Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cells Dev ; 177: 203908, 2024 03.
Article in English | MEDLINE | ID: mdl-38403117

ABSTRACT

The Notch signaling pathway, an evolutionarily highly conserved pathway, participates in various essential physiological processes in organisms. Activation of Notch signaling in the canonical manner requires the combination of ligand and receptor. There are two ligands of Notch in Drosophila: Delta (Dl) and Serrate (Ser). A mutation mf157 is identified for causing nicks of fly wings in genetic analysis from a mutant library (unpublished) that was established previously. Immunofluorescent staining illustrates that mf157 represses the expression of Cut and Wingless (Wg), the targets of Notch signaling. MARCM cloning analysis reveals that mf157 functions at the same level or the upstream of ligands of Notch in signaling sending cells. Sequencing demonstrates that mf157 is a novel allele of the Ser gene. Subsequently, mf553 and mf167 are also identified as new alleles of Ser from our library. Furthermore, the complementary assays and the examination of transcripts confirm the sequencing results. Besides, the repressed phenotypes of Notch signaling were reverted by transposon excision experiments of mf157. In conclusion, we identify three fresh alleles of Ser. Our works supply additional genetic resources for further study of functions of Ser and Notch signaling regulation.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila/metabolism , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Alleles , Membrane Proteins/genetics , Membrane Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism
2.
Cell Mol Life Sci ; 80(7): 182, 2023 Jun 18.
Article in English | MEDLINE | ID: mdl-37330998

ABSTRACT

The Notch pathway is an ancient, evolutionary conserved intercellular signaling mechanism that is involved in cell fate specification and proper embryonic development. The Jagged2 gene, which encodes a ligand for the Notch family of receptors, is expressed from the earliest stages of odontogenesis in epithelial cells that will later generate the enamel-producing ameloblasts. Homozygous Jagged2 mutant mice exhibit abnormal tooth morphology and impaired enamel deposition. Enamel composition and structure in mammals are tightly linked to the enamel organ that represents an evolutionary unit formed by distinct dental epithelial cell types. The physical cooperativity between Notch ligands and receptors suggests that Jagged2 deletion could alter the expression profile of Notch receptors, thus modifying the whole Notch signaling cascade in cells within the enamel organ. Indeed, both Notch1 and Notch2 expression are severely disturbed in the enamel organ of Jagged2 mutant teeth. It appears that the deregulation of the Notch signaling cascade reverts the evolutionary path generating dental structures more reminiscent of the enameloid of fishes rather than of mammalian enamel. Loss of interactions between Notch and Jagged proteins may initiate the suppression of complementary dental epithelial cell fates acquired during evolution. We propose that the increased number of Notch homologues in metazoa enabled incipient sister cell types to form and maintain distinctive cell fates within organs and tissues along evolution.


Subject(s)
Membrane Proteins , Receptors, Notch , Pregnancy , Female , Mice , Animals , Cell Lineage/genetics , Membrane Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Serrate-Jagged Proteins/metabolism , Cell Differentiation/physiology , Carrier Proteins , Mammals/metabolism
3.
Trends Plant Sci ; 28(7): 841-853, 2023 07.
Article in English | MEDLINE | ID: mdl-37019716

ABSTRACT

The SERRATE (SE) protein is involved in the processing of RNA polymerase II (RNAPII) transcripts. It is associated with different complexes engaged in different aspects of plant RNA metabolism, including assemblies involved in transcription, splicing, polyadenylation, miRNA biogenesis, and RNA degradation. SE stability and interactome properties can be influenced by phosphorylation. SE exhibits an intriguing liquid-liquid phase separation property that may be important in the assembly of different RNA-processing bodies. Therefore, we propose that SE seems to participate in the coordination of different RNA-processing steps and can direct the fate of transcripts, targeting them for processing or degradation when they cannot be properly processed or are synthesized in excess.


Subject(s)
Arabidopsis Proteins , Arabidopsis , MicroRNAs , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , RNA Processing, Post-Transcriptional/genetics , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism , RNA/metabolism , MicroRNAs/genetics , RNA, Plant/genetics , RNA, Plant/metabolism , Gene Expression Regulation, Plant
4.
Hepatology ; 78(5): 1337-1351, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37021797

ABSTRACT

BACKGROUND AND AIMS: Paucity of intrahepatic bile ducts (BDs) is caused by various etiologies and often leads to cholestatic liver disease. For example, in patients with Alagille syndrome (ALGS), which is a genetic disease primarily caused by mutations in jagged 1 ( JAG1) , BD paucity often results in severe cholestasis and liver damage. However, no mechanism-based therapy exists to restore the biliary system in ALGS or other diseases associated with BD paucity. Based on previous genetic observations, we investigated whether postnatal knockdown of the glycosyltransferase gene protein O -glucosyltransferase 1 ( Poglut1) can improve the ALGS liver phenotypes in several mouse models generated by removing one copy of Jag1 in the germline with or without reducing the gene dosage of sex-determining region Y-box 9 in the liver. APPROACH AND RESULTS: Using an ASO established in this study, we show that reducing Poglut1 levels in postnatal livers of ALGS mouse models with moderate to profound biliary abnormalities can significantly improve BD development and biliary tree formation. Importantly, ASO injections prevent liver damage in these models without adverse effects. Furthermore, ASO-mediated Poglut1 knockdown improves biliary tree formation in a different mouse model with no Jag1 mutations. Cell-based signaling assays indicate that reducing POGLUT1 levels or mutating POGLUT1 modification sites on JAG1 increases JAG1 protein level and JAG1-mediated signaling, suggesting a likely mechanism for the observed in vivo rescue. CONCLUSIONS: Our preclinical studies establish ASO-mediated POGLUT1 knockdown as a potential therapeutic strategy for ALGS liver disease and possibly other diseases associated with BD paucity.


Subject(s)
Alagille Syndrome , Glycosyltransferases , Liver , Oligonucleotides, Antisense , Animals , Mice , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Alagille Syndrome/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Calcium-Binding Proteins/genetics , Cholestasis/genetics , Cholestasis/metabolism , Gene Silencing , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Phenotype , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism
5.
Environ Toxicol ; 37(12): 2957-2964, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36039874

ABSTRACT

The purpose of this study is to explore the anti-colorectal cancer of Xiaotansanjiefang, a famous traditional Chinese medicine, and its potential anti-cancer mechanism. In this study, the HCT116 cell spheres were prepared as in vitro study model. We found the Xiaotansanjiefang medication was able to inhibit the proliferation of HCT116 cell spheres in a dose-dependent manner, especially in 3 and 6 mg/ml Xiaotansanjiefang medication treated groups. We also found the high concentration of Xiaotansanjiefang medication could suppress the migration and promote the apoptosis of HCT116 cell spheres. Moreover, we found the expression of Jagged 1, Notch 3, Snail, and Hes 1 were decreased in HCT116 cell spheres treated with Xiaotansanjiefang medication. Furthermore, the proliferation and apoptosis behaviors of HCT116 cell spheres treated with Xiaotansanjiefang medication were reversed with the addition of Jagged 1 Fc chimera protein. The expression of Jagged 1, Notch 3, Snail, and Hes 1 were also increased again in HCT116 cells treated with Xiaotansanjiefang medication plus with Jagged 1 Fc chimera protein. The presented study may provide a promising strategy to treat and prevent colorectal cancer.


Subject(s)
Intercellular Signaling Peptides and Proteins , Neoplasms , Jagged-1 Protein/metabolism , Serrate-Jagged Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation , Membrane Proteins/metabolism , Signal Transduction
6.
Vascul Pharmacol ; 145: 107087, 2022 08.
Article in English | MEDLINE | ID: mdl-35792302

ABSTRACT

BACKGROUND: Notch signaling is an evolutionarily conserved pathway that functions via direct cell-cell contact. The Notch ligand Jagged1 (Jag1) has been extensively studied in vascular development, particularly for its role in smooth muscle cell maturation. Endothelial cell-expressed Jag1 is essential for blood vessel formation by signaling to nascent vascular smooth muscle cells and promoting their differentiation. Given the established importance of Jag1 in endothelial cell/smooth muscle crosstalk during development, we sought to determine the extent of this communication in the adult vasculature for blood vessel function and homeostasis. METHODS: We conditionally deleted Jag1 in endothelial cells of adult mice and examined the phenotypic consequences on smooth muscle cells of the vasculature. RESULTS: Our results show that genetic loss of Jag1 in endothelial cells has a significant impact on Notch signaling and vascular smooth muscle function in mature blood vessels. Endothelial cell-specific deletion of Jag1 causes a concomitant loss of JAG1 and NOTCH3 expression in vascular smooth muscle cells, resulting in a transition to a less differentiated state. Aortic vascular smooth muscle cells isolated from the endothelial cell-specific Jag1 deficient mice retain an altered phenotype in culture with fixed changes in gene expression and reduced Notch signaling. Utilizing comparative RNA-sequence analysis, we found that Jag1 deficiency preferentially affects extracellular matrix and adhesion protein gene expression. Vasoreactivity studies revealed a reduced contractile response and impaired agonist-induced relaxation in endothelial cell Jag1-deficient aortas compared to controls. CONCLUSIONS: These data are the first to demonstrate that Jag1 in adult endothelial cells is required for the regulation and homeostasis of smooth muscle cell function in arterial vessels partially through the autoregulation of Notch signaling and cell matrix/adhesion components in smooth muscle cells.


Subject(s)
Endothelial Cells , Receptors, Notch , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Phenotype , RNA/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism
7.
JCI Insight ; 7(15)2022 08 08.
Article in English | MEDLINE | ID: mdl-35819850

ABSTRACT

Mucosecretory lung disease compromises airway epithelial function and is characterized by goblet cell hyperplasia and ciliated cell hypoplasia. Goblet and ciliated cell types are derived from tracheobronchial stem/progenitor cells via a Notch-dependent mechanism. Although specific arrays of Notch receptors regulate cell fate determination, the function of the ligands Jagged1 (JAG1) and JAG2 is unclear. This study examined JAG1 and JAG2 function using human air-liquid-interface cultures that were treated with γ-secretase complex (GSC) inhibitors, neutralizing peptides/antibodies, or WNT/ß-catenin pathway antagonists/agonists. These experiments revealed that JAG1 and JAG2 regulated cell fate determination in the tracheobronchial epithelium; however, their roles did not adhere to simple necessity and sufficiency rules. Biochemical studies indicated that JAG1 and JAG2 underwent posttranslational modifications that resulted in generation of a JAG1 C-terminal peptide and regulated the abundance of full-length JAG2 on the cell surface. GSC and glycogen synthase kinase 3 were implicated in these posttranslational events, but WNT agonist/antagonist studies and RNA-Seq indicated a WNT-independent mechanism. Collectively, these data suggest that posttranslational modifications create distinct assemblies of JAG1 and JAG2, which regulate Notch signal strength and determine the fate of tracheobronchial stem/progenitor cells.


Subject(s)
Intercellular Signaling Peptides and Proteins , Lung Diseases , Calcium-Binding Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/genetics , Jagged-2 Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Serrate-Jagged Proteins/metabolism , Signal Transduction
8.
Adv Med Sci ; 67(2): 196-202, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35421813

ABSTRACT

Notch signaling is a highly conserved pathway and it plays an essential role in regulating cellular proliferation, differentiation, and apoptosis. The human Notch family includes four receptors, Notch 1-4, and five ligands, delta-like ligand 1 (DLL1), delta-like ligand 3 (DLL3), delta-like ligand 4 (DLL4), Jagged-1 (JAG1), and Jagged-2 (JAG2). It is widely known, that Notch signaling components are often mutated and have deregulated expression in many types of cancer and other diseases. Thus, various therapeutic approaches targeting receptors and ligands of the Notch pathway are being investigated. Human JAG1 is closely related to tumor biology among the Notch ligands, and recent studies have shown potential for monoclonal antibodies targeting JAG1 in cancer therapy. Therefore, this review focuses on current reports on the significance of JAG1 directed cancer treatment, emphasizing immunotherapy.


Subject(s)
Intercellular Signaling Peptides and Proteins , Neoplasms , Humans , Serrate-Jagged Proteins/metabolism , Jagged-1 Protein/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Calcium-Binding Proteins/metabolism , Ligands , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Neoplasms/therapy , Immunotherapy , Antibodies, Monoclonal/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism
9.
Cell Death Dis ; 12(9): 832, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34482375

ABSTRACT

Oncogenic signaling pathway reprograms cancer cell metabolism to promote aerobic glycolysis in favor of tumor growth. The ability of cancer cells to evade immunosurveillance and the role of metabolic regulators in T-cell functions suggest that oncogene-induced metabolic reprogramming may be linked to immune escape. Notch1 signaling, dysregulated in lung cancer, is correlated with increased glycolysis. Herein, we demonstrate in lung cancer that Notch1 promotes glycolytic gene expression through functional interaction with histone acetyltransferases p300 and pCAF. Notch1 signaling forms a positive feedback loop with TAZ. Notch1 transcriptional activity was increased in the presence of TAZ and the activation was TEAD1 independent. Notably, aerobic glycolysis was critical for Notch1/TAZ axis modulation of lung cancer growth in vitro and in vivo. Increased level of extracellular lactate via Notch1/TAZ axis inhibited cytotoxic T-cell activity, leading to the invasive characteristic of lung cancer cells. Interaction between Notch1 and TAZ promoted aerobic glycolysis and immune escape in lung cancer. Our findings provide potential therapeutic targets against Notch1 and TAZ and would be important for clinical translation in lung cancer.


Subject(s)
Glycolysis , Immune Evasion , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Aerobiosis , Animals , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Genes, Reporter , Glycolysis/genetics , Humans , Immune Evasion/genetics , Killer Cells, Natural/immunology , Lactic Acid/metabolism , Lung Neoplasms/genetics , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Protein Binding , Receptor, Notch1/metabolism , Serrate-Jagged Proteins/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , TEA Domain Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , p300-CBP Transcription Factors/metabolism
10.
Mol Biol Cell ; 32(1): 28-44, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33175598

ABSTRACT

Evolutionarily conserved RNA-binding protein Musashi1 (Msi1) can regulate developmentally relevant genes. Here we report the generation and characterization of a mouse model that allows inducible Msi1 overexpression in a temporal and tissue-specific manner. We show that ubiquitous Msi1 induction in ∼5-wk-old mice delays overall growth, alters organ-to-body proportions, and causes premature death. Msi1-overexpressing mice had shortened intestines, diminished intestinal epithelial cell (IEC) proliferation, and decreased growth of small intestine villi and colon crypts. Although Lgr5-positive intestinal stem cell numbers remained constant in Msi1-overexpressing tissue, an observed reduction in Cdc20 expression provided a potential mechanism underlying the intestinal growth defects. We further demonstrated that Msi1 overexpression affects IEC differentiation in a region-specific manner, with ileum tissue being influenced the most. Ilea of mutant mice displayed increased expression of enterocyte markers, but reduced expression of the goblet cell marker Mucin2 and fewer Paneth cells. A higher hairy and enhancer of split 1:mouse atonal homolog 1 ratio in ilea from Msi1-overexpressing mice implicated Notch signaling in inducing enterocyte differentiation. Together, this work implicates Msi1 in mouse postnatal development of multiple organs, with Notch signaling alterations contributing to intestinal defects. This new mouse model will be a useful tool to further elucidate the role of Msi1 in other tissue settings.


Subject(s)
Growth and Development , Homeostasis , Intestines/growth & development , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cdc20 Proteins/metabolism , Cell Proliferation , Intestinal Mucosa/metabolism , Membrane Proteins/metabolism , Mice, Transgenic , Microvilli/metabolism , Models, Animal , Organ Specificity , Receptors, G-Protein-Coupled/metabolism , Serrate-Jagged Proteins/metabolism , Transgenes , Up-Regulation
11.
Immunol Lett ; 223: 106-114, 2020 07.
Article in English | MEDLINE | ID: mdl-32325090

ABSTRACT

BACKGROUND: Notch pathway is highly conserved across species and is involved in the regulation of cell differentiation and activity both in embryonic development and adult life. Notch signaling has an important role in the development of hematopoietic stem cells and their differentiation to committed lineages, as well as in the regulation of several non-hematopoietic cell lines. OBJECTIVE: As Notch signaling has been implicated in various inflammatory and autoimmune diseases, it is of interest to elucidate what role do Notch receptors and ligands have in inflammatory arthritides. METHODS: We performed a search on the role of Notch receptors (1-4) and Notch ligands Delta-like (DLL) 1, 3, 4 and Jagged (Jag) 1 and 2 in animal models of inflammatory arthritis and most common types of human inflammatory arthritis (rheumatoid arthritis, psoriatic arthritis or ankylosing spondylitis). The initial search identified 135 unique articles, of which 24 were ultimately deemed relevant and included in this systematic review. RESULTS: Overall, identified articles describe roles for Notch ligands and receptors in inflammatory arthritis, with Notch activation resulting in enhanced Th1/17 polarization, osteoclast differentiation, macrophage activation and fibroblast-like synoviocyte proliferation. However, the inhibitory role of Notch signaling, especially by Jag1 is also described. CONCLUSION: There is evidence that Notch pathway activation affects multiple cell lineages present within the arthritic environment, therefore potentially acting as one of the drivers of disease pathogenesis. Since cell lineage-selective transgenic mouse models and specific Notch receptor inhibitors are becoming increasingly available, it can be expected that future research will evaluate whether Notch signaling components initiate crucial pathogenic impulses and, therefore, present viable therapeutic targets in inflammatory arthritis.


Subject(s)
Arthritis/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Osteoclasts/physiology , Receptors, Notch/metabolism , Serrate-Jagged Proteins/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Disease Models, Animal , Humans
12.
Dev Genes Evol ; 230(3): 213-225, 2020 05.
Article in English | MEDLINE | ID: mdl-31960122

ABSTRACT

Size and shape constitute fundamental aspects in the description of morphology. Yet while the developmental-genetic underpinnings of trait size, in particular with regard to scaling relationships, are increasingly well understood, those of shape remain largely elusive. Here we investigate the potential function of the Notch signaling pathway in instructing the shape of beetle horns, a highly diversified and evolutionarily novel morphological structure. We focused on the bull-headed dung beetle Onthophagus taurus due to the wide range of horn sizes and shapes present among males in this species, in order to assess the potential function of Notch signaling in the specification of horn shape alongside the regulation of shape changes with allometry. Using RNA interference-mediated transcript depletion of Notch and its ligands, we document a highly conserved role of Notch signaling in general appendage formation. By integrating our functional genetic approach with a geometric morphometric analysis, we find that Notch signaling moderately but consistently affects horn shape, and does so differently for the horns of minor, intermediate-sized, and major males. Our results suggest that the function of Notch signaling during head horn formation may vary in a complex manner across male morphs, and highlights the power of integrating functional genetic and geometric morphometric approaches in analyzing subtle but nevertheless biologically important phenotypes in the face of significant allometric variation.


Subject(s)
Body Patterning , Coleoptera/growth & development , Coleoptera/genetics , Receptors, Notch/physiology , Serrate-Jagged Proteins/metabolism , Signal Transduction , Animals , Biological Evolution , Coleoptera/anatomy & histology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genes, Insect , Insect Proteins/genetics , Insect Proteins/metabolism , Male , Morphogenesis , Phenotype , RNA Interference , Serrate-Jagged Proteins/genetics , Sex Characteristics
13.
Dev Biol ; 460(1): 77-85, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31866513

ABSTRACT

Spatial patterning during embryonic development emerges from the differentiation of progenitor cells that share the same genetic program. One of the main challenges in systems biology is to understand the relationship between gene network and patterning, especially how the cells communicate to coordinate their differentiation. This review aims to describe the principles of pattern formation from local cell-cell interactions mediated by the Notch signalling pathway. Notch mediates signalling via direct cell-cell contact and regulates cell fate decisions in many tissues during embryonic development. Here, I will describe the patterning mechanisms via different Notch ligands and the critical role of Notch oscillations during the segmentation of the vertebrate body, brain development, and blood vessel formation.


Subject(s)
Body Patterning/physiology , Embryonic Development/physiology , Neovascularization, Physiologic/physiology , Neurogenesis/physiology , Receptors, Notch/metabolism , Animals , Cell Communication/physiology , Gene Expression Regulation, Developmental/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Serrate-Jagged Proteins/metabolism , Signal Transduction/physiology , Somites/embryology , Transcription Factor HES-1/metabolism , Zebrafish
14.
Elife ; 82019 10 21.
Article in English | MEDLINE | ID: mdl-31631837

ABSTRACT

Notch signaling regulates cell fate selection during development in multiple organs including the lung. Previous studies on the role of Notch in the lung focused mostly on Notch pathway core components or receptor-specific functions. It is unclear, however, how Jagged or Delta-like ligands collectively or individually (Jag1, Jag2, Dll1, Dll4) influence differentiation of airway epithelial progenitors. Using mouse genetic models we show major differences in Jag and Dll in regulation and establishment of cell fate. Jag ligands had a major impact in balancing distinct cell populations in conducting airways, but had no role in the establishment of domains and cellular abundance in the neuroendocrine (NE) microenvironment. Surprisingly, Dll ligands were crucial in restricting cell fate and size of NE bodies and showed an overlapping role with Jag in differentiation of NE-associated secretory (club) cells. These mechanisms may potentially play a role in human conditions that result in aberrant NE differentiation, including NE hyperplasias and cancer.


Subject(s)
Cell Differentiation , Intracellular Signaling Peptides and Proteins/metabolism , Lung/cytology , Membrane Proteins/metabolism , Serrate-Jagged Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cellular Microenvironment , Ligands , Mice , Neurosecretory Systems/metabolism , Receptors, Notch/metabolism , Signal Transduction
15.
Int J Biochem Cell Biol ; 116: 105599, 2019 11.
Article in English | MEDLINE | ID: mdl-31494224

ABSTRACT

Yorki (Yki), a transcriptional co-activator that is a key component of the Hippo pathway, induces the transcription of a number of targets that promote cell proliferation and survival. Bombyx mori Yki3 (BmYki3), with 445 amino acid residues, facilitates cell migration and cell division, and enlarges cultured cell and wing disc size. In this study, cellular localization, transcriptional co-activator activity, cell migration, cell cycle, and cell size were characterized in alternative isoforms of BmYki. BmYki1 and BmYki3 are mainly located in the cytoplasm and nucleus, respectively, while, BmYki2 is located in both the cytoplasm and nucleus. The mutation BmYki1S97A (S97mutated to A) was transported from the cytoplasm to nucleus. Cell migration, cell cycle, and cell size could be enhanced by BmYki, however, the effect of BmYki1 and BmYki2 on cell proliferation was less compared to BmYki3. Moreover, wing discs could be enlarged by overexpressing BmYki1 or BmYki2 isoforms. Dual-luciferase reporter assay showed that BmYki3 had the highest activity to B. mori ovarian tumor gene. In BmN cells overexpressing one of the BmYki isoforms, expression levels of kibra ortholog (kibra), inhibitor of apoptosis protein (iap), four-jointed (fj), expanded (ex), crumbs (crb) and BMP and activin membrane-bound inhibitor homolog (Bmpr) genes were upregulated, while those of α-catenin (α-cat), decapentaplegic (dpp), serrate (serr) and signal transducer and activator of transcription (stat) genes were down-regulated. There was some difference in the regulation of gene expression between different isoforms. These results suggested that the activity of BmYki isoforms was different in the silkworm.


Subject(s)
Bombyx/genetics , Insect Proteins/genetics , Ovary/metabolism , Trans-Activators , Transcriptional Activation , Wings, Animal/metabolism , Animals , Bombyx/metabolism , Cell Cycle , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cell Size , Cell Survival , Cytosol/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Insect Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Ovary/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wings, Animal/cytology , alpha Catenin/genetics , alpha Catenin/metabolism
16.
Cell Mol Life Sci ; 76(1): 163-178, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30327840

ABSTRACT

Low-density lipoprotein receptor-related protein 4 (LRP4) is a multi-functional protein implicated in bone, kidney and neurological diseases including Cenani-Lenz syndactyly (CLS), sclerosteosis, osteoporosis, congenital myasthenic syndrome and myasthenia gravis. Why different LRP4 mutation alleles cause distinct and even contrasting disease phenotypes remain unclear. Herein, we utilized the zebrafish model to search for pathways affected by a deficiency of LRP4. The lrp4 knockdown in zebrafish embryos exhibits cyst formations at fin structures and the caudal vein plexus, malformed pectoral fins, defective bone formation and compromised kidney morphogenesis; which partially phenocopied the human LRP4 mutations and were reminiscent of phenotypes resulting form a perturbed Notch signaling pathway. We discovered that the Lrp4-deficient zebrafish manifested increased Notch outputs in addition to enhanced Wnt signaling, with the expression of Notch ligand jagged1b being significantly elevated at the fin structures. To examine conservatism of signaling mechanisms, the effect of LRP4 missense mutations and siRNA knockdowns, including a novel missense mutation c.1117C > T (p.R373W) of LRP4, were tested in mammalian kidney and osteoblast cells. The results showed that LRP4 suppressed both Wnt/ß-Catenin and Notch signaling pathways, and these activities were perturbed either by LRP4 missense mutations or by a knockdown of LRP4. Our finding underscore that LRP4 is required for limiting Jagged-Notch signaling throughout the fin/limb and kidney development, whose perturbation representing a novel mechanism for LRP4-related diseases. Moreover, our study reveals an evolutionarily conserved relationship between LRP4 and Jagged-Notch signaling, which may shed light on how the Notch signaling is fine-tuned during fin/limb development.


Subject(s)
Gene Expression Regulation, Developmental , LDL-Receptor Related Proteins/genetics , Receptors, Notch/metabolism , Serrate-Jagged Proteins/metabolism , Signal Transduction , Zebrafish Proteins/genetics , Zebrafish/genetics , Animal Fins/embryology , Animal Fins/metabolism , Animals , Extremities/embryology , Extremities/physiology , Gene Knockdown Techniques , HEK293 Cells , Humans , Kidney/embryology , Kidney/metabolism , LDL-Receptor Related Proteins/metabolism , Mutation , Mutation, Missense , Organogenesis , Wnt Signaling Pathway , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
17.
Development ; 145(16)2018 08 13.
Article in English | MEDLINE | ID: mdl-30042180

ABSTRACT

T-cell development is a complex dynamic process that relies on ordered stromal signals delivered to thymus-seeding progenitors that migrate throughout different thymus microenvironments (TMEs). Particularly, Notch signaling provided by thymic epithelial cells (TECs) is crucial for T-cell fate specification and generation of mature T cells. Four canonical Notch ligands (Dll1, Dll4, Jag1 and Jag2) are expressed in the thymus, but their spatial distribution in functional TMEs is largely unknown, especially in humans, and their impact on Notch1 activation during T-lymphopoiesis remains undefined. Based on immunohistochemistry and quantitative confocal microscopy of fetal, postnatal and adult human and mouse thymus samples, we show that spatial regulation of Notch ligand expression defines discrete Notch signaling niches and dynamic species-specific TMEs. We further show that Notch ligand expression, particularly DLL4, is tightly regulated in cortical TECs during human thymus ontogeny and involution. Also, we provide the first evidence that NOTCH1 activation is induced in vivo in CD34+ progenitors and developing thymocytes at particular cortical niches of the human fetal and postnatal thymus. Collectively, our results show that human thymopoiesis involves complex spatiotemporal regulation of Notch ligand expression, which ensures the coordinated delivery of niche-specific NOTCH1 signals required for dynamic T-cell development.


Subject(s)
Receptor, Notch1/metabolism , Thymus Gland/growth & development , Thymus Gland/metabolism , Adolescent , Adult , Aging/metabolism , Animals , Antigens, CD34/metabolism , Child , Fetus/embryology , Gene Expression Regulation, Developmental , Humans , Infant , Infant, Newborn , Ligands , Mice , Mice, Inbred C57BL , Organogenesis , Serrate-Jagged Proteins/metabolism , Signal Transduction , Stem Cells/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/embryology
18.
Sci Rep ; 8(1): 9277, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29915190

ABSTRACT

Radiation exposure severely damages the hematopoietic system. Although several radio-protectors have been proposed to prevent radiation-induced damage, most agents have limited efficacy. In the present study, we investigated whether mesenchymal stem cells (MSCs) could contribute to the expansion of hematopoietic cells and mitigate radiation-induced hematopoietic injury in vitro and in vivo. We found that co-culture with MSCs promoted hematopoietic progenitor/stem cell (HPSCs) maintenance by providing a bone marrow-like microenvironment. In addition, we showed that MSCs prevented radiation-induced damage to HPSCs, as evidenced by the lack of DNA damage and apoptosis. Intravenously injected MSCs rapidly migrated to the bone marrow (BM) and prevented loss of BM cellularity, which reduced lethality and ameliorated pancytopenia in the BM of whole body-irradiated mice. We demonstrated that MSC-derived Jagged1 attenuated radiation-induced cytotoxicity of HPSCs, and that this was mediated by Notch signaling and expression of downstream proteins Bcl2 and p63 in HPSCs. In addition, Notch2 depletion significantly reduced the MSC-mediated radio-protective effect in human- and mouse-derived HPSCs. Collectively, our data show that activation of Notch and its associated downstream signaling pathways prevent radiation-induced hematopoietic injury. Therefore, enhancing Jagged1-Notch2 signaling could provide therapeutic benefit by protecting the hematopoietic system against damage after radiation.


Subject(s)
Hematopoietic Stem Cells/pathology , Mesenchymal Stem Cells/metabolism , Radiation Injuries/pathology , Receptor, Notch2/metabolism , Animals , Apoptosis/radiation effects , Bone Marrow/pathology , Bone Marrow/radiation effects , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Radiation Injuries/metabolism , Radiation, Ionizing , Serrate-Jagged Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Umbilical Cord/cytology
19.
Int J Mol Med ; 42(1): 674-684, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693137

ABSTRACT

The clinical use of doxorubicin (Dox) is limited by its cardiotoxicity. The fundamental changes it induces include interstitial myocardial fibrosis and the appearance of senescent cardiomyocytes. Mesenchymal stem cell (MSC)­based therapies have also been reported to modulate cellular senescence, and have been used effectively to treat age­related cardiovascular diseases. In the present study, the Transwell system was used to coculture H9c2 cells with MSCs, and their proliferation and viability were assessed. The expression of senescence­related genes p53 and p16, and telomere length were measured using reverse transcription­quantitative polymerase chain reaction analysis, and the Jagged­1/Notch­1 signaling pathway was detected using western blot analysis. The results revealed that Dox induced the senescence of H9c2 cells, characterized by a low proliferation rate, poor viability, reduced telomere length and impaired telomerase activity, and by marked increases in the expression of p53 and p16. By contrast, when cocultured with MSCs in the presence of Dox, H9c2 cell proliferation and viability increased, whereas the expression levels of p53 and p16 decreased, and telomere length and telomerase activity increased. The mechanism underlying the antisenescence function of MSCs was clarified, which involved the vascular endothelial growth factor (VEGF)/Jagged­1/Notch­1/transforming growth factor­ß1 (TGF­ß1) signaling pathway. It was confirmed that inhibiting VEGF, or silencing Jagged­1 or Notch­1 with small interfering RNA, or using recombinant TGF­ß1 eliminated the antisenescence effects of MSCs on the Dox­treated H9c2 cells. The results revealed that MSCs rescued H9c2 cells from Dox­induced senescence through the release of VEGF, which activated the Jagged­1/Notch­1 signaling pathway, leading to the inhibition of TGF­ß1 release. Therefore, treatment with MSCs may have important therapeutic implications on the attenuation of cardiotoxicity in patients with cancer treated with Dox.


Subject(s)
Cellular Senescence/drug effects , Doxorubicin/pharmacology , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Receptors, Notch/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line , Coculture Techniques , Male , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Rats, Sprague-Dawley , Serrate-Jagged Proteins/metabolism , Signal Transduction/drug effects
20.
Exp Cell Res ; 359(1): 257-265, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28734865

ABSTRACT

BACKGROUND: Epithelial to mesenchymal transition (EMT) is considered to be important for cancer invasion and metastasis. Tumour hypoxia, in addition to Transforming Growth Factor-ß (TGF-ß) and Notch, amongst others, have been suggested to be involved in EMT. We therefore investigated if hypoxia, TGF-ß1 and the Notch ligand Jagged-1 alone induced morphological changes with corresponding EMT signatures in different epithelial breast cancer cell lines in vitro. Furthermore, we also studied whether or not TGF-ß1, or Jagged-1 in combination with hypoxia added any effect on EMT. METHODS: The cells were exposed to normoxia or hypoxia alone or in combination with TGF-ß1 or Jagged-1. Morphological responses to treatment were investigated by light microscopy, and changes in markers for EMT and hypoxia were evaluated by western blot analysis and immunofluorescence studies. RESULTS: One of the four cell lines (MCF7) became elongated and highly multipolar, indicative of EMT, following hypoxia, TGF-ß1 and Jagged-1 treatment per se with the most distinct morphological shift seen with Jagged-1 treatment in combination with hypoxia. Also, when regarding hypoxia, MCF7 cells showed the greatest change in EMT-markers of the four cell lines tested, but these changes were not consistent with a typical EMT pattern. The morphology of BT474 cells was not altered following Jagged-1 treatment, however, Jagged-1 increased E-cadherin levels. Morphology was changed following TGF-ß1 treatment of BT474 cells, but it did not affect E-cadherin levels. Neither Jagged-1 nor TGF-ß1 altered the levels of Vimentin in the BT474 cell line. The E-cadherin responses to hypoxia varied with end-point in both MCF7 and BT474 cells, and in most cases were not consistent with EMT. CONCLUSION: Our results using four different breast cancer cell lines in vitro do not provide evidence that EMT is induced by hypoxia alone or in combination with TGF-ß1 or the Notch ligand Jagged-1. The inconsistency in morphological appearance and EMT-markers, as well as the time dependent variation in E-cadherin responses could not support EMT. Importantly, there was not one single common response pattern to the stimuli used, suggesting that cell lines with different hormone statuses display individual traits that respond differently to the stimuli applied. Thus, based on the present results, common statements that single factors by themselves can induce EMT seem questionable.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Hormones/metabolism , Mesoderm/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Hypoxia/drug effects , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ligands , Mesoderm/drug effects , Receptors, Notch/metabolism , Serrate-Jagged Proteins/metabolism , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...