Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Neurosci Res ; 101(8): 1345-1359, 2023 08.
Article in English | MEDLINE | ID: mdl-37031448

ABSTRACT

Classical dynamins (DNMs) are GTPase proteins engaged in endocytosis, a fundamental process for cargo internalization from the plasma membrane. In mammals, three DNM genes are present with different expression patterns. DNM1 is expressed at high levels in neurons, where it takes place in the recycling of synaptic vesicles; DNM2 is ubiquitously expressed, while DNM3 is found in the brain and in the testis. Due to the conservation of genes in comparison to mammals, we took advantage of a zebrafish model for functional characterization of dnm1a, ortholog of mammalian DNM1. Our data strongly demonstrated that dnm1a has a nervous tissue-specific expression pattern and plays a role in the formation of both axon and synapse. This is the first in vivo study that collects evidence about the effects of dnm1a loss of function in zebrafish, thus providing a new excellent model to be used in different scientific fields.


Subject(s)
Nerve Tissue , Zebrafish , Animals , Male , Axons , Neurons/metabolism , Synapses/metabolism , Mammals
2.
Hum Mol Genet ; 28(1): 64-73, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30239720

ABSTRACT

Cornelia de Lange syndrome (CdLS), which is reported to affect ∼1 in 10 000 to 30 000 newborns, is a multisystem organ developmental disorder with relatively mild to severe effects. Among others, intellectual disability represents an important feature of this condition. CdLS can result from mutations in at least five genes: nipped-B-like protein, structural maintenance of chromosomes 1A, structural maintenance of chromosomes 3, RAD21 cohesin complex component and histone deacetylase 8 (HDAC8). It is believed that mutations in these genes cause CdLS by impairing the function of the cohesin complex (to which all the aforementioned genes contribute to the structure or function), disrupting gene regulation during critical stages of early development. Since intellectual disorder might result from alterations in neural development, in this work, we studied the role of Hdac8 gene in mouse neural stem cells (NSCs) and in vertebrate (Danio rerio) brain development by knockdown and chemical inhibition experiments. Underlying features of Hdac8 deficiency is an increased cell death in the developing neural tissues, either in mouse NSCs or in zebrafish embryos.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Proteins, Non-Histone/physiology , De Lange Syndrome/genetics , Histone Deacetylases/genetics , Animals , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromosomal Proteins, Non-Histone/genetics , De Lange Syndrome/physiopathology , Gene Expression Regulation/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Neural Stem Cells/physiology , Neurons/physiology , Phenotype , Repressor Proteins/genetics , Zebrafish , Zebrafish Proteins , Cohesins
3.
Nucleic Acids Res ; 47(3): 1278-1293, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30544196

ABSTRACT

SMYD3 is a methylase previously linked to cancer cell invasion and migration. Here we show that SMYD3 favors TGFß-induced epithelial-mesenchymal transition (EMT) in mammary epithelial cells, promoting mesenchymal and EMT transcription factors expression. SMYD3 directly interacts with SMAD3 but it is unnecessary for SMAD2/3 phosphorylation and nuclear translocation. Conversely, SMYD3 is indispensable for SMAD3 direct association to EMT genes regulatory regions. Accordingly, SMYD3 knockdown or its pharmacological blockade with the BCI121 inhibitor dramatically reduce TGFß-induced SMAD3 association to the chromatin. Remarkably, BCI121 treatment attenuates mesenchymal genes transcription in the mesenchymal-like MDA-MB-231 cell line and reduces their invasive ability in vivo, in a zebrafish xenograft model. In addition, clinical datasets analysis revealed that higher SMYD3 levels are linked to a less favorable prognosis in claudin-low breast cancers and to a reduced metastasis free survival in breast cancer patients. Overall, our data point at SMYD3 as a pivotal SMAD3 cofactor that promotes TGFß-dependent mesenchymal gene expression and cell migration in breast cancer, and support SMYD3 as a promising pharmacological target for anti-cancer therapy.


Subject(s)
Breast Neoplasms/genetics , Histone-Lysine N-Methyltransferase/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Chromatin/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Zebrafish
4.
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
5.
J Cell Physiol ; 233(7): 5396-5404, 2018 07.
Article in English | MEDLINE | ID: mdl-29215726

ABSTRACT

SEL1L (suppressor/enhancer of Lin-12-like) is a highly conserved gene associated with the endoplasmic reticulum-associated degradation (ERAD) pathway and involved in mediating the balance between stem cells self-renewal and differentiation of neural progenitors. It has been recently shown that SEL1L KO mice are embryonic lethal and display altered organogenesis. To better characterize the function of SEL1L in the early stages of embryonic development, we turned to the zebrafish model (Danio rerio). After exploring sel1l expression by RT-PCR and in situ hybridization, we employed a morpholino-mediated down-regulation approach. Results showed extensive impairments in the vasculature, which supports the mice knock-out findings.


Subject(s)
Embryonic Development/genetics , Endoplasmic Reticulum-Associated Degradation/genetics , Endoplasmic Reticulum/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Endoplasmic Reticulum/metabolism , Endothelium/cytology , Gene Expression Regulation, Developmental/genetics , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
6.
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
7.
Exp Cell Res ; 361(2): 236-245, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29107067

ABSTRACT

The CFDP1 proteins have been linked to craniofacial development and osteogenesis in vertebrates, though specific human syndromes have not yet been identified. Alterations of craniofacial development represent the main cause of infant disability and mortality in humans. For this reason, it is crucial to understand the cellular functions and mechanism of action of the CFDP1 protein in model vertebrate organisms. Using a combination of genomic, molecular and cell biology approaches, we have performed a functional analysis of the cfdp1 gene and its encoded protein, zCFDP1, in the zebrafish model system. We found that zCFDP1 is present in the zygote, is rapidly produced after MTZ transition and is highly abundant in the head structures. Depletion of zCFDP1, induced by an ATG-blocking morpholino, produces considerable defects in craniofacial structures and bone mineralization. Together, our results show that zCFDP1 is an essential protein required for proper development and provide the first experimental evidence showing that in vertebrates it actively participates to the morphogenesis of craniofacial territories.


Subject(s)
Gene Expression Regulation, Developmental , Osteogenesis/genetics , Phosphoproteins/genetics , Skull/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Calcification, Physiologic/genetics , Cloning, Molecular , Embryo, Mammalian , Morpholinos/genetics , Morpholinos/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Skull/growth & development , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
8.
J Cell Physiol ; 231(3): 613-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26206533

ABSTRACT

Genetic variants within components of the cohesin complex (NIPBL, SMC1A, SMC3, RAD21, PDS5, ESCO2, HDAC8) are believed to be responsible for a spectrum of human syndromes known as "cohesinopathies" that includes Cornelia de Lange Syndrome (CdLS). CdLS is a multiple malformation syndrome affecting almost any organ and causing severe developmental delay. Cohesinopathies seem to be caused by dysregulation of specific developmental pathways downstream of mutations in cohesin components. However, it is still unclear how mutations in different components of the cohesin complex affect the output of gene regulation. In this study, zebrafish embryos and SMC1A-mutated patient-derived fibroblasts were used to analyze abnormalities induced by SMC1A loss of function. We show that the knockdown of smc1a in zebrafish impairs neural development, increases apoptosis, and specifically down-regulates Ccnd1 levels. The same down-regulation of cohesin targets is observed in SMC1A-mutated patient fibroblasts. Previously, we have demonstrated that haploinsufficiency of NIPBL produces similar effects in zebrafish and in patients fibroblasts indicating a possible common feature for neurological defects and mental retardation in cohesinopathies. Interestingly, expression analysis of Smc1a and Nipbl in developing mouse embryos reveals a specific pattern in the hindbrain, suggesting a role for cohesins in neural development in vertebrates.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cyclin D1/metabolism , De Lange Syndrome/metabolism , Transcription Factors/metabolism , Zebrafish/embryology , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , De Lange Syndrome/genetics , Down-Regulation , Humans , Mice , Mutation/genetics , Transcription Factors/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Anal Chem ; 88(14): 7115-22, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27348197

ABSTRACT

In vivo studies of blood circulation pathologies have great medical relevance and need methods for the characterization of time varying flows at high spatial and time resolution in small animal models. We test here the efficacy of the combination of image correlation techniques and single plane illumination microscopy (SPIM) in characterizing time varying flows in vitro and in vivo. As indicated by numerical simulations and by in vitro experiments on straight capillaries, the complex analytical form of the cross-correlation function for SPIM detection can be simplified, in conditions of interest for hemodynamics, to a superposition of Gaussian components, easily amenable to the analysis of variable flows. The possibility to select a wide field of view with a good spatial resolution along the collection optical axis and to compute the cross-correlation between regions of interest at varying distances on a single time stack of images allows one to single out periodic flow components from spurious peaks on the cross-correlation functions and to infer the duration of each flow component. We apply this cross-correlation analysis to the blood flow in Zebrafish embryos at 4 days after fertilization, measuring the average speed and the duration of the systolic and diastolic phases.


Subject(s)
Hemodynamics , Zebrafish/physiology , Animals , Fluorescence , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Microspheres , Pulse , Rhodamines/chemistry , Time , Unilamellar Liposomes/chemistry
10.
Development ; 140(7): 1528-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23482488

ABSTRACT

During mouse skeletal muscle development, the Nfix gene has a pivotal role in regulating fetal-specific transcription. Zebrafish and mice share related programs for muscle development, although zebrafish develops at a much faster rate. In fact, although mouse fetal muscle fibers form after 15 days of development, in fish secondary muscle fibers form by 48 hours post-fertilization in a process that until now has been poorly characterized mechanically. In this work, we studied the zebrafish ortholog Nfix (nfixa) and its role in the proper switch to the secondary myogenic wave. This allowed us to highlight evolutionarily conserved and divergent functions of Nfix. In fact, the knock down of nfixa in zebrafish blocks secondary myogenesis, as in mouse, but also alters primary slow muscle fiber formation. Moreover, whereas Nfix mutant mice are motile, nfixa knockdown zebrafish display impaired motility that probably depends upon disruption of the sarcoplasmic reticulum. We conclude that, during vertebrate evolution, the transcription factor Nfix lost some specific functions, probably as a consequence of the different environment in which teleosts and mammals develop.


Subject(s)
Evolution, Molecular , Muscle, Skeletal/embryology , NFI Transcription Factors/physiology , Vertebrates/genetics , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Conserved Sequence/physiology , Embryo, Mammalian , Embryo, Nonmammalian , Genetic Speciation , Mice , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/metabolism , NFI Transcription Factors/genetics , Phylogeny , Vertebrates/embryology , Vertebrates/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
11.
J Cell Physiol ; 230(4): 821-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25205658

ABSTRACT

The Coiled-Coil Domain Containing 80 (CCDC80) gene has been identified as strongly induced in rat thyroid PC CL3 cells immortalized by the adenoviral E1A gene. In human, CCDC80 is a potential oncosoppressor due to its down-regulation in several tumor cell lines and tissues and it is expressed in almost all tissues. CCDC80 has homologous in mouse, chicken, and zebrafish. We cloned the zebrafish ccdc80 and analyzed its expression and function during embryonic development. The in-silico translated zebrafish protein shares high similarity with its mammalian homologous, with nuclear localization signals and a signal peptide. Gene expression analysis demonstrates that zebrafish ccdc80 is maternally and zygotically expressed throughout the development. In particular, ccdc80 is strongly expressed in the notochord and it is under the regulation of the Hedgehog pathway. In this work we investigated the functional effects of ccdc80-loss-of-function during embryonic development and verified its interaction with gadd45ß2 in somitogenesis.


Subject(s)
Antigens, Differentiation/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction/genetics , Somites/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Down-Regulation/physiology , Hedgehog Proteins/metabolism , Zebrafish/embryology
12.
J Cell Biochem ; 116(3): 418-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25335865

ABSTRACT

ß-Catenin is a central effector of the Wnt pathway and one of the players in Ca(+)-dependent cell-cell adhesion. While many wnts are present and expressed in vertebrates, only one ß-catenin exists in the majority of the organisms. One intriguing exception is zebrafish that carries two genes for ß-catenin. The maternal recessive mutation ichabod presents very low levels of ß-catenin2 that in turn affects dorsal axis formation, suggesting that ß-catenin1 is incapable to compensate for ß-catenin2 loss and raising the question of whether these two ß-catenins may have differential roles during early axis specification. Here we identify a specific antibody that can discriminate selectively for ß-catenin1. By confocal co-immunofluorescent analysis and low concentration gain-of-function experiments, we show that ß-catenin1 and 2 behave in similar modes in dorsal axis induction and cellular localization. Surprisingly, we also found that in the ich embryo the mRNAs of the components of ß-catenin regulatory pathway, including ß-catenin1, are more abundant than in the Wt embryo. Increased levels of ß-catenin1 are found at the membrane level but not in the nuclei till high stage. Finally, we present evidence that ß-catenin1 cannot revert the ich phenotype because it may be under the control of a GSK3ß-independent mechanism that required Axin's RGS domain function.


Subject(s)
Axin Protein/metabolism , Mutation/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Antibody Specificity , Axin Protein/genetics , Blastula/drug effects , Blastula/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Genes, Dominant , Immunohistochemistry , Lithium Chloride/pharmacology , Phenotype , Protein Stability/drug effects , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics , beta Catenin/metabolism
13.
J Med Genet ; 51(7): 436-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24711647

ABSTRACT

BACKGROUND: Cardiovascular malformations have a higher incidence in patients with NF1 microdeletion syndrome compared to NF1 patients with intragenic mutation, presumably owing to haploinsufficiency of one or more genes included in the deletion interval and involved in heart development. In order to identify which genes could be responsible for cardiovascular malformations in the deleted patients, we carried out expression studies in mouse embryos and functional studies in zebrafish. METHODS AND RESULTS: The expression analysis of three candidate genes included in the NF1 deletion interval, ADAP2, SUZ12 and UTP6, performed by in situ hybridisation, showed the expression of ADAP2 murine ortholog in heart during fundamental phases of cardiac morphogenesis. In order to investigate the role of ADAP2 in cardiac development, we performed loss-of-function experiments of zebrafish ADAP2 ortholog, adap2, by injecting two different morpholino oligos (adap2-MO and UTR-adap2-MO). adap2-MOs-injected embryos (morphants) displayed in vivo circulatory and heart shape defects. The molecular characterisation of morphants with cardiac specific markers showed that the injection of adap2-MOs causes defects in heart jogging and looping. Additionally, morphological and molecular analysis of adap2 morphants demonstrated that the loss of adap2 function leads to defective valvulogenesis, suggesting a correlation between ADAP2 haploinsufficiency and the occurrence of valve defects in NF1-microdeleted patients. CONCLUSIONS: Overall, our findings indicate that ADAP2 has a role in heart development, and might be a reliable candidate gene for the occurrence of cardiovascular malformations in patients with NF1 microdeletion and, more generally, for the occurrence of a subset of congenital heart defects.


Subject(s)
Cardiovascular Abnormalities/genetics , Craniofacial Abnormalities/genetics , GTPase-Activating Proteins/genetics , Intellectual Disability/genetics , Learning Disabilities/genetics , Neurofibromatoses/genetics , Animals , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Disease Models, Animal , Genetic Association Studies , Genetic Predisposition to Disease , Heart/embryology , Humans , Mice , Morphogenesis , Zebrafish
14.
Arterioscler Thromb Vasc Biol ; 33(6): 1238-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23520166

ABSTRACT

OBJECTIVE: Lymphangiogenesis is regulated by transcription factors and by growth factor pathways, but their interplay has not been extensively studied so far. We addressed this issue in zebrafish. APPROACH AND RESULTS: Mutations in the transcription factor-coding gene SOX18 and in VEGFR3 cause lymphedema, and the VEGFR3/Flt4 ligand VEGFC plays an evolutionarily conserved role in lymphangiogenesis. Here, we report a strong genetic interaction between Sox18 and VegfC in the early phases of lymphatic development in zebrafish. Knockdown of sox18 selectively impaired lymphatic sprouting from the cardinal vein and resulted in defective lymphatic thoracic duct formation. Sox18 and the related protein Sox7 play redundant roles in arteriovenous differentiation. We used a novel transgenic line that enables inducible expression of a dominant-negative mutant form of mouse Sox18 protein. Our data led us to conclude that Sox18 is crucially involved in lymphangiogenesis after arteriovenous differentiation. Combined partial knockdown of sox18 and vegfc, using subcritical doses of specific morpholinos, revealed a synergistic interaction in both venous and lymphatic sprouting from the cardinal vein and greatly impaired thoracic duct formation. CONCLUSIONS: This interaction suggests a previously unappreciated crosstalk between the growth factor and transcription factor pathways that regulate lymphangiogenesis in development and disease.


Subject(s)
Gene Expression Regulation, Developmental , Lymphangiogenesis/genetics , SOXF Transcription Factors/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor C/genetics , Animals , Animals, Genetically Modified , Blood Vessels/embryology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice , Models, Animal , Protein Interaction Domains and Motifs/genetics , SOXF Transcription Factors/metabolism , Sensitivity and Specificity , Vascular Endothelial Growth Factor C/metabolism , Zebrafish
15.
Biochim Biophys Acta ; 1823(10): 1925-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22867989

ABSTRACT

Blood vessel formation depends on the highly coordinated actions of a variety of angiogenic regulators. Vascular endothelial growth factor (VEGF) and Angiopoietin-1 (Ang-1) are both potent and essential proangiogenic factors with complementary roles in vascular development and function. Whereas VEGF is required for the formation of the initial vascular plexus, Ang-1 contributes to the stabilization and maturation of growing blood vessels. Here, we provide evidence of a novel microRNA (miRNA)-dependent molecular mechanism of Ang-1 signalling modulation aimed at stabilizing adult vasculature. MiRNAs are short non-coding RNA molecules that post-trascriptionally regulate gene expression by translational suppression or in some instances by cleavage of the respective mRNA target. Our data indicate that endothelial cells of mature vessels express high levels of miR-126, which primarily targets phosphoinositide-3-kinase regulatory subunit 2 (p85ß). Down-regulation of miR-126 and over-expression of p85ß in endothelial cells inhibit the biological functions of Ang-1. Additionally, knockdown of miR-126 in zebrafish resulted in vascular remodelling and maturation defects, reminiscent of the Ang-1 loss-of-function phenotype. Our findings suggest that miR-126-mediated phosphoinositide-3-kinase regulation, not only fine-tunes VEGF-signaling, but it strongly enhances the activities of Ang-1 on vessel stabilization and maturation.


Subject(s)
Angiopoietin-1/metabolism , Blood Vessels/embryology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , MicroRNAs/metabolism , Morphogenesis , Signal Transduction/genetics , Angiopoietin-1/genetics , Animals , Blood Vessels/metabolism , Cell Survival , Down-Regulation/genetics , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/metabolism , Enzyme Activation , Gene Expression Regulation, Developmental , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Mice , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
16.
J Neurochem ; 127(1): 22-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23800350

ABSTRACT

Transcriptional dysregulation is a hallmark of Huntington's disease (HD) and one cause of this dysregulation is enhanced activity of the REST-mSIN3a-mSIN3b-CoREST-HDAC repressor complex, which silences transcription through REST binding to the RE1/NRSE silencer. Normally, huntingtin (HTT) prevents this binding, allowing expressing of REST target genes. Here, we aimed to identify HTT mimetics that disrupt REST complex formation in HD. From a structure-based virtual screening of 7 million molecules, we selected 94 compounds predicted to interfere with REST complex formation by targeting the PAH1 domain of mSIN3b. Primary screening using DiaNRSELuc8 cells revealed two classes of compounds causing a greater than two-fold increase in luciferase. In particular, quinolone-like compound 91 (C91) at a non-toxic nanomolar concentration reduced mSIN3b nuclear entry and occupancy at the RE1/NRSE within the Bdnf locus, and restored brain-derived neurotrophic factor (BDNF) protein levels in HD cells. The mRNA levels of other RE1/NRSE-regulated genes were similarly increased while non-REST-regulated genes were unaffected. C91 stimulated REST-regulated gene expression in HTT-knockdown Zebrafish and increased BDNF mRNA in the presence of mutant HTT. Thus, a combination of virtual screening and biological approaches can lead to compounds reducing REST complex formation, which may be useful in HD and in other pathological conditions.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Animals , Animals, Genetically Modified , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Co-Repressor Proteins , Embryo, Nonmammalian , Enzyme-Linked Immunosorbent Assay , Humans , Huntingtin Protein , Immunohistochemistry , Luciferases/metabolism , Microinjections , Models, Molecular , Nerve Tissue Proteins/biosynthesis , Polymerase Chain Reaction , RNA, Messenger/administration & dosage , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Transcription, Genetic/genetics , Transfection , Zebrafish
17.
Arterioscler Thromb Vasc Biol ; 32(7): 1563-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22516065

ABSTRACT

OBJECTIVE: The goal of this study was to determine the in vivo functions of the synaptic proteins neurexins and neuroligins in embryonic vascular system development using zebrafish as animal model. METHODS AND RESULTS: In the present study, we show that the knockdown of the α-form of neurexin 1a induces balance defects and reduced locomotory activity, whereas ß-neurexin 1a and neuroligin 1 morphants present defects in sprouting angiogenesis and vascular remodeling, in particular in the caudal plexus and subintestinal vessels. Coinjection of low doses of morpholinos for ß-neurexin 1a and neuroligin 1 together or in combination with morpholinos targeting the -heparin--binding isoforms of vascular endothelial growth factor A (encoded by the VEGFAb gene) recapitulates the observed abnormalities, suggesting synergistic activity of these molecules. Similar coinjection experiments with morpholinos, targeting the enzyme heparan sulfate 6-O-sulfotransferase 2, confirm the presence of a functional correlation between extracellular matrix maturation and ß-neurexin 1a or neuroligin 1. CONCLUSIONS: Our data represent the first in vivo evidence of the role of neurexin and neuroligin in embryonic blood vessel formation and provide insights into their mechanism of action.


Subject(s)
Blood Vessels/embryology , Cell Adhesion Molecules, Neuronal/physiology , Glycoproteins/physiology , Heparin/metabolism , Neovascularization, Physiologic , Neuropeptides/physiology , Vascular Endothelial Growth Factor A/physiology , Zebrafish/embryology , Animals , Extracellular Matrix/physiology , Sulfotransferases/physiology
18.
Arterioscler Thromb Vasc Biol ; 31(7): 1589-97, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21527751

ABSTRACT

OBJECTIVE: The vascular competence of human-derived hematopoietic progenitors for postnatal vascularization is still poorly characterized. It is unclear whether, in the absence of ischemia, hematopoietic progenitors participate in neovascularization and whether they play a role in new blood vessel formation by incorporating into developing vessels or by a paracrine action. METHODS AND RESULTS: In the present study, human cord blood-derived CD34(+) (hCD34(+)) cells were transplanted into pre- and postgastrulation zebrafish embryos and in an adult vascular regeneration model induced by caudal fin amputation. When injected before gastrulation, hCD34(+) cells cosegregated with the presumptive zebrafish hemangioblasts, characterized by Scl and Gata2 expression, in the anterior and posterior lateral mesoderm and were involved in early development of the embryonic vasculature. These morphogenetic events occurred without apparent lineage reprogramming, as shown by CD45 expression. When transplanted postgastrulation, hCD34(+) cells were recruited into developing vessels, where they exhibited a potent paracrine proangiogenic action. Finally, hCD34(+) cells rescued vascular defects induced by Vegf-c in vivo targeting and enhanced vascular repair in the zebrafish fin amputation model. CONCLUSIONS: These results indicate an unexpected developmental ability of human-derived hematopoietic progenitors and support the hypothesis of an evolutionary conservation of molecular pathways involved in endothelial progenitor differentiation in vivo.


Subject(s)
Animal Fins/blood supply , Antigens, CD34/analysis , Cell Differentiation , Cord Blood Stem Cell Transplantation , Endothelial Cells/transplantation , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Neovascularization, Physiologic , Zebrafish , Amputation, Surgical , Animal Fins/surgery , Animals , Animals, Genetically Modified , Caco-2 Cells , Cell Differentiation/drug effects , Cell Movement , Endothelial Cells/immunology , Fetal Blood/immunology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/immunology , Humans , Paracrine Communication , Phenotype , RNA Interference , Recombinant Fusion Proteins/metabolism , Regeneration , Signal Transduction , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
19.
J Sports Med Phys Fitness ; 62(6): 788-794, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34156181

ABSTRACT

BACKGROUND: We tested the hypothesis that whole-body vibration (WBV) positively affects the fatigue process ensuing from repeated bouts of maximal efforts, as induced by repeated sprints' ability (RSA). Eleven male soccer players performed three sets of six repeated shuttle sprints (40 meters). METHODS: Eleven male soccer players (age 23.6±4.5 years) were cross-randomized to perform WBW before RSA and during the recovery between sets (WBV-with) or to warm-up and passive recovery between sets (WBV-without). The effects of WBV were quantified by sprint time (ST) and blood lactate concentration (LA), collected up to 15 min after completion of tests. RESULTS: ST during RSA showed a better maintenance of performance in the WBV-with compared to WBV-without condition in all three sets, reaching a statistical significance between-groups during the 2nd and 3rd set (P<0.05). No significant differences in ST over the sets were detected in WBV-with, whereas a significant decrease was observed in the WBV-without condition (P<0.001). LA recovered significantly faster from the 9th to 15th minute of recovery in WBV-with as compared to WBV-without (P<0.05). CONCLUSIONS: These findings would indicate that WBV performed during recovery between RSA sets can delay the onset of muscle fatigue resulting in a better maintenance of sprint performance.


Subject(s)
Athletic Performance , Running , Soccer , Adult , Athletic Performance/physiology , Exercise Test/methods , Humans , Lactic Acid , Male , Muscle Fatigue/physiology , Running/physiology , Soccer/physiology , Vibration , Young Adult
20.
FASEB J ; 24(7): 2385-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20207941

ABSTRACT

Cerebral amyloid angiopathy (CAA) caused by amyloid beta (Abeta) deposition around brain microvessels results in vascular degenerative changes. Antiangiogenic Abeta properties are known to contribute to the compromised cerebrovascular architecture. Here we hypothesize that Abeta peptides impair angiogenesis by causing endothelial cells to enter senescence at an early stage of vascular development. Wild-type (WT) Abeta and its mutated variant E22Q peptide, endowed with marked vascular tropism, were used in this study. In vivo, in zebrafish embryos, the WT or E22Q peptides reduced embryo survival with an IC(50) of 6.1 and 4.7 microM, respectively. The 2.5 microM concentration, showing minimal toxicity, was chosen. Alkaline phosphatase staining revealed disorganized vessel patterning, narrowing, and reduced branching of vessels. Beta-galactosidase staining and the cyclin-dependent kinase inhibitor p21 expression, indicative of senescence, were increased. In vitro, WT and E22Q reduced endothelial cell survival with an IC(50) of 12.3 and 8.8 microM, respectively. The 5 microM concentration, devoid of acute effects on the endothelium, was applied chronically to long-term cultured human umbilical vein endothelial cells (HUVECs). We observed reduced cumulative population doubling, which coincided with beta-galactosidase accumulation, down-regulation of telomerase reverse-transcriptase mRNA expression, decreased telomerase activity, and p21 activation. Senescent HUVECs showed marked angiogenesis impairment, as Abeta treatment reduced tube sprouting. The endothelial injuries caused by the E22Q peptide were much more aggressive than those induced by the WT peptide. Premature Abeta-induced senescence of the endothelium, producing progressive alterations of microvessel morphology and functions, may represent one of the underlying mechanisms for sporadic or heritable CAA.


Subject(s)
Amyloid beta-Peptides/toxicity , Cellular Senescence/drug effects , Endothelial Cells/drug effects , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/pharmacology , Angiogenesis Inhibitors , Animals , Blood Vessels/growth & development , Blood Vessels/pathology , Cerebral Amyloid Angiopathy , Embryo, Nonmammalian/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Humans , Mutation, Missense , Neovascularization, Physiologic/drug effects , Survival Rate , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL