Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Transplant Proc ; 56(2): 440-447, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38368129

ABSTRACT

BACKGROUND: Humanized mouse models with engraftment of human peripheral blood mononuclear cells (PBMCs) or hematopoietic stem cells (HSCs) are effective tools for the study of human immunity. Busulfan has been used as a substitute for irradiation in human hematopoietic stem cell (HSC) transplantation models, but it has not been tested in human peripheral blood mononuclear cell (PBMC) transplantation models. METHODS: This study evaluated PBMC engraftment using cytometry and enzyme-linked immunosorbent assay (ELISA) in female NOD.CB17/Prkdcscid/JKrb/ IL2 receptor γ-/- (NIG) mice treated with busulfan. RESULTS: In this model, the percentage of human CD3+ T cell engraftment in the blood was 28.2%, with dominant infiltration of CD8+ cells in the spleen 3 weeks post PBMC transplantation. Production of human cytokines, including Interleukin (IL)-12p70, IL-4, IL-5, IFN-γ, IL-6, IL-8, IL-22, Tumor Necrosis Factor alpha, and IL-10, was determined in mice treated with busulfan. CONCLUSIONS: Our findings demonstrate that busulfan treatment is a beneficial alternative for simple and efficient PBMC engraftment in a rodent model, possibly helping to evaluate human immunity in preclinical studies.


Subject(s)
Busulfan , Leukocytes, Mononuclear , Humans , Female , Animals , Mice , Mice, SCID , Mice, Inbred NOD , Transplantation, Heterologous
2.
Cell Mol Life Sci ; 80(3): 69, 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36821008

ABSTRACT

Animal models have been utilized to understand the pathogenesis of Zellweger spectrum disorders (ZSDs); however, the link between clinical manifestations and molecular pathways has not yet been clearly established. We generated peroxin 5 homozygous mutant zebrafish (pex5-/-) to gain insight into the molecular pathogenesis of peroxisome dysfunction. pex5-/- display hallmarks of ZSD in humans and die within one month after birth. Fasting rapidly depletes lipids and glycogen in pex5-/- livers and expedites their mortality. Mechanistically, deregulated mitochondria and mechanistic target of rapamycin (mTOR) signaling act together to induce metabolic alterations that deplete hepatic nutrients and accumulate damaged mitochondria. Accordingly, chemical interventions blocking either the mitochondrial function or mTOR complex 1 (mTORC1) or a combination of both improve the metabolic imbalance shown in the fasted pex5-/- livers and extend the survival of animals. In addition, the suppression of oxidative stress by N-acetyl L-cysteine (NAC) treatment rescued the apoptotic cell death and early mortality observed in pex5-/-. Furthermore, an autophagy activator effectively ameliorated the early mortality of fasted pex5-/-. These results suggest that fasting may be detrimental to patients with peroxisome dysfunction, and that modulating the mitochondria, mTORC1, autophagy activities, or oxidative stress may provide a therapeutic option to alleviate the symptoms of peroxisomal diseases associated with metabolic dysfunction.


Subject(s)
Fasting , Mitochondria , Peroxisome-Targeting Signal 1 Receptor , Zebrafish , Animals , Humans , Autophagy/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Peroxisome-Targeting Signal 1 Receptor/genetics , Peroxisome-Targeting Signal 1 Receptor/metabolism
3.
Pharmacol Res ; 184: 106423, 2022 10.
Article in English | MEDLINE | ID: mdl-36064078

ABSTRACT

BMP2 is clinically used as an ectopic bone inducer and plays a significant role in bone development, formation, and diseases. Chitinase 3-like 1 protein (Chi3L1) is found in the skeletal system. However, Chi3L1-mediated bone metabolism and aging-related bone erosion via BMP2 signaling have not yet been demonstrated. Herein, Chi3L1 increased BMP2-induced osteoblast differentiation in mesenchymal precursor cells and human primary osteoblasts. Chi3L1KO(-/-) showed abnormal bone development, and primary osteoblasts isolated from Chi3L1KO(-/-) exhibited impaired osteoblast differentiation and maturation. Chi3L1 also potentiated BMP2 signaling and RUNX2 expression in primary osteoblasts. Chi3L1 interacted with BMPRIa, which increased the surface expression of BMPRIa and promoted BMP2 signaling to induce osteoblast differentiation. Chi3L1KO(-/-) mice showed bone formation reduced with a decrease in RUNX2 expression in calvarial defects. Chi3L1KO(-/-) mice exhibited aging-related osteoporotic bone loss with decreases in the levels of RUNX2 and OPG, while serum PYD level and osteoclast number increased. Chi3L1 increased OPG via non-canonical BMP2 signaling in osteoblasts, which suppressed osteoclastogenesis in BMMs. Furthermore, ROC analysis showed that serum Chi3L1 level clinically decreased in osteoporosis patients. Our findings demonstrate that Chi3L1 promotes bone formation, suppresses osteoclastogenesis, and prevents aging-related osteoporosis.


Subject(s)
Chitinases , Osteoporosis , Animals , Biomarkers/metabolism , Cell Differentiation , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/metabolism , Chitinases/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mice , Osteoblasts/metabolism , Osteogenesis , Osteoporosis/drug therapy , Osteoporosis/metabolism
4.
Lab Anim Res ; 36: 27, 2020.
Article in English | MEDLINE | ID: mdl-32817844

ABSTRACT

Genome editing has recently emerged as a powerful tool for generating mutant mice. Small deletions of nucleotides in the target genes are frequently found in CRISPR/Cas9 mediated mutant mice. However, there are very few reports analyzing the phenotypes in small deleted mutant mice generated by CRISPR/Cas9. In this study, we generated a mutant by microinjecting sgRNAs targeting the IL2 receptor γ gene and Cas9 protein, into the cytoplasm of IVF-derived NOD.CB17/Prkdcscid/JKrb (NOD/SCID) mice embryos, and further investigated whether a 2 bp deletion of the IL2 receptor γ gene affects severe deficiency of immune cells as seen in NOD/LtSz-scid IL2 receptor γ-/- (NSG) mice. Our results show that the thymus weight of mutant mice is significantly less than that of NOD/SCID mice, whereas the spleen weight was marginally less. T and B cells in the mutant mice were severely deficient, and NK cells were almost absent. In addition, tumor growth was exceedingly increased in the mutant mice transplanted with HepG2, Raji and A549 cells, but not in nude and NOD/SCID mice. These results suggest that the NOD/SCID mice with deletion of 2 bp in the IL2 receptor γ gene shows same phenotype as NSG mice. Taken together, our data indicates that small deletions by genome editing is sufficient to generate null mutant mice.

5.
Cell Commun Signal ; 18(1): 5, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32127023

ABSTRACT

BACKGROUND: Chitinase 3 like 1 protein (Chi3L1) is expressed in several cancers, and a few evidences suggest that the secreted Chi3L1 contributes to tumor development. However, the molecular mechanisms of intracellular Chi3L1 are unknown in the lung tumor development. METHODS: In the present study, we generated Chi3L1 knockout mice (Chi3L1KO(-/-)) using CRISPR/Cas9 system to investigate the role of Chi3L1 on lung tumorigenesis. RESULTS: We established lung metastasis induced by i.v. injections of B16F10 in Chi3L1KO(-/-). The lung tumor nodules were significantly reduced in Chi3L1KO(-/-) and protein levels of p53, p21, BAX, and cleaved-caspase 3 were significantly increased in Chi3L1KO(-/-), while protein levels of cyclin E1, CDK2, and phsphorylation of STAT3 were decreased in Chi3L1KO(-/-). Allograft mice inoculated with B16F10 also suppressed tumor growth and increased p53 and its target proteins including p21 and BAX. In addition, knockdown of Chi3L1 in lung cancer cells inhibited lung cancer cell growth and upregulated p53 expression with p21 and BAX, and a decrease in phosphorylation of STAT3. Furthermore, we found that intracellular Chi3L1 physically interacted and colocalized with p53 to inhibit its protein stability and transcriptional activity for target genes related with cell cycle arrest and apoptosis. In lung tumor patient, we clinically found that Chi3L1 expression was upregulated with a decrease in p53 expression, as well as we validated that intracellular Chi3L1 was colocalized, reversely expressed, and physically interacted with p53, which results in suppression of the expression and function of p53 in lung tumor patient. CONCLUSIONS: Our studies suggest that intracellular Chi3L1 plays a critical role in the lung tumorigenesis by regulating its novel target protein, p53 in both an in vitro and in vivo system.


Subject(s)
Carcinogenesis/pathology , Chitinase-3-Like Protein 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Allografts , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Chitinase-3-Like Protein 1/chemistry , Down-Regulation , Humans , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Protein Binding , Protein Stability , Transcription, Genetic , Ubiquitination
6.
Sci Rep ; 9(1): 13911, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31558757

ABSTRACT

Cpf1 is an RNA-guided endonuclease that can be programmed to cleave DNA targets. Specific features, such as containing a short crRNA, creating a staggered cleavage pattern and having a low off-target rate, render Cpf1 a promising gene-editing tool. Here, we present a new Cpf1 ortholog, EeCpf1, as a genome-editing tool; this ortholog is derived from the gut bacterial species Eubacterium eligens. EeCpf1 exhibits a higher cleavage activity with the Mn2+ metal cofactor and efficiently cuts the target DNA with an engineered, nucleotide extended crRNA at the 5' target site. When mouse blastocysts were injected with multitargeting crRNAs against the IL2R-γ gene, an essential gene for immunodeficient mouse model production, EeCpf1 efficiently generated IL2R-γ knockout mice. For the first time, these results demonstrate that EeCpf1 can be used as an in vivo gene-editing tool for the production of knockout mice. The utilization of engineered crRNA with multiple target sites will help to explore the in vivo DNA cleavage activities of Cpf1 orthologs from other species that have not been demonstrated.


Subject(s)
Bacterial Proteins/metabolism , Endonucleases/metabolism , Eubacterium/enzymology , Gene Editing/methods , Animals , Bacterial Proteins/genetics , Blastocyst/metabolism , Endonucleases/genetics , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Magnesium/metabolism , Mice , Mice, Inbred C57BL , RNA, Circular/genetics
7.
Mol Cells ; 40(4): 271-279, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28359144

ABSTRACT

Ran-binding protein family member, RanBP9 has been reported in various basic cellular mechanisms and neuropathological conditions including schizophrenia. Previous studies have reported that RanBP9 is highly expressed in the mammalian brain and retina; however, the role of RanBP9 in retinal development is largely unknown. Here, we present the novel and regulatory roles of RanBP9 in retinal development of a vertebrate animal model, zebrafish. Zebrafish embryos exhibited abundant expression of ranbp9 in developing brain tissues as well as in the developing retina. Yeast two-hybrid screening demonstrated the interaction of RanBP9 with Mind bomb, a component of Notch signaling involved in both neurogenesis and neural disease autism. The interaction is further substantiated by co-localization studies in cultured cells. Knockdown of ranbp9 resulted in retinal dysplasia with defective proliferation of retinal cells, downregulation of neuronal differentiation marker huC, elevation of neural proliferation marker her4, and alteration of cell cycle marker p57kip2. Expression of the Müller glial cell marker glutamine synthase was also affected in knockdown morphants. Our results suggest that Mind bomb-binding partner RanBP9 plays a role during retinal cell development of zebrafish embryogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Nuclear Proteins/metabolism , Retina/embryology , Ubiquitin-Protein Ligases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Brain/embryology , Brain/metabolism , COS Cells , Cell Proliferation , Chlorocebus aethiops , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Cytoskeletal Proteins/genetics , Down-Regulation , ELAV-Like Protein 3/genetics , ELAV-Like Protein 3/metabolism , Ependymoglial Cells/physiology , Gene Knockdown Techniques , Neurogenesis/physiology , Nuclear Proteins/genetics , Retina/cytology , Retina/metabolism , Retinal Dysplasia/genetics , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
8.
Anim Cells Syst (Seoul) ; 21(2): 93-99, 2017.
Article in English | MEDLINE | ID: mdl-30460056

ABSTRACT

Fish lineage-specific gene, sinup [Siaz-interacting nuclear protein], modulates neural plate formation in embryogenesis and shares homology with human TPX2 protein, a member of the vertebrate mitogen-activating protein family. In spite of the presence of the TPX2 domain in Sinup, its cellular function has been unknown. As an initial approach to this question, we expressed Sinup by injecting sinup-EGFP mRNAs into zebrafish embryos at the one- to two-cell stage. First of all, Sinup-EGFP was associated with centrosomes and mitotic spindles. In particular, Sinup was localized to the spindle poles and midbody microtubules during the period between anaphase and cytokinesis. Second, various deleted mutants of Sinup-EGFP failed to be associated with the centrosomes and mitotic spindles. Third, a Sinup mutant, where the 144th Serine residue was converted to alanine, not only disturbed the mitotic spindle organization, such as multipolar spindles, fragmented spindle poles, and flattened spindles, but also arrested the cell cycle at metaphase and cell movement. Finally, Sinup is phosphorylated by Aurora A and the 144th Serine mutant of Sinup is partially phosphorylated by Aurora A kinase. We thus propose that Sinup is an essential element for the integrity of centrosomes and mitotic spindle fibers as well as for the normal process of cell cycle and cellular movement in vertebrate embryos.

9.
Sci Rep ; 5: 18011, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26657540

ABSTRACT

Estrogen-related receptor alpha (ESRRa) regulates a number of cellular processes including development of bone and muscles. However, direct evidence regarding its involvement in cartilage development remains elusive. In this report, we establish an in vivo role of Esrra in cartilage development during embryogenesis in zebrafish. Gene expression analysis indicates that esrra is expressed in developing pharyngeal arches where genes necessary for cartilage development are also expressed. Loss of function analysis shows that knockdown of esrra impairs expression of genes including sox9, col2a1, sox5, sox6, runx2 and col10a1 thus induces abnormally formed cartilage in pharyngeal arches. Importantly, we identify putative ESRRa binding elements in upstream regions of sox9 to which ESRRa can directly bind, indicating that Esrra may directly regulate sox9 expression. Accordingly, ectopic expression of sox9 rescues defective formation of cartilage induced by the knockdown of esrra. Taken together, our results indicate for the first time that ESRRa is essential for cartilage development by regulating sox9 expression during vertebrate development.


Subject(s)
Chondrogenesis , Gene Expression Regulation, Developmental , Receptors, Estrogen/metabolism , SOX9 Transcription Factor/genetics , Zebrafish/genetics , Zebrafish/metabolism , Animals , Branchial Region/embryology , Cartilage/embryology , Cartilage/metabolism , Cell Survival/genetics , Chondrocytes/metabolism , Chondrogenesis/genetics , Embryonic Development/genetics , Gene Knockdown Techniques , Neural Crest/embryology , Nucleotide Motifs , Protein Binding , Receptors, Estrogen/genetics , Response Elements , Zebrafish/embryology , ERRalpha Estrogen-Related Receptor
10.
PLoS One ; 9(10): e109517, 2014.
Article in English | MEDLINE | ID: mdl-25347788

ABSTRACT

OBJECTIVE: It has been shown that Mindbomb (Mib), an E3 Ubiquitin ligase, is an essential modulator of Notch signaling during development. However, its effects on vascular development remain largely unknown. APPROACHES AND RESULTS: We identified a number of novel proteins that physically interact with Mib, including the Factor Inhibiting Hypoxia Inducible Factor 1 (FIH-1, also known as HIF1AN) from a yeast two hybrid screen, as previously reported. In cultured cells, FIH-1 colocalizes with Mib1, corroborating their potential interaction. In zebrafish embryos, FIH-1 appears to modulate VEGF-A signaling activity; depletion of fih-1 induces ectopic expression of vascular endothelial growth factor-a (vegfa) and leads to exuberant ectopic sprouts from intersegmental vessels (ISVs). Conversely, over-expression of fih-1 substantially attenuates the formation of ISVs, which can be rescued by concurrent over-expression of vegfa, indicating that FIH-1/HIF1AN may fine tune VEGF-A signaling. CONCLUSIONS: Taken together, our data suggest that FIH-1 interacts with Mib E3 Ubiquitin ligase and modulates vascular development by attenuating VEGF-A signaling activity.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Neovascularization, Physiologic/physiology , Ubiquitin-Protein Ligases/metabolism , Angiogenesis Inducing Agents/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Gene Expression , Hypoxia-Inducible Factor 1/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Physiologic/drug effects , Protein Binding , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
11.
Mol Cells ; 37(1): 74-80, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24552713

ABSTRACT

The peroxisome is an intracellular organelle that responds dynamically to environmental changes. Various model organisms have been used to study the roles of peroxisomal proteins in maintaining cellular homeostasis. By taking advantage of the zebrafish model whose early stage of embryogenesis is dependent on yolk components, we examined the developmental roles of the D-bifunctional protein (Dbp), an essential enzyme in the peroxisomal ß-oxidation. The knockdown of dbp in zebrafish phenocopied clinical manifestations of its deficiency in human, including defective craniofacial morphogenesis, growth retardation, and abnormal neuronal development. Overexpression of murine Dbp rescued the morphological phenotypes induced by dbp knockdown, indicative of conserved roles of Dbp during zebrafish and mammalian development. Knockdown of dbp impaired normal development of blood, blood vessels, and most strikingly, endoderm-derived organs including the liver and pancreas - a phenotype not reported elsewhere in connection with peroxisome dysfunction. Taken together, our results demonstrate for the first time that zebrafish might be a useful model animal to study the role of peroxisomes during vertebrate development.


Subject(s)
Peroxisomal Disorders/enzymology , Peroxisomal Multifunctional Protein-2/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Conserved Sequence , Disease Models, Animal , Embryonic Development , Gastrointestinal Tract/abnormalities , Gene Expression , Gene Knockdown Techniques , Genetic Complementation Test , Humans , Mice , Molecular Sequence Data , Neurogenesis , Peroxisomal Disorders/genetics , Peroxisomal Multifunctional Protein-2/metabolism , Peroxisomes/enzymology , Yolk Sac/metabolism , Zebrafish/embryology , Zebrafish Proteins/metabolism
12.
Biochem Biophys Res Commun ; 442(1-2): 22-7, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24211584

ABSTRACT

Fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, is an anti-hyperlipidemic agent that has been widely used in the treatment of dyslipidemia. In this study, we examined the effect of fenofibrate on liver damage caused by refeeding a high-fat diet (HFD) in mice after 24h fasting. Here, we showed that refeeding HFD after fasting causes liver damage in mice determined by liver morphology and liver cell death. A detailed analysis revealed that hepatic lipid droplet formation is enhanced and triglyceride levels in liver are increased by refeeding HFD after starvation for 24h. Also, NF-κB is activated and consequently induces the expression of TNF-α, IL1-ß, COX-2, and NOS2. However, treating with fenofibrate attenuates the liver damage and triglyceride accumulation caused by the fasting-refeeding HFD process. Fenofibrate reduces the expression of NF-κB target genes but induces genes for peroxisomal fatty acid oxidation, peroxisome biogenesis and mitochondrial fatty acid oxidation. These results strongly suggest that the treatment of fenofibrate ameliorates the liver damage induced by fasting-refeeding HFD, possibly through the activation of fatty acid oxidation.


Subject(s)
Diet, High-Fat/adverse effects , Fasting/adverse effects , Fenofibrate/administration & dosage , Hypolipidemic Agents/administration & dosage , Liver/drug effects , PPAR alpha/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Fatty Acids/metabolism , Ligands , Liver/metabolism , Liver/pathology , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Oxidation-Reduction , Triglycerides/metabolism
13.
Mol Cells ; 36(2): 145-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23852131

ABSTRACT

During skeletal development, both osteogenic and chondrogenic programs are initiated from multipotent mesenchymal cells, requiring a number of signaling molecules, transcription factors, and downstream effectors to orchestrate the sophisticated process. Col10a1, an important downstream effector gene, has been identified as a marker for maturing chondrocytes in higher vertebrates, such as mammals and birds. In zebrafish, this gene has been shown to be expressed in both osteoblasts and chondrocytes, but no study has reported its role in osteoblast development. To initially delineate the osteogenic program from chondrogenic lineage development, we used the zebrafish col10a1 promoter to establish a transgenic zebrafish expressing a GFP reporter specifically in osteoblast-specific bone structures that do not involve cartilaginous programs. A construct harboring a -2.2-kb promoter region was found to be sufficient to drive the reporter gene in osteoblast-specific bone structures within the endogenous col10a1 expression domain, confirming that separable cis-acting elements exist for distinct cell type-specific expression of col10a1 during zebrafish skeletal development. The -2.2-kb col10a1:GFP transgenic zebrafish marking only bone structures derived from osteoblasts will undoubtedly be an invaluable tool for identifying and characterizing molecular events driving osteoblast development in zebrafish, which may further provide a differential mechanism where col10a1 is involved in the development of chondrocytes undergoing maturation in other vertebrate systems.


Subject(s)
Animals, Genetically Modified , Collagen Type X/genetics , Green Fluorescent Proteins/genetics , Osteoblasts/metabolism , Osteogenesis , Zebrafish/genetics , Animals , Chondrocytes/metabolism , Collagen Type X/metabolism , Green Fluorescent Proteins/metabolism , Promoter Regions, Genetic , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Dev Dyn ; 237(12): 3921-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19035353

ABSTRACT

XB51 protein is known to interact with the amino-terminal of the X11L protein and to be involved in Abeta40 generation, a hallmark of Alzheimer's disease. In this study, we isolated a zebrafish xb51 homologue and analyzed its spatio-temporal expression pattern during early brain development. The xb51 transcript was first detected in the forebrain at 22 hr post-fertilization. Expression of xb51 in the brain persisted by 36 hpf and became more complex in the brain after 48 hpf. The detailed expression domain of xb51 in the dorsal telencephalon was defined by several molecular markers: emx1, dlx2, lim1, islet1, neurod4/zath3, ngn1, her4, and elavl3/huC. The location of xb51-expressing cells was restricted in a subset of cells positive for elavl3/huC and acetylated alpha-tubulin, markers of differentiating and/or differentiated neurons. Together, these results suggest that xb51 may be required for maturation and maintenance of xb51-expressing neurons in the forebrain.


Subject(s)
Alzheimer Disease , Calcium-Binding Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Molecular Sequence Data , Neurons/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Telencephalon/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
15.
J Biol Chem ; 281(47): 36391-400, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17003037

ABSTRACT

Mutations in Drosophila neuralized (Dneur) result in a variety of developmental defects that closely resemble those of Notch mutants and other Notch pathway mutants. However, mice with disrupted neur1 do not show any aberrant cell fate specifications in neurogenesis and somitogenesis. Thus, we speculated that other vertebrate neur homolog(s) might compensate for loss of the neur gene. Here, we report the paralog of mouse Neur1, named Neuralized-2 (Neur2), which is a ubiquitin-protein isopeptide ligase (E3) that interacts with and ubiquitinates Delta. Both murine Neur1 and Neur2 have similar degrees of homology to DNeur, and neur2 is expressed in patterns similar to those of neur1 in embryos, suggesting potential functional redundancy. Interestingly, two distinct classes of E3 ligases, Mind bomb-1 (Mib1) and Neur2, have cooperative but distinct roles in Delta endocytosis to Hrs-positive vesicles, i.e. Mib1 functions in the initial step of Delta endocytosis, and Neur2 is required for targeting endocytosed Delta to Hrs-positive vesicles. Thus, our study provides a new insight into how distinct E3 ligases work together in the endocytic pathways for Notch signaling.


Subject(s)
Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Mutation , Nerve Tissue Proteins/physiology , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , Animals , Drosophila melanogaster , Humans , Ligands , Mice , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Sequence Homology, Amino Acid , Zebrafish , Zebrafish Proteins/metabolism
16.
FEBS Lett ; 580(18): 4409-16, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16857196

ABSTRACT

Notch signaling has an evolutionarily conserved function for cell fate determination and stem cell maintenance. Previously, we identified a novel component of the Notch signaling pathway in zebrafish, mind bomb, which encodes an E3 ubiquitin ligase essential for Notch signal activation. Further studies showed that Mind bomb(-/-) mouse embryos exhibited pan-Notch phenotypes in various tissues, suggesting that Mind bomb function is conserved in mammals. Therefore we sought to understand the various molecular partners of Mind bomb using yeast two-hybrid screening. In this search we identified Sorting nexin 5 (Snx5) as a novel interacting partner of Mind bomb. Furthermore we demonstrated that Snx5 colocalizes with Mind bomb in early endosomal compartments, suggesting that Snx5 is important for Mind bomb trafficking. In addition, we identified zebrafish orthologue of Snx5 and showed that snx5 is predominantly expressed in hematopoietic and endothelial precursor cells in zebrafish. We also found defects in hematopoiesis and blood vessel development in snx5 morpholino-injected embryos. Taken together, we show that Snx5, a novel interacting partner of Mind bomb, may have an essential role for cell fate determination in early development.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Endothelial Cells/cytology , Humans , Immunoprecipitation , Mice , Neovascularization, Physiologic , Sorting Nexins , Stem Cells/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/chemistry , Vesicular Transport Proteins , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/chemistry
17.
Development ; 132(15): 3459-70, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16000382

ABSTRACT

The Delta-Notch signaling pathway is an evolutionarily conserved intercellular signaling mechanism essential for cell fate specification. Mind bomb 1 (Mib1) has been identified as a ubiquitin ligase that promotes the endocytosis of Delta. We now report that mice lacking Mib1 die prior to embryonic day 11.5, with pan-Notch defects in somitogenesis, neurogenesis, vasculogenesis and cardiogenesis. The Mib1-/- embryos exhibit reduced expression of Notch target genes Hes5, Hey1, Hey2 and Heyl, with the loss of N1icd generation. Interestingly, in the Mib1-/- mutants, Dll1 accumulated in the plasma membrane, while it was localized in the cytoplasm near the nucleus in the wild types, indicating that Mib1 is essential for the endocytosis of Notch ligand. In accordance with the pan-Notch defects in Mib1-/- embryos, Mib1 interacts with and regulates all of the Notch ligands, jagged 1 and jagged 2, as well as Dll1, Dll3 and Dll4. Our results show that Mib1 is an essential regulator, but not a potentiator, for generating functional Notch ligands to activate Notch signaling.


Subject(s)
Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Blood Vessels/embryology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Ligands , Mice , Mice, Knockout , Receptors, Cell Surface/metabolism , Receptors, Notch , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases/deficiency
18.
J Biol Chem ; 280(23): 22335-42, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15824097

ABSTRACT

The zebrafish gene, mind bomb (mib), encodes a protein that positively regulates of the Delta-mediated Notch signaling. It interacts with the intracellular domain of Delta to promote its ubiquitination and endocytosis. In our search for the mouse homologue of zebrafish mind bomb, we cloned two homologues in the mouse genome: a mouse orthologue (mouse mib1) and a paralogue, named mind bomb-2 (mib2), which is evolutionarily conserved from Drosophila to human. Both Mib1 and Mib2 have an E3 ubiquitin ligase activity in their C-terminal RING domain and interact with Xenopus Delta (XD) via their N-terminal region. Mib2 is also able to ligate ubiquitin to XD and shift the membrane localization of Delta to intracellular vesicles. Importantly, Mib2 rescues both the neuronal and vascular defects in the zebrafish mib(ta52b) mutants. In contrast to the functional similarities between Mib1 and Mib2, mib2 is highly expressed in adult tissues, but almost not at all in embryos, whereas mib1 is abundantly expressed in both embryos and adult tissues. These data suggest that Mib2 has functional similarities to Mib1, but might have distinct roles in Notch signaling as an E3 ubiquitin ligase.


Subject(s)
Membrane Proteins/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , Animals , Blotting, Northern , Cell Line , Cell Membrane/metabolism , Cloning, Molecular , Drosophila , Drosophila Proteins , HeLa Cells , Humans , Immunoprecipitation , In Situ Hybridization , Ligands , Mice , Molecular Sequence Data , Mutation , Phenotype , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptors, Notch , Sequence Homology, Amino Acid , Signal Transduction , Subcellular Fractions/metabolism , Transfection , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Xenopus , Zebrafish , Zebrafish Proteins
19.
Biochem Biophys Res Commun ; 312(3): 601-7, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14680807

ABSTRACT

Three genes, alpha-synuclein, parkin, and ubiquitin C-terminal hydrolase L1 (UCH-L1), have been associated with inherited forms of Parkinson's disease (PD), although their in vivo functions have remained largely unknown. To develop an animal model for the molecular study of PD, we cloned zebrafish uch-L1 cDNA and its gene promoter. Sequence analysis revealed that the zebrafish Uch-L1 is highly homologous (79%) to the human UCH-L1, which is a member of the deubiquitinating enzymes. By whole-mount in situ hybridization, we examined the spatiotemporal expression of uch-L1 mRNA in developing zebrafish embryos. The uch-L1 mRNAs are detected in neuronal cells at the first day of embryo development. The expression domain of uch-L1 overlaps with that of tyrosine hydroxylase, a molecular marker for dopaminergic neurons, in the ventral diencephalon, an equivalent structure to the substantia nigra where PD progresses in human. To further analyze the tissue-specific regulation of uch-L1 gene expression, we also tested its gene promoter activity and showed a preferential neuronal expression in transient transgenic zebrafish embryos. These results suggest that uch-L1 may have an important role in the development of neuronal cells in early embryos as well as in the degeneration and disease of neuronal cells in late adult brain.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Parkinson Disease/genetics , Parkinson Disease/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Humans , Mice , Models, Animal , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Sequence Homology , Species Specificity , Tissue Distribution , Ubiquitin Thiolesterase/chemistry , Zebrafish/embryology , Zebrafish Proteins/chemistry
20.
Biochem Biophys Res Commun ; 308(1): 184-90, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12890499

ABSTRACT

We have isolated a basic helix-loop-helix (bHLH) gene homologous to the Drosophila proneural gene atonal, termed zath3, from zebrafish. zath3 is expressed in neurons of the central nervous system and in subsets of cranial ganglia. Zebrafish mindbomb (mib) mutants have a higher density of zath3 expressing cells and narrowminded (nrd) mutants lack zath3 expression in a domain corresponding to primary sensory neurons showing that the expression of zath3 is regulated by both mib and nrd. Injection of synthetic zath3 RNA into zebrafish embryos expands the neural plate size, promotes ectopic expression of neuronal markers, and partially rescues the deficit of sensory neurons seen in nrd mutants. Interfering with zath3 function using antisense morpholino oligonucleotides (MO) has no significant effect on early neurogenesis. However, a double knock down of zath3 and neurogenin1 (ngn1), another atonal homologue, with morpholinos (MOs) leads to more severe defects in neurogenesis than are seen with ngn1 MO alone: a subtle reduction of motor and inter-neurons, and an almost complete loss all cranial ganglia. This study suggests that zath3 and ngn1 have partially overlapping roles in early neurogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Helix-Loop-Helix Motifs , Nerve Tissue Proteins/genetics , Nervous System/embryology , Zebrafish Proteins/genetics , Zebrafish/embryology , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Cloning, Molecular , DNA Primers , DNA-Binding Proteins/chemistry , Humans , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Zebrafish Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL