formation of a single apical lumen. Genetic removal of IRSp53 results in abnormal renal tubulogenesis, with altered tubular polarity and architectural organization. Thus, IRSp53 acts as a membrane curvature-sensing platform for the assembly of multi-protein complexes that control the trafficking of apical determinants and the integrity of the luminal plasma membrane.


Subject(s)
Cell Membrane/metabolism , Nerve Tissue Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Cell Polarity/genetics , Cell Polarity/physiology , Epithelial Cells/metabolism , Female , Humans , Morphogenesis/genetics , Morphogenesis/physiology , Nerve Tissue Proteins/genetics , Protein Transport/genetics , Protein Transport/physiology , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , rab GTP-Binding Proteins/genetics
10.
Sci Rep ; 9(1): 1211, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718891

ABSTRACT

Homozygous mutations in SNAP29, encoding a SNARE protein mainly involved in membrane fusion, cause CEDNIK (Cerebral Dysgenesis, Neuropathy, Ichthyosis and Keratoderma), a rare congenital neurocutaneous syndrome associated with short life expectancy, whose pathogenesis is unclear. Here, we report the analysis of the first genetic model of CEDNIK in zebrafish. Strikingly, homozygous snap29 mutant larvae display CEDNIK-like features, such as microcephaly and skin defects. Consistent with Snap29 role in membrane fusion during autophagy, we observe accumulation of the autophagy markers p62 and LC3, and formation of aberrant multilamellar organelles and mitochondria. Importantly, we find high levels of apoptotic cell death during early development that might play a yet uncharacterized role in CEDNIK pathogenesis. Mutant larvae also display mouth opening problems, feeding impairment and swimming difficulties. These alterations correlate with defective trigeminal nerve formation and excess axonal branching. Since the paralog Snap25 is known to promote axonal branching, Snap29 might act in opposition with, or modulate Snap25 activity during neurodevelopment. Our vertebrate genetic model of CEDNIK extends the description in vivo of the multisystem defects due to loss of Snap29 and could provide the base to test compounds that might ameliorate traits of the disease.


Subject(s)
Keratoderma, Palmoplantar/metabolism , Neurocutaneous Syndromes/metabolism , SNARE Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Autophagy , Humans , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/physiopathology , Membrane Fusion , Models, Genetic , Mutation , Nervous System Malformations/metabolism , Neurocutaneous Syndromes/genetics , Neurocutaneous Syndromes/physiopathology , Phenotype , Protein Binding , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , SNARE Proteins/physiology , Synaptosomal-Associated Protein 25/metabolism , Synaptosomal-Associated Protein 25/physiology , Zebrafish/metabolism , Zebrafish Proteins/physiology
11.
Sci Rep ; 9(1): 1527, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30728389

ABSTRACT

Cystic fibrosis (CF) is a hereditary disease due to mutations in the CFTR gene and causes mortality in humans mainly due to respiratory infections caused by Pseudomonas aeruginosa. In a previous work we used phage therapy, which is a treatment with a mix of phages, to actively counteract acute P. aeruginosa infections in mice and Galleria mellonella larvae. In this work we apply phage therapy to the treatment of P. aeruginosa PAO1 infections in a CF zebrafish model. The structure of the CFTR channel is evolutionary conserved between fish and mammals and cftr-loss-of-function zebrafish embryos show a phenotype that recapitulates the human disease, in particular with destruction of the pancreas. We show that phage therapy is able to decrease lethality, bacterial burden, and the pro-inflammatory response caused by PAO1 infection. In addition, phage administration relieves the constitutive inflammatory state of CF embryos. To our knowledge, this is the first time that phage therapy is used to cure P. aeruginosa infections in a CF animal model. We also find that the curative effect against PAO1 infections is improved by combining phages and antibiotic treatments, opening a useful therapeutic approach that could reduce antibiotic doses and time of administration.


Subject(s)
Cystic Fibrosis/complications , Disease Models, Animal , Embryo, Nonmammalian/immunology , Pseudomonas Infections/therapy , Pseudomonas Phages/growth & development , Pseudomonas aeruginosa/virology , Respiratory Tract Infections/therapy , Animals , Anti-Bacterial Agents/therapeutic use , Embryo, Nonmammalian/microbiology , Embryo, Nonmammalian/virology , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/virology , Pseudomonas Phages/isolation & purification , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/physiology , Respiratory Tract Infections/microbiology , Zebrafish
12.
Dev Neurosci ; 30(1-3): 65-81, 2008.
Article in English | MEDLINE | ID: mdl-18075256

ABSTRACT

We used transgenic zebrafish expressing GFP or YFP in subpopulations of neurons to study the migration, homing process and axon extension of groups of CNS neurons in different regions of the zebrafish brain. We found that extensive migration takes place at all levels of the CNS and gives rise to nuclei or cell populations with specific identities. Here, we describe 4 previously unknown or only partially characterized migratory events taking place in the zebrafish telencephalon and rhombic lip, using 3 different transgenic lines, and identify the phenotypes of the cells undertaking these migrations. The migration of a subgroup of mitral cell precursors from the dorsocaudal telencephalon to the olfactory bulb, visualized in the tg(tbr1:YFP) transgenic line, is coupled with morphogenetic transformation of the dorsal telencephalon. The tg(1.4dlx5a-6a:GFP) transgenic line provides a means to analyze the migration of GABAergic interneurons from the ventral to the dorsal telencephalon, thus extending the occurrence of this migration to another vertebrate. The tg(Xeom:GFP) transgenic line provides the first demonstration of the dorsoventral migration of glutamatergic septal neurons, present in mammals and now described in fish, thus reconciling the contrasting evidence of dorsal patterning genes (tbr1, eomes) expressed in a ventral cell population. Furthermore, migration studies in the tg(1.4dlx5a-6a:GFP) and tg(Xeom:GFP) lines help determine the origin of 2 important cell populations in the fish cerebellum: projection neurons and Purkinje cells. These examples reinforce the concept that migratory events contribute to the distribution of cell types with diverse identities through the CNS and that zebrafish transgenic lines represent excellent tools to study these events.


Subject(s)
Cell Movement/physiology , Central Nervous System/embryology , Morphogenesis/physiology , Neurons/physiology , Stem Cells/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Lineage/genetics , Central Nervous System/cytology , Central Nervous System/physiology , Gene Expression Regulation, Developmental/genetics , Neurons/cytology , Stem Cells/cytology , Zebrafish/physiology
13.
Oncotarget ; 7(34): 55302-55312, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27486814

ABSTRACT

Nucleophosmin (NPM1) is a ubiquitous multifunctional phosphoprotein with both oncogenic and tumor suppressor functions. Mutations of the NPM1 gene are the most frequent genetic alterations in acute myeloid leukemia (AML) and result in the expression of a mutant protein with aberrant cytoplasmic localization, NPMc+. Although NPMc+ causes myeloproliferation and AML in animal models, its mechanism of action remains largely unknown. Here we report that NPMc+ activates canonical Wnt signaling during the early phases of zebrafish development and determines a Wnt-dependent increase in the number of progenitor cells during primitive hematopoiesis. Coherently, the canonical Wnt pathway is active in AML blasts bearing NPMc+ and depletion of the mutant protein in the patient derived OCI-AML3 cell line leads to a decrease in the levels of active ß-catenin and of Wnt target genes. Our results reveal a novel function of NPMc+ and provide insight into the molecular pathogenesis of AML bearing NPM1 mutations.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/physiology , Wnt Signaling Pathway/physiology , Zebrafish/embryology , Animals , Axin Protein/analysis , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/etiology , Mutation , Nuclear Proteins/genetics , Nucleophosmin
14.
Sci Rep ; 6: 30213, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27458029

ABSTRACT

Histone deacetylases (HDACs) catalyze the removal of acetyl molecules from histone and non-histone substrates playing important roles in chromatin remodeling and control of gene expression. Class I HDAC1 is a critical regulator of cell cycle progression, cellular proliferation and differentiation during development; it is also regulated by many post-translational modifications (PTMs). Herein we characterize a new mitosis-specific phosphorylation of HDAC1 driven by Aurora kinases A and B. We show that this phosphorylation affects HDAC1 enzymatic activity and it is critical for the maintenance of a proper proliferative and developmental plan in a complex organism. Notably, we find that Aurora-dependent phosphorylation of HDAC1 regulates histone acetylation by modulating the expression of genes directly involved in the developing zebrafish central nervous system. Our data represent a step towards the comprehension of HDAC1 regulation by its PTM code, with important implications in unravelling its roles both in physiology and pathology.


Subject(s)
Aurora Kinases/metabolism , Embryonic Development , Histone Deacetylase 1/metabolism , Mitosis , Zebrafish/embryology , Acetylation , Animals , Genes, Regulator , Histones/metabolism , Phosphorylation
15.
Mech Dev ; 115(1-2): 133-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12049777

ABSTRACT

We have identified the cDNAs of two new zebrafish preprosomatostatins, PPSS1 and PPSS3, in addition to the previously cloned PPSS2 (Argenton et al., 1999). PPSS1 is the orthologue of mammalian PPSSs, with a conserved C-terminal SS-14 sequence, PPSS2 is a divergent SS precursor and PPSS3 is a cortistatin-like prohormone. Using whole-mount in situ hybridisation, we have analysed the expression of PPSS1 and PPSS2 in zebrafish embryos up to 5 days post fertilisation. PPSS1 was expressed in the developing pancreas and central nervous system (CNS), whereas PPSS2 expression was exclusively pancreatic. In the CNS, PPSS1 was detected in several areas, in particular in the vagal motor nucleus and in cells that pioneer the tract of the postoptic commissure. PPSS1 was also expressed transiently in the telencephalon and spinal motor neurons. In all areas but the telencephalon PPSS1 was coexpressed with islet-1.


Subject(s)
Central Nervous System/metabolism , Gene Expression , Pancreas/metabolism , Protein Precursors/genetics , Repressor Proteins , Somatostatin/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Axin Protein , Base Sequence , Brain/embryology , Brain/metabolism , Central Nervous System/embryology , DNA, Complementary , Gene Expression Profiling , Humans , Molecular Sequence Data , Pancreas/embryology , Phylogeny , Protein Precursors/classification , Proteins/genetics , Sequence Homology, Amino Acid , Somatostatin/classification , Zebrafish/embryology , Zebrafish Proteins
16.
Mech Dev ; 121(2): 131-42, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15037315

ABSTRACT

Sox proteins are DNA-binding proteins belonging to the HMG box superfamily and they play key roles in animal embryonic development. Zebrafish Sox21a is part of group B Sox proteins and its chicken and mouse orthologs have been described as transcriptional repressor and activator, respectively, in two different target gene contexts. Zebrafish sox21a is present as a maternal transcript in the oocyte and is mainly expressed at the developing midbrain-hindbrain boundary from the onset of neurulation. In order to understand its role in vivo, we ectopically expressed sox21a by microinjection. Ectopic expression of full length sox21a leads to dorsalization of the embryos. A subset of the dorsalized embryos shows a partial axis splitting, and hence an ectopic neural tube, as an additional phenotype. At gastrulation, injected embryos show expansion of the expression domains of organizer-specific genes, such as chordin and goosecoid. Molecular markers used in somitogenesis highlight that sox21a-injected embryos have shortened AP axis, undulating axial structures, enlarged or even radialized paraxial territory. The developmental abnormalities caused by ectopic expression of sox21a are suggestive of defects in convergence-extension morphogenetic movements. Antisense morpholino oligonucleotides, designed to functionally knockdown sox21a, cause ventralization of the embryos. Moreover, gain-of-function experiments with chimeric constructs, where Sox21a DNA-binding domain is fused to a transcriptional activator (VP16) or repressor (EnR) domain, suggests that zebrafish Sox21a acts as a repressor in dorso-ventral patterning.


Subject(s)
DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Base Sequence , Body Patterning/genetics , Body Patterning/physiology , DNA-Binding Proteins/physiology , Female , Gastrula , Gene Expression , Gene Targeting , High Mobility Group Proteins/physiology , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Repressor Proteins/genetics , Repressor Proteins/physiology , SOXB2 Transcription Factors , Zebrafish/physiology , Zebrafish Proteins/physiology
17.
Nat Commun ; 6: 8479, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26446569

ABSTRACT

Vascular lumen formation is a fundamental step during angiogenesis; yet, the molecular mechanisms underlying this process are poorly understood. Recent studies have shown that neural and vascular systems share common anatomical, functional and molecular similarities. Here we show that the organization of endothelial lumen is controlled at the post-transcriptional level by the alternative splicing (AS) regulator Nova2, which was previously considered to be neural cell-specific. Nova2 is expressed during angiogenesis and its depletion disrupts vascular lumen formation in vivo. Similarly, Nova2 depletion in cultured endothelial cells (ECs) impairs the apical distribution and the downstream signalling of the Par polarity complex, resulting in altered EC polarity, a process required for vascular lumen formation. These defects are linked to AS changes of Nova2 target exons affecting the Par complex and its regulators. Collectively, our results reveal that Nova2 functions as an AS regulator in angiogenesis and is a novel member of the 'angioneurins' family.


Subject(s)
Alternative Splicing/physiology , Antigens, Neoplasm/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/physiology , Neovascularization, Physiologic/physiology , RNA-Binding Proteins/metabolism , Animals , Antigens, Neoplasm/genetics , Cells, Cultured , Mice , Neuro-Oncological Ventral Antigen , RNA-Binding Proteins/genetics
18.
Mol Oncol ; 8(2): 207-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24309677

ABSTRACT

Notch signaling in prominently involved in growth regulation in metazoan tissues. Because of this, Notch is often upregulated in cancer and current efforts point to developing drugs that block its activation. Notch receptor endocytosis towards acidic compartments is a recently appreciated determinant of signaling activation. Vacuolar H(+) ATPase (V-ATPase) is responsible for acidification of endocytic organelles and mutants in V-ATPase subunit encoding genes in model organisms have been recently shown to display loss of Notch signaling. Here, we show that administration of BafilomycinA1 (BafA1), a highly specific V-ATPase inhibitor decreases Notch signaling during Drosophila and Zebrafish development, and in human cells in culture. In normal breast cells, we find that BafA1 treatment leads to accumulation of Notch in the endo-lysosomal system, and reduces its processing and signaling activity. In Notch-addicted breast cancer cells, BafA1 treatment reduces growth in cells expressing membrane tethered forms of Notch, while sparing cells expressing cytoplasmic forms. In contrast, we find that V-ATPase inhibition reduces growth of leukemia cells, without affecting Notch activatory cleavage. However, consistent with the emerging roles of V-ATPase in controlling multiple signaling pathways, in these cells Akt activation is reduced, as it is also the case in BafA1-treated breast cancer cells. Our data support V-ATPase inhibition as a novel therapeutic approach to counteract tumor growth via signaling pathways regulated at the endo-lysosomal level.


Subject(s)
Breast Neoplasms/metabolism , Drosophila Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Macrolides/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Receptors, Notch/antagonists & inhibitors , Signal Transduction , Vacuolar Proton-Translocating ATPases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
19.
Dev Cell ; 31(4): 420-33, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25458010

ABSTRACT

Cells entering mitosis become rounded, lose attachment to the substrate, and increase their cortical rigidity. Pivotal to these events is the dismantling of focal adhesions (FAs). How mitotic reshaping is linked to commitment to divide is unclear. Here, we show that DEPDC1B, a protein that accumulates in G2, coordinates de-adhesion events and cell-cycle progression at mitosis. DEPDC1B functions as an inhibitor of a RhoA-based signaling complex, which assembles on the FA-associated protein tyrosine phosphatase, receptor type, F (PTPRF) and mediates the integrity of FAs. By competing with RhoA for the interaction with PTPRF, DEPDC1B promotes the dismantling of FAs, which is necessary for the morphological changes preceding mitosis. The circuitry is relevant in whole organisms, as shown by the control exerted by the DEPDC1B/RhoA/PTPRF axis on mitotic dynamics during zebrafish development. Our results uncover an adhesion-dependent signaling mechanism that coordinates adhesion events with the control of cell-cycle progression.


Subject(s)
Cell Cycle Proteins/metabolism , Mitosis/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Adhesion , Cells, Cultured , Humans , Phosphorylation , Signal Transduction/physiology
20.
PLoS One ; 5(12): e15047, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21203543

ABSTRACT

Prep1 is a developmentally essential TALE class homeodomain transcription factor. In zebrafish and mouse, Prep1 is already ubiquitously expressed at the earliest stages of development, with important tissue-specific peculiarities. The Prep1 gene in mouse is developmentally essential and has haploinsufficient tumor suppressor activity [1]. We have determined the human Prep1 transcription start site (TSS) by primer extension analysis and identified, within 20 bp, the transcription start region (TSR) of the zebrafish Prep1.1 promoter. The functions of the zebrafish 5' upstream sequences were analyzed both by transient transfections in Hela Cells and by injection in zebrafish embryos. This analysis revealed a complex promoter with regulatory sequences extending up to -1.8, possibly -5.0 Kb, responsible for tissue specific expression. Moreover, the first intron contains a conserved tissue-specific enhancer both in zebrafish and in human cells. Finally, a two nucleotides mutation of an EGR-1 site, conserved in all species including human and zebrafish and located at a short distance from the TSS, destroyed the promoter activity of the -5.0 Kb promoter. A transgenic fish expressing GFP under the -1.8 Kb zebrafish promoter/enhancer co-expressed GFP and endogenous Prep1.1 during embryonic development. In the adult fish, GFP was expressed in hematopoietic regions like the kidney, in agreement with the essential function of Prep1 in mouse hematopoiesis. Sequence comparison showed conservation from man to fish of the sequences around the TSS, within the first intron enhancer. Moreover, about 40% of the sequences spread throughout the 5 Kbof the zebrafish promoter are concentrated in the -3 to -5 Kb of the human upstream region.


Subject(s)
Homeodomain Proteins/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Binding Sites , Green Fluorescent Proteins/metabolism , Humans , Mice , Models, Genetic , Mutation , Nucleotides/chemistry , Protein Structure, Tertiary , Transcription Factors/chemistry , Transcription Initiation Site , Zebrafish , Zebrafish Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL