Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; : 133652, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38971273

ABSTRACT

Eighteen S rRNA factor 1 (ESF1) is a predominantly nucleolar protein essential for embryogenesis. Our previous studies have suggested that Esf1 is a negative regulator of the tumor suppressor protein p53. However, it remains unclear whether ESF1 contributes to tumorigenesis. In this current research, we find that increased ESF1 expression correlates with poor survival in multiple tumors including pancreatic cancer. ESF1 is able to regulate cell proliferation, migration, DNA damage-induced apoptosis, and tumorigenesis. Mechanistically, ESF1 physically interacts with MDM2 and is essential for maintaining the stability of MDM2 protein by inhibiting its ubiquitination. Additionally, ESF1 also prevented stress-induced stabilization of p53 in multiple cancer cells. Hence, our findings suggest that ESF1 is a potent regulator of the MDM2-p53 pathway and promotes tumor progression.

2.
Life Sci ; 344: 122564, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38492922

ABSTRACT

AIMS: Prolyl hydroxylase domain 2 (PHD2), encoded by the Egln1 gene, serves as a pivotal regulator of the hypoxia-inducible factor (HIF) pathway and acts as a cellular oxygen sensor. Somatic inactivation of Phd2 in mice results in polycythemia and congestive heart failure. However, due to the embryonic lethality of Phd2 deficiency, its role in development remains elusive. Here, we investigated the function of two egln1 paralogous genes, egln1a and egln1b, in zebrafish. MAIN METHODS: The egln1 null zebrafish were generated using the CRISPR/Cas9 system. Quantitative real-time PCR assays and Western blot analysis were employed to detect the effect of egln1 deficiency on the hypoxia signaling pathway. The hypoxia response of egln1 mutant zebrafish were assessed by analyzing heart rate, gill agitation frequency, and blood flow velocity. Subsequently, o-dianisidine staining and in situ hybridization were used to investigate the role of egln1 in zebrafish hematopoietic function. KEY FINDINGS: Our data show that the loss of egln1a or egln1b individually has no visible effects on growth rate. However, the egln1a; egln1b double mutant displayed significant growth retardation and elevated mortality at around 2.5 months old. Both egln1a-null and egln1b-null zebrafish embryo exhibited enhanced tolerance to hypoxia, systemic hypoxic response that include hif pathway activation, increased cardiac activity, and polycythemia. SIGNIFICANCE: Our research introduces zebrafish egln1 mutants as the first congenital embryonic viable systemic vertebrate animal model for PHD2, providing novel insights into hypoxic signaling and the progression of PHD2- associated disease.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Hypoxia , Polycythemia , Zebrafish , Animals , Mice , Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Polycythemia/genetics , Procollagen-Proline Dioxygenase/genetics , Zebrafish/genetics , Zebrafish/metabolism
3.
J Biol Chem ; 299(6): 104829, 2023 06.
Article in English | MEDLINE | ID: mdl-37201586

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a critical transcription factor that regulates the expression of genes involved in cellular adaptation to low oxygen levels. Aberrant regulation of the HIF-1 signaling pathway is linked to various human diseases. Previous studies have established that HIF-1α is rapidly degraded in a von Hippel-Lindau protein (pVHL)-dependent manner under normoxic conditions. In this study, we find that pVHL binding protein 1 (VBP1) is a negative regulator of HIF-1α but not HIF-2α using zebrafish as an in vivo model and in vitro cell culture models. Deletion of vbp1 in zebrafish caused Hif-1α accumulation and upregulation of Hif target genes. Moreover, vbp1 was involved in the induction of hematopoietic stem cells (HSCs) under hypoxic conditions. However, VBP1 interacted with and promoted the degradation of HIF-1α in a pVHL-independent manner. Mechanistically, we identify the ubiquitin ligase CHIP and HSP70 as new VBP1 binding partners and demonstrate that VBP1 negatively regulated CHIP and facilitated CHIP-mediated degradation of HIF-1α. In patients with clear cell renal cell carcinoma (ccRCC), lower VBP1 expression was associated with worse survival outcomes. In conclusion, our results link VBP1 with CHIP stability and provide insights into underlying molecular mechanisms of HIF-1α-driven pathological processes.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Humans , Zebrafish/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Transcription Factors/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cytoskeletal Proteins , Molecular Chaperones
4.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33914867

ABSTRACT

A key step in the activation of canonical Wnt signaling is the interaction between ß-catenin and Tcf/Lefs that forms the transcription activation complex and facilitates the expression of target genes. Eukaryotic initiation factor 4A3 (EIF4A3) is an ATP-dependent DEAD box-family RNA helicase and acts as a core subunit of the exon junction complex (EJC) to control a series of RNA post-transcriptional processes. In this study, we uncover that EIF4A3 functions as a Wnt inhibitor by interfering with the formation of ß-catenin/Tcf transcription activation complex. As Wnt stimulation increases, accumulated ß-catenin displaces EIF4A3 from a transcriptional complex with Tcf/Lef, allowing the active complex to facilitate the expression of target genes. In zebrafish embryos, eif4a3 depletion inhibited the development of the dorsal organizer and pattern formation of the anterior neuroectoderm by increasing Wnt/ß-catenin signaling. Conversely, overexpression of eif4a3 decreased Wnt/ß-catenin signaling and inhibited the formation of the dorsal organizer before gastrulation. Our results reveal previously unreported roles of EIF4A3 in the inhibition of Wnt signaling and the regulation of embryonic development in zebrafish.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , Wnt Signaling Pathway/physiology , Zebrafish/embryology , Animals , Embryo, Nonmammalian/metabolism , Eukaryotic Initiation Factor-4A/genetics , Gene Expression Regulation, Developmental , Transcriptional Activation , Wnt Proteins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
5.
J Biol Chem ; 295(49): 16826-16839, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32989053

ABSTRACT

The Wnt/ß-catenin pathway is one of the major pathways that regulates embryonic development, adult homeostasis, and stem cell self-renewal. In this pathway, transcription factors T-cell factor and lymphoid enhancer factor (TCF/LEF) serve as a key switch to repress or activate Wnt target gene transcription by recruiting repressor molecules or interacting with the ß-catenin effector, respectively. It has become evident that the protein stability of the TCF/LEF family members may play a critical role in controlling the activity of the Wnt/ß-catenin signaling pathway. However, factors that regulate the stability of TCF/LEFs remain largely unknown. Here, we report that pVHL binding protein 1 (VBP1) regulates the Wnt/ß-catenin signaling pathway by controlling the stability of TCF/LEFs. Surprisingly, we found that either overexpression or knockdown of VBP1 decreased Wnt/ß-catenin signaling activity in both cultured cells and zebrafish embryos. Mechanistically, VBP1 directly binds to all four TCF/LEF family members and von Hippel-Lindau tumor-suppressor protein (pVHL). Either overexpression or knockdown of VBP1 increases the association between TCF/LEFs and pVHL and then decreases the protein levels of TCF/LEFs via proteasomal degradation. Together, our results provide mechanistic insights into the roles of VBP1 in controlling TCF/LEFs protein stability and regulating Wnt/ß-catenin signaling pathway activity.


Subject(s)
Cytoskeletal Proteins/metabolism , Molecular Chaperones/metabolism , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Cell Line , Cell Proliferation , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Embryo, Nonmammalian/metabolism , Humans , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , TCF Transcription Factors/genetics , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Transcriptional Activation , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
6.
FEBS J ; 285(18): 3464-3484, 2018 09.
Article in English | MEDLINE | ID: mdl-30073783

ABSTRACT

Craniofacial malformations are common congenital birth defects and usually caused by abnormal development of the cranial neural crest cells. Some nucleolar ribosome biogenesis factors are implicated in neural crest disorders also known as neurocristopathies. However, the underlying mechanisms linking ribosome biogenesis and neural crest cell (NCC) development remain to be elucidated. Here we report a novel zebrafish model with a CRISPR/Cas9-generated esf1 mutation, which exhibits severe NCC-derived pharyngeal cartilage loss and defects in the eyes, brain, and heart. The expression of several typical NCC markers, including sox10, dlx2a, nrp2b, crestin, vgll2a, and sox9a, was reduced in the head of the esf1 mutants, which indicates that esf1 plays a role in the development of zebrafish NCCs. We demonstrate that, similar to the yeast, loss of esf1 in zebrafish leads to defects in 18S rRNA biogenesis and ribosome biogenesis. We also show strong upregulation of p53 signaling as well as apoptosis, and poor proliferation in mutants. Inactivation of p53 rescues the early tissue defects and pharyngeal cartilage loss observed in esf1 mutants, indicating that increased cell death and pharyngeal cartilage defects observed in esf1 mutants are mediated via upregulated p53 signaling pathways. Based on transplantation analysis, we found esf1 functions in NCC in a cell autonomous fashion. Together, our results suggest that esf1 is required for NCC development and pharyngeal cartilage formation. These studies provide a potential model for investigating the relationship between ribosome biogenesis defects and craniofacial neurocristopathies.


Subject(s)
Cartilage/embryology , Embryo, Nonmammalian/cytology , Nuclear Proteins/metabolism , Pharynx/embryology , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Body Patterning , Cartilage/metabolism , Embryo, Nonmammalian/metabolism , Mutation , Nuclear Proteins/genetics , Pharynx/metabolism , Ribosomes/metabolism , Tumor Suppressor Protein p53/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
7.
FEBS Lett ; 592(16): 2769-2775, 2018 08.
Article in English | MEDLINE | ID: mdl-30047986

ABSTRACT

Hif-3α, a member of the hypoxia-inducible factor (HIF) family, enters the nucleus and regulates gene expression in response to hypoxia. The molecular basis of its nuclear localization is not clear. HIF-1α and HIF-2α use a bipartite nuclear localization signal (NLS) to enter the nucleus. This motif is not conserved in Hif-3α. Although there is a conserved Arg/Lys rich motif in the Hif-3α N-terminal region, deletion of this region has minimal effect on Hif-3α nuclear localization. Here, we mapped the functional NLS to the unique C-terminal region of Hif-3α and identified two clusters of basic residues critical for its nuclear localization. The two NLS motifs are functionally redundant. Our results, thus, suggest that Hif-3α nuclear localization is mediated through two redundant NLS motifs located in its unique C-terminal region.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Apoptosis Regulatory Proteins , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia , Gene Expression Regulation , HeLa Cells , Humans , Nuclear Localization Signals , Protein Domains , Repressor Proteins
8.
Mol Cell Biochem ; 432(1-2): 199-208, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28374141

ABSTRACT

Insulin-like growth factor II (IGF-II) can stimulate myogenesis and is critically involved in skeletal muscle differentiation. The presence of negative regulators of this process, however, is not well explored. Here, we showed that in myoblast cells, IGF-II negatively regulated peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) mRNA expression, while constitutive expression of PGC-1α induced myoblast differentiation. These results suggest that the negative regulation of PGC-1α by IGF-II may act as a negative feedback mechanism in IGF-II-induced myogenic differentiation. Reporter assays demonstrated that IGF-II suppresses the basal PGC-1α promoter activity. Blocking the IGF-II signaling pathway increased the endogenous PGC-1α levels. In addition, pharmacological inhibition of PI3 kinase activity prevented the downregulation of PGC-1α but the activation of mTOR was not required for this process. Importantly, further analysis showed that forkhead transcription factor FoxO1 contributes to mediating the effects of IGF-II on PGC-1 promoter activity. These findings indicate that IGF-II reduces PGC-1α expression in skeletal muscle cells through a mechanism involving PI3K-Akt-FoxO1 but not p38 MAPK or Erk1/2 MAPK pathways.


Subject(s)
Forkhead Box Protein O1/metabolism , Insulin-Like Growth Factor II/metabolism , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cell Line , Forkhead Box Protein O1/genetics , Insulin-Like Growth Factor II/genetics , Mice , Muscle, Skeletal/cytology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
9.
Development ; 144(9): 1687-1697, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28302747

ABSTRACT

The Wnt/ß-catenin signaling pathway plays pivotal roles in axis formation during embryogenesis and in adult tissue homeostasis. Glutathione peroxidase 4 (GPX4) is a selenoenzyme and participates in the reduction of peroxides. Its synthesis depends on the availability of the element selenium. However, the roles of GPX4 in vertebrate embryonic development and underlying mechanisms are largely unknown. Here, we show that maternal loss of zebrafish gpx4b promotes embryonic dorsal organizer formation, whereas overexpression of gpx4b inhibits the development of the dorsal organizer. Depletion of human GPX4 and zebrafish gpx4b (GPX4/gpx4b) increases, while GPX4/gpx4b overexpression decreases, Wnt/ß-catenin signaling in vivo and in vitro Functional and epistatic studies showed that GPX4 functions at the Tcf/Lef level, independently of selenocysteine activation. Mechanistically, GPX4 interacts with Tcf/Lefs and inhibits Wnt activity by preventing the binding of Tcf/Lefs to the promoters of Wnt target genes, resulting in inhibitory action in the presence of Wnt/ß-catenin signaling. Our findings unravel GPX4 as a suppressor of Wnt/ß-catenin signals, suggesting a possible relationship between the Wnt/ß-catenin pathway and selenium via the association of Tcf/Lef family proteins with GPX4.


Subject(s)
Embryo, Nonmammalian/enzymology , Glutathione Peroxidase/metabolism , Organizers, Embryonic/enzymology , Wnt Signaling Pathway , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Embryo, Nonmammalian/cytology , Evolution, Molecular , Gene Expression Regulation, Developmental , Glutathione Peroxidase/chemistry , Glutathione Peroxidase/deficiency , HEK293 Cells , Humans , Phenotype , Phospholipid Hydroperoxide Glutathione Peroxidase , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Selenium/metabolism , Signal Transduction/genetics , Transcription, Genetic , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zygote/metabolism
10.
Article in English | MEDLINE | ID: mdl-28228749

ABSTRACT

Neuronal survival and growth in the embryo is controlled partly by trophic factors. For most trophic factors (such as Insulin-like growth factor-1), the ability to regulate cell survival has been attributed to the phosphoinositide 3-kinase (PI3K)/Akt kinase cascade. This study presents data illustrating the role of PI3K/Akt in attainment of normal brain size during zebrafish embryogenesis. Blocking PI3K with inhibitor LY294002 caused a significant reduction in brain size (in addition to global growth retardation) during zebrafish embryogenesis. This PI3 Kinase inhibition-induced brain size decrease was recovered by the overexpression of myristoylated Akt (myr-Akt), a constitutive form of Akt. Further analysis reveals that expressing exogenous myr-Akt significantly augmented brain size. Whole mount in situ hybridization analysis of several marker genes showed that myr-Akt overexpression did not alter brain patterning. Furthermore, the expression of myr-Akt was found to protect neuronal cells from apoptosis induced by heat shock and UV light, suggesting that inhibition of neuronal cell death may be part of the underlying cause of the increased brain size. These data provide a foundation for addressing the role of PI3K/Akt in brain growth during zebrafish embryogenesis.

11.
Mar Biotechnol (NY) ; 18(2): 264-70, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26838966

ABSTRACT

Marine organisms often protect themselves against their predators by chemical defensive strategy. The second metabolites isolated from marine organisms and their symbiotic microbes have been proven to play a vital role in marine chemical ecology, such as ichthyotoxicity, allelopathy, and antifouling. It is well known that the microscale models for marine chemoecology assessment are urgently needed for trace quantity of marine natural products. Zebrafish model has been widely used as a microscale model in the fields of environment ecological evaluation and drug safety evaluation, but seldom reported for marine chemoecology assessment. In this work, zebrafish embryo toxicity microscale model was established for ichthyotoxicity evaluation of marine natural products by using 24-well microplate based on zebrafish embryo. Ichthyotoxicity was evaluated by observation of multiple toxicological endpoints, including coagulation egg, death, abnormal heartbeat, no spontaneous movement, delayed hatch, and malformation of the different organs during zebrafish embryogenesis periods at 24, 48, and 72 h post-fertilization (hpf). 3,4-Dichloroaniline was used as the positive control for method validation. Subsequently, the established model was applied to test the ichthyotoxic activity of the compounds isolated from corals and their symbiotic microbes and to isolate the bioactive secondary metabolites from the gorgonian Subergorgia mollis under bioassay guidance. It was suggested that zebrafish embryo toxicity microscale model is suitable for bioassay-guided isolation and preliminary bioactivity screening of marine natural products.


Subject(s)
Biological Assay , Biological Products/toxicity , Embryonic Development/drug effects , Heart/drug effects , Zygote/drug effects , Animals , Anthozoa/chemistry , Biological Products/isolation & purification , Embryo, Nonmammalian , Heart/growth & development , Lethal Dose 50 , Microscopy , Mutagenicity Tests , Toxicity Tests, Acute , Toxicity Tests, Chronic , Zebrafish
12.
PLoS One ; 9(6): e99514, 2014.
Article in English | MEDLINE | ID: mdl-24918770

ABSTRACT

The Wnt/ß-catenin or canonical Wnt signaling pathway plays fundamental roles in early development and in maintaining adult tissue homeostasis. R-spondin 3 (Rspo3) is a secreted protein that has been implicated in activating the Wnt/ß-catenin signaling in amphibians and mammals. Here we report that zebrafish Rspo3 plays a negative role in regulating the zygotic Wnt/ß-catenin signaling. Zebrafish Rspo3 has a unique domain structure. It contains a third furin-like (FU3) domain. This FU3 is present in other four ray-finned fish species studied but not in elephant shark. In zebrafish, rspo3 mRNA is maternally deposited and has a ubiquitous expression in early embryonic stages. After 12 hpf, its expression becomes tissue-specific. Forced expression of rspo3 promotes dorsoanterior patterning and increases the expression of dorsal and anterior marker genes. Knockdown of rspo3 increases ventral-posterior development and stimulates ventral and posterior marker genes expression. Forced expression of rspo3 abolishes exogenous Wnt3a action and reduces the endogenous Wnt signaling activity. Knockdown of rspo3 results in increased Wnt/ß-catenin signaling activity. Further analyses indicate that Rspo3 does not promote maternal Wnt signaling. Human RSPO3 has similar action when tested in zebrafish embryos. These results suggest that Rspo3 regulates dorsoventral and anteroposterior patterning by negatively regulating the zygotic Wnt/ß-catenin signaling in zebrafish embryos.


Subject(s)
Body Patterning , Intracellular Signaling Peptides and Proteins/physiology , Wnt Signaling Pathway , Zebrafish Proteins/physiology , Amino Acid Sequence , Animals , Evolution, Molecular , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Protein Structure, Tertiary , Thrombospondins/genetics , Thrombospondins/physiology , Zebrafish/embryology , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
13.
J Biol Chem ; 289(30): 20664-76, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24928507

ABSTRACT

The receptor-tyrosine kinase Ror2 acts as an alternative receptor or co-receptor for Wnt5a and mediates Wnt5a-induced convergent extension movements during embryogenesis in mice and Xenopus as well as the polarity and migration of several cell types during development. However, little is known about whether Ror2 function is conserved in other vertebrates or is involved in other non-canonical Wnt ligands in vivo. In this study we demonstrated that overexpression of dominant-negative ror2 (ror2-TM) mRNA in zebrafish embryos resulted in convergence and extension defects and incompletely separated eyes, which is consistent with observations from slb/wnt11 mutants or wnt11 knockdown morphants. Moreover, the co-injection of ror2-TM mRNA and a wnt11 morpholino or the coexpression of ror2 and wnt11 in zebrafish embryos synergetically induced more severe convergence and extension defects. Transplantation studies further demonstrated that the Ror2 receptor responded to the Wnt11 ligand and regulated cell migration and cell morphology during gastrulation. DnRor2 inhibited the action of Wnt11, which was revealed by a decreased percentage of Wnt11-induced convergence and extension defects. Ror2 physically interacts with Wnt11. Theintracellular Tyr-647andSer-863 sites ofRor2are essential for mediating the action of Wnt11. Dishevelled and RhoA act downstream of Wnt11-Ror2 to regulate convergence and extension movements. Overall, our data suggest an important role of Ror2 in mediating Wnt11 signaling and in regulating convergence and extension movements in zebrafish.


Subject(s)
Cell Movement/physiology , Gastrula/embryology , Gastrulation/physiology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dishevelled Proteins , Gastrula/cytology , Gene Knockdown Techniques , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Gen Comp Endocrinol ; 197: 82-91, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24362258

ABSTRACT

The growth and developmental rate of developing embryos and fetus are tightly controlled and coordinated to maintain proper body shape and size. The insulin receptor substrate (IRS) proteins, key intracellular transducers of insulin and insulin-like growth factor signaling, play essential roles in the regulation of growth and development. A short isoform of apoptosis-stimulating protein of p53 2 (ASPP2) was recently identified as a binding partner of IRS-1 and IRS-2 in mammalian cells in vitro. However, it is unclear whether ASPP2 plays any role in vertebrate embryonic growth and development. Here, we show that zebrafish Aspp2a and Aspp2b negatively regulate embryonic growth without affecting developmental rate. Human ASPP2 had similar effects on body growth in zebrafish embryos. Aspp2a and 2b inhibit Akt signaling. This inhibition was reversed by coinjection of myr-Akt1, a constitutively active form of Akt1. Zebrafish Aspp2a and Aspp2b physically bound with Irs-1, and the growth inhibitory effects of ASPP2/Aspp2 depend on the presence of their ankyrin repeats and SH3 domains. These findings uncover a novel role of Aspp2 in regulating vertebrate embryonic growth.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation, Developmental/physiology , Insulin Receptor Substrate Proteins/metabolism , Insulin/metabolism , MAP Kinase Signaling System/physiology , Zebrafish/embryology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ankyrins/metabolism , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Body Size/physiology , Evolution, Molecular , HEK293 Cells , Humans , Proto-Oncogene Proteins c-akt/metabolism , Somatomedins/metabolism , Somites/embryology , Somites/physiology , Zebrafish/genetics , Zebrafish Proteins , src Homology Domains/physiology
15.
Am J Physiol Regul Integr Comp Physiol ; 303(11): R1165-74, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23034716

ABSTRACT

Hypoxia-inducible factors 1-3 (HIF1-3) are transcription factors that regulate gene expression in response to hypoxia. Compared with our extensive understanding of HIF-1 and HIF-2, our knowledge of HIF-3 is limited. In this study, we characterized the zebrafish hif-3α gene and determined its temporal and spatial expression, physiological regulation, and biological activity. We show that the chromosomal location, gene structure, and protein structure of zebrafish hif-3α are similar to its mammalian orthologs. When tagged with enhanced green fluorescent protein and transfected into cultured cells, zebrafish Hif-3α was localized in the nucleus and stimulated reporter gene expression in a hypoxia response element-dependent manner. During early development, hif-3α mRNA was detected in all tissues with higher levels in the head. This expression pattern became more apparent in larvae at the 72, 96, and 120 hours post fertilization stages. In the adult stage, hif-3α mRNA was detected in all examined tissues with the highest levels in the ovary. Hypoxia treatment increased Hif-3α protein levels in both embryos and adults. Hypoxia also increased hif-3α mRNA expression levels, and this regulation was tissue-specific. Expression of a stabilized form of Hif-1α in zebrafish embryos increased the expression of igfbp-1a, a Hif-1 target gene, whereas it did not change hif-3α mRNA levels, suggesting that hif-3α is not a Hif-1α target. These results provide new information about the structural and functional conservation, spatial and temporal expression, and physiological regulation of hif-3α in a teleost model organism.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Evolution, Molecular , HEK293 Cells , HeLa Cells , Humans , Hypoxia , Larva/metabolism , Molecular Sequence Data , Oxygen/metabolism , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/physiology , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish/embryology , Zebrafish/genetics
16.
Int J Cancer ; 130(9): 2003-12, 2012 May 01.
Article in English | MEDLINE | ID: mdl-21618524

ABSTRACT

Hypoxia stimulates tumor angiogenesis by inducing the expression of angiogenic molecules. The negative regulators of this process, however, are not well understood. Here, we report that hypoxia induced the expression of insulin-like growth factor binding protein-6 (IGFBP-6), a tumor repressor, in human and rodent vascular endothelial cells (VECs) via a hypoxia-inducible factor (HIF)-mediated mechanism. Addition of human IGFBP-6 to cultured human VECs inhibited angiogenesis in vitro. An IGFBP-6 mutant with at least 10,000-fold lower binding affinity for IGFs was an equally potent inhibitor of angiogenesis, suggesting that this action of IGFBP-6 is IGF-independent. The functional relationship between IGFBP-6 and vascular endothelial growth factor (VEGF), a major hypoxia-inducible angiogenic molecule, was examined. While VEGF alone increased angiogenesis in vitro, co-incubation with IGFBP-6 abolished VEGF-stimulated angiogenesis. The in vivo role of IGFBP-6 in angiogenesis was tested in flk1:GFP zebrafish embryos, which exhibit green fluorescence protein in developing vascular endothelium, permitting visualization of developing blood vessels. Injection of human IGFBP-6 mRNA reduced the number of embryonic inter-segmental blood vessels by ∼40%. This anti-angiogenic activity is conserved in zebrafish because expression of zebrafish IGFBP-6b had similar effects. To determine the anti-angiogenic effect of IGFBP-6 in a tumor model, human Rh30 rhabdomyosarcoma cells stably transfected with IGFBP-6 were inoculated into athymic BALB/c nude mice. Vessel density was 52% lower in IGFBP-6-transfected xenografts than in vector control xenografts. These results suggest that the expression of IGFBP-6 in VECs is up-regulated by hypoxia and IGFBP-6 inhibits angiogenesis in vitro and in vivo.


Subject(s)
Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Protein 6/metabolism , Insulin-Like Growth Factor I/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Female , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Insulin-Like Growth Factor Binding Protein 6/genetics , Insulin-Like Growth Factor I/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Rats , Rhabdomyosarcoma/blood supply , Rhabdomyosarcoma/metabolism , Vascular Endothelial Growth Factor A/genetics , Zebrafish
17.
PLoS One ; 7(12): e52674, 2012.
Article in English | MEDLINE | ID: mdl-23300740

ABSTRACT

REDD1/redd1 is a stress-response gene that is induced under various stressful conditions such as hypoxia, DNA damage, and energy stress. The increased REDD1 inhibits mTOR signaling and cell growth. Here we report an unexpected role of Redd1 in regulating dorsoventral patterning in zebrafish embryos and the underlying mechanisms. Zebrafish redd1 mRNA is maternally deposited. Although it is ubiquitously detected in many adult tissues, its expression is highly tissue-specific and dynamic during early development. Hypoxia and heat shock strongly induce redd1 expression in zebrafish embryos. Knockdown of Redd1 using two independent morpholinos results in dorsalized embryos and this effect can be rescued by injecting redd1 mRNA. Forced expression of Redd1 ventralizes embryos. Co-expression of Redd1 with Wnt3a or a constitutively active form of ß-catenin suggests that Redd1 alters dorsoventral patterning by antagonizing the Wnt/ß-catenin signaling pathway. These findings have unraveled a novel role of Redd1 in early development by antagonizing Wnt/ß-catenin signaling.


Subject(s)
Body Patterning , Intracellular Signaling Peptides and Proteins/genetics , Stress, Physiological , Wnt Signaling Pathway , Zebrafish Proteins/genetics , Animals , Embryo, Nonmammalian/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Morpholinos/genetics , Organ Specificity , Wnt3A Protein/metabolism , Zebrafish , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , beta Catenin/metabolism
18.
J Cell Sci ; 124(Pt 11): 1925-35, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21558420

ABSTRACT

IGFBP3 is a multi-functional protein that has IGF-dependent and IGF-independent actions in cultured cells. Here we show that the IGF binding domain (IBD), nuclear localization signal (NLS) and transactivation domain (TA) are conserved and functional in zebrafish Igfbp3. The in vivo roles of these domains were investigated by expression of Igfbp3 and its mutants in zebrafish embryos. Igfbp3, and its NLS and TA mutants had equally strong dorsalizing effects. Human IGFBP3 had similar dorsalizing effects in zebrafish embryos. The activities of IBD and IBD+NLS mutants were lower, but they still caused dorsalization. Thus, the IGF-independent action of Igfbp3 is not related to NLS or TA in this in vivo model. We next tested the hypothesis that Igfbp3 exerts its IGF-independent action by affecting Bmp signaling. Co-expression of Igfbp3 with Bmp2b abolished Bmp2b-induced gene expression and inhibited its ventralizing activity. Biochemical assays and in vitro experiments revealed that IGFBP3 bound BMP2 and inhibited BMP2-induced Smad signaling in cultured human cells. In vivo expression of Igfbp3 increased chordin expression in zebrafish embryos by alleviating the negative regulation of Bmp2. The elevated level of Chordin acted together with Igfbp3 to inhibit the actions of Bmp2. Knockdown of Igfbp3 enhanced the ventralized phenotype caused by chordin knockdown. These results suggest that Igfbp3 exerts its IGF-independent actions by antagonizing Bmp signaling and that this mechanism is conserved.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/metabolism , Zebrafish/embryology , Amino Acid Sequence , Animals , Binding Sites , Bone Morphogenetic Protein 2/metabolism , Cell Line , Conserved Sequence , Gene Expression , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Sequence Data , Nuclear Localization Signals/metabolism , Protein Structure, Tertiary , Protein Transport , Sequence Alignment , Transcriptional Activation , Up-Regulation , Zebrafish/metabolism
19.
Endocrinology ; 152(5): 1860-71, 2011 May.
Article in English | MEDLINE | ID: mdl-21385939

ABSTRACT

Recent genome-wide association studies have implicated the clusterin gene in the etiology of Alzheimer's disease. The expression and function of clusterin in the developing brain, however, is poorly understood. In this study, we have characterized the zebrafish clusterin gene and determined its structural conservation, developmental expression, and physiological regulation. The structure of the zebrafish clusterin gene and protein is similar to its human orthologue. Biochemical assays show that zebrafish Clusterin is a secreted protein that cannot bind IGFs. In adult zebrafish, clusterin mRNA is detected in many tissues. In early development, clusterin mRNA becomes detectable at 12 h postfertilization, and its levels gradually increase thereafter. In situ hybridization analysis indicates that clusterin mRNA is specifically expressed in the developing diencephalic and myelencephalic choroid plexus. Among various stresses tested, heat shock, but not hypoxic or ionic stresses, increases the levels of clusterin mRNA. Inhibition of the IGF-I receptor-mediated signaling or overexpression of IGF ligands did not change clusterin mRNA levels. In comparison, inhibition or targeted knockdown of Notch signaling significantly increased clusterin mRNA expression in choroid plexus. These results suggest that clusterin is a marker of choroid plexus in zebrafish, and its expression in the developing choroid plexus is under the regulation of Notch but not IGF signaling.


Subject(s)
Choroid Plexus/metabolism , Clusterin/genetics , Receptor, IGF Type 1/genetics , Receptors, Notch/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Choroid Plexus/embryology , Clusterin/classification , Clusterin/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , In Situ Hybridization , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Microscopy, Fluorescence , Molecular Sequence Data , Phylogeny , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction/drug effects , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
20.
Article in Chinese | MEDLINE | ID: mdl-17511158

ABSTRACT

OBJECTIVE: To explore the importance and preventive measure of vocal scar after fiber laryngoscope surgery. METHOD: The preventive measures such as treatment of the pathogeny, voice exercise, adjustment of operative skill, vocal rest after operation and drug treatment for vocal scars in 350 patients with polyps of vocal cord, vocal nodules and vocal cyst after fiber laryngoscope surgery were adopted in order to reduce the rate of vocal scar. The rate of vocal scar was calculated and analysed to evaluate the effect of preventive measures two month later. RESULT: The incidence rate of vocal scar after fiber laryngoscope surgery was 12.3%. And vocal scar was the main difficulty in vocalizing after operation. There is yet no specific treatment for vocal scar. Prevention is more important. Preventive measures should be carried out through the perioperative period, i. e., before, in and after the surgery. CONCLUSION: The prevention of vocal scar complication is very important in the perioperative period of fiber laryngoscope surgery. And as the preventive measures are adopted, the incidence rate of vocal scar will be significantly reduced.


Subject(s)
Cicatrix/prevention & control , Postoperative Complications/prevention & control , Vocal Cords/pathology , Adolescent , Adult , Aged , Cicatrix/etiology , Female , Humans , Laryngoscopy/adverse effects , Male , Middle Aged , Voice Quality , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...