Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Cell Sci ; 136(16)2023 08 15.
Article in English | MEDLINE | ID: mdl-37519219

ABSTRACT

The ErbB-family receptors play pivotal roles in the proliferation, migration and survival of epithelial cells. Because our knowledge on the ErbB-family receptors has been largely obtained by the exogenous application of their ligands, it remains unknown to what extent each of the ErbB members contributes to these outputs. We here knocked out each ErbB gene, various combinations of ErbB genes or all ErbB genes in Madin-Darby canine kidney cells to delineate the contribution of each gene. ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1, respectively) activation waves during collective cell migration were mediated primarily by ErbB1 and secondarily by the ErbB2 and ErbB3 heterodimer. Either ErbB1 or the ErbB2 and ErbB3 complex was sufficient for the G1/S progression. The saturation cell density was markedly reduced in cells deficient in all ErbB proteins, but not in cells retaining only ErbB2, which cannot bind to ligands. Thus, a ligand-independent ErbB2 activity is sufficient for preventing apoptosis at high cell density. In short, systematic knockout of ErbB-family genes has delineated the roles of each ErbB receptor.


Subject(s)
Receptor, ErbB-2 , Signal Transduction , Animals , Dogs , Ligands , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Phosphorylation , Genes, erbB , Cell Proliferation/genetics , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
2.
Cell Struct Funct ; 48(2): 241-249, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37813623

ABSTRACT

Calcium transients drive cells to discharge prostaglandin E2 (PGE2). We visualized PGE2-induced protein kinase A (PKA) activation and quantitated PGE2 secreted from a single cell by combining fluorescence microscopy and a simulation model. For this purpose, we first prepared PGE2-producer cells that express either an optogenetic or a chemogenetic calcium channel stimulator: OptoSTIM1 or Gq-DREADD, respectively. Second, we prepared reporter cells expressing the Gs-coupled PGE2 reporter EP2 and the PKA biosensor Booster-PKA, which is based on the principle of Förster resonance energy transfer (FRET). Upon the stimulation-induced triggering of calcium transients, a single producer cell discharges PGE2 to stimulate PKA in the surrounding reporter cells. Due to the flow of the medium, the PKA-activated area exhibited a comet-like smear when HeLa cells were used. In contrast, radial PKA activation was observed when confluent MDCK cells were used, indicating that PGE2 diffusion was restricted to the basolateral space. By fitting the radius of the PKA-activated area to a simulation model based on simple diffusion, we estimated that a single HeLa cell secretes 0.25 fmol PGE2 upon a single calcium transient to activate PKA in more than 1000 neighboring cells. This model also predicts that the PGE2 discharge rate is comparable to the diffusion rate. Thus, our method quantitatively envisions that a single calcium transient affects more than 1000 neighboring cells via PGE2.Key words: prostaglandin E2, imaging, intercellular communication, biosensor, quantification.


Subject(s)
Dinoprostone , Fluorescence Resonance Energy Transfer , Animals , Dogs , Humans , HeLa Cells , Dinoprostone/pharmacology , Dinoprostone/metabolism , Madin Darby Canine Kidney Cells
3.
Nat Methods ; 16(10): 1029-1036, 2019 10.
Article in English | MEDLINE | ID: mdl-31501546

ABSTRACT

Optical dimerizers have been developed to untangle signaling pathways, but they are of limited use in vivo, partly due to their inefficient activation under two-photon (2P) excitation. To overcome this problem, we developed Förster resonance energy transfer (FRET)-assisted photoactivation, or FRAPA. On 2P excitation, mTagBFP2 efficiently absorbs and transfers the energy to the chromophore of CRY2. Based on structure-guided engineering, a chimeric protein with 40% FRET efficiency was developed and named 2P-activatable CRY2, or 2paCRY2. 2paCRY2 was employed to develop a RAF1 activation system named 2paRAF. In three-dimensionally cultured cells expressing 2paRAF, extracellular signal-regulated kinase (ERK) was efficiently activated by 2P excitation at single-cell resolution. Photoactivation of ERK was also accomplished in the epidermal cells of 2paRAF-expressing mice. We further developed an mTFP1-fused LOV domain that exhibits efficient response to 2P excitation. Collectively, FRAPA will pave the way to single-cell optical control of signaling pathways in vivo.


Subject(s)
Flavoproteins/metabolism , Fluorescence Resonance Energy Transfer/methods , Optogenetics , Photons , Animals , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice
4.
Am J Pathol ; 191(1): 194-203, 2021 01.
Article in English | MEDLINE | ID: mdl-33069718

ABSTRACT

Contraction of vascular smooth muscle is regulated primarily by calcium concentration and secondarily by ROCK activity within the cells. In contrast to the wealth of information regarding regulation of calcium concentration, little is known about the spatiotemporal regulation of ROCK activity in live blood vessels. Here, we report ROCK activation in subcutaneous arterioles in a transgenic mouse line that expresses a genetically encoded ROCK biosensor based on the principle of FÓ§rster resonance energy transfer by two-photon excitation in vivo imaging. Rapid vasospasm was induced upon laser ablation of arterioles, concomitant with a transient increase in calcium concentration in arteriolar smooth muscles. Unlike the increase in calcium concentration, vasoconstriction and ROCK activation continued for several minutes after irradiation. Both the ROCK inhibitor, fasudil, and the ganglionic nicotinic acetylcholine receptor blocker, hexamethonium, inhibited laser-induced ROCK activation and reduced the duration of vasospasm at the segments distant from the irradiated point. These observations suggest that vasoconstriction is initially triggered by a rapid surge of cytoplasmic calcium and then maintained by sympathetic nerve-mediated ROCK activation.


Subject(s)
Muscle, Smooth, Vascular/enzymology , Vasoconstriction/physiology , rho-Associated Kinases/metabolism , Animals , Autonomic Nervous System/physiology , Calcium Signaling/physiology , Fluorescence Resonance Energy Transfer , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/innervation
5.
FASEB J ; 35(9): e21880, 2021 09.
Article in English | MEDLINE | ID: mdl-34449091

ABSTRACT

In vertebrates, retinal rod and cone photoreceptor cells rely significantly on glycolysis. Lactate released from photoreceptor cells fuels neighboring retinal pigment epithelium cells and Müller glial cells through oxidative phosphorylation. To understand this highly heterogeneous metabolic environment around photoreceptor cells, single-cell analysis is needed. Here, we visualized cellular AMP-activated protein kinase (AMPK) activity and ATP levels in the retina by two-photon microscopy. Transgenic mice expressing a hyBRET-AMPK biosensor were used for measuring the AMPK activity. GO-ATeam2 transgenic mice were used for measuring the ATP level. Temporal metabolic responses were successfully detected in the live retinal explants upon drug perfusion. A glycolysis inhibitor, 2-deoxy-d-glucose (2-DG), activated AMPK and reduced ATP. These effects were clearly stronger in rods than in cones. Notably, rod AMPK and ATP started to recover at 30 min from the onset of 2-DG perfusion. Consistent with these findings, ex vivo electroretinogram recordings showed a transient slowdown in rod dim flash responses during a 60-min 2-DG perfusion, whereas cone responses were not affected. Based on these results, we propose that cones surrounded by highly glycolytic rods become less dependent on glycolysis, and rods also become less dependent on glycolysis within 60 min upon the glycolysis inhibition.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Glycolysis/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Ependymoglial Cells/metabolism , Light , Mice , Mice, Inbred C57BL , Oxidative Phosphorylation , Photons , Retina/metabolism
6.
Cell Struct Funct ; 46(2): 103-111, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34744115

ABSTRACT

IFN-γ secreted from immune cells exerts pleiotropic effects on tumor cells, including induction of immune checkpoint and antigen presentation, growth inhibition, and apoptosis induction. We combined a dual promoter system with an IFN-γ signaling responsive promoter to generate a reporter named the interferon sensing probe (ISP), which quantitates the response to IFN-γ by means of fluorescence and bioluminescence. The integration site effect of the transgene is compensated for by the PGK promoter-driven expression of a fluorescent protein. Among five potential IFN-γ-responsive elements, we found that the interferon γ-activated sequence (GAS) exhibited the best performance. When ISP-GAS was introduced into four cell lines and subjected to IFN-γ stimulation, dose-dependency was observed with an EC50 ranging from 0.2 to 0.9 ng/mL, indicating that ISP-GAS can be generally used as a sensitive biosensor of IFN-γ response. In a syngeneic transplantation model, the ISP-GAS-expressing cancer cells exhibited bioluminescence and fluorescence signals in an IFN-γ receptor-dependent manner. Thus, ISP-GAS could be used to quantitatively monitor the IFN-γ response both in vitro and in vivo.Key words: in vivo imaging, tumor microenvironment, interferon-gamma, dual promoter system.


Subject(s)
Interferon-gamma , Transcription, Genetic , Interferon-gamma/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger , Signal Transduction
7.
Cell Struct Funct ; 45(2): 131-141, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32581154

ABSTRACT

Tissue absorbance, light scattering, and autofluorescence are significantly lower in the near-infrared (NIR) range than in the visible range. Because of these advantages, NIR fluorescent proteins (FPs) are in high demand for in vivo imaging. Nevertheless, application of NIR FPs such as iRFP is still limited due to their dimness in mammalian cells. In contrast to GFP and its variants, iRFP requires biliverdin (BV) as a chromophore. The dimness of iRFP is at least partly due to rapid reduction of BV by biliverdin reductase-A (BLVRA). Here, we established biliverdin reductase-a knockout (Blvra-/-) mice to increase the intracellular BV concentration and, thereby, to enhance iRFP fluorescence intensity. As anticipated, iRFP fluorescence intensity was significantly increased in all examined tissues of Blvra-/- mice. Similarly, the genetically encoded calcium indicator NIR-GECO1, which is engineered based on another NIR FP, mIFP, exhibited a marked increase in fluorescence intensity in mouse embryonic fibroblasts derived from Blvra-/- mice. We expanded this approach to an NIR light-sensing optogenetic tool, the BphP1-PpsR2 system, which also requires BV as a chromophore. Again, deletion of the Blvra gene markedly enhanced the light response in HeLa cells. These results indicate that the Blvra-/- mouse is a versatile tool for the in vivo application of NIR FPs and NIR light-sensing optogenetic tools.Key words: in vivo imaging, near-infrared fluorescent protein, biliverdin, biliverdin reductase, optogenetic tool.


Subject(s)
Biliverdine/metabolism , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Animals , Biliverdine/genetics , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Oxidoreductases Acting on CH-CH Group Donors/metabolism
8.
Development ; 144(2): 334-344, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27993976

ABSTRACT

The heart is an endocrine organ, as cardiomyocytes (CMs) secrete natriuretic peptide (NP) hormones. Since the discovery of NPs, no other peptide hormones that affect remote organs have been identified from the heart. We identified osteocrin (Ostn) as an osteogenesis/chondrogenesis regulatory hormone secreted from CMs in zebrafish. ostn mutant larvae exhibit impaired membranous and chondral bone formation. The impaired bones were recovered by CM-specific overexpression of OSTN. We analyzed the parasphenoid (ps) as a representative of membranous bones. In the shortened ps of ostn morphants, nuclear Yap1/Wwtr1-dependent transcription was increased, suggesting that Ostn might induce the nuclear export of Yap1/Wwtr1 in osteoblasts. Although OSTN is proposed to bind to NPR3 (clearance receptor for NPs) to enhance the binding of NPs to NPR1 or NPR2, OSTN enhanced C-type NP (CNP)-dependent nuclear export of YAP1/WWTR1 of cultured mouse osteoblasts stimulated with saturable CNP. OSTN might therefore activate unidentified receptors that augment protein kinase G signaling mediated by a CNP-NPR2 signaling axis. These data demonstrate that Ostn secreted from the heart contributes to bone formation as an endocrine hormone.


Subject(s)
Chondrogenesis/genetics , Myocytes, Cardiac/metabolism , Osteogenesis/genetics , Skull/embryology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animal Structures/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Chondrogenesis/drug effects , Embryo, Nonmammalian , HEK293 Cells , Heart/metabolism , Humans , Mice , Organogenesis/drug effects , Organogenesis/genetics , Osteogenesis/drug effects , Peptide Hormones/genetics , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Peptide Hormones/physiology , Skull/drug effects , Transcription Factors/metabolism , Transcription Factors/pharmacology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/pharmacology
9.
Pathol Int ; 70(7): 379-390, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32270554

ABSTRACT

The invention of two-photon excitation microscopes widens the potential application of intravital microscopy (IVM) to the broad field of experimental pathology. Moreover, the recent development of fluorescent protein-based, genetically encoded biosensors provides an ideal tool to visualize the cell function in live animals. We start from a brief review of IVM with two-photon excitation microscopes and genetically encoded biosensors based on the principle of Förster resonance energy transfer (FRET). Then, we describe how IVM using biosensors has revealed the pathogenesis of several disease models.


Subject(s)
Biosensing Techniques/methods , Intravital Microscopy/methods , Microscopy, Fluorescence/methods , Pathology/methods , Animals , Disease Models, Animal , Fluorescence Resonance Energy Transfer/methods , Intravital Microscopy/instrumentation , Microscopy, Fluorescence/instrumentation
10.
Cell Struct Funct ; 44(2): 153-169, 2019 Nov 02.
Article in English | MEDLINE | ID: mdl-30905922

ABSTRACT

Two decades have passed since the development of the first calcium indicator based on the green fluorescent protein (GFP) and the principle of Förster resonance energy transfer (FRET). During this period, researchers have advanced many novel ideas for the improvement of such genetically encoded FRET biosensors, which have allowed them to expand their targets from small molecules to signaling proteins and physicochemical properties. Although the merits of "genetically encoded" FRET biosensors became clear once various cell lines were established and several transgenic organisms were generated, the road to these developments was not necessarily a smooth one. Moreover, even today the development of new FRET biosensors remains a very labor-intensive, trial-and-error process. Therefore, at this junction, it may be worthwhile to summarize the progress of the FRET biosensor and discuss the future direction of its development and application.Key words: FRET, biosensor, fluorescent protein.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/genetics , Organisms, Genetically Modified/genetics , Animals , Humans
11.
Cell Struct Funct ; 43(2): 129-140, 2018 Aug 10.
Article in English | MEDLINE | ID: mdl-29962383

ABSTRACT

For more than a century, hematoxylin and eosin (H&E) staining has been the de facto standard for histological studies. Consequently, the legacy of histological knowledge is largely based on H&E staining. Due to the recent advent of multi-photon excitation microscopy, the observation of live tissue is increasingly being used in many research fields. Adoption of this technique has been further accelerated by the development of genetically encoded biosensors for ions and signaling molecules. However, H&E-based histology has not yet begun to fully utilize in vivo imaging due to the lack of proper morphological markers. Here, we report a genetically encoded fluorescent marker, NuCyM (Nucleus, Cytosol, and Membrane), which is designed to recapitulate H&E staining patterns in vivo. We generated a transgenic mouse line ubiquitously expressing NuCyM by using a ROSA26 bacterial artificial chromosome (BAC) clone. NuCyM evenly marked the plasma membrane, cytoplasm and nucleus in most tissues, yielding H&E staining-like images. In the NuCyM-expressing cells, cell division of a single cell was clearly observed as five basic phases during M phase by three-dimensional imaging. We next crossed NuCyM mice with transgenic mice expressing an ERK biosensor based on the principle of Förster resonance energy transfer (FRET). Using NuCyM, ERK activity in each cell could be extracted from the FRET images. To further accelerate the image analysis, we employed machine learning-based segmentation methods, and thereby automatically quantitated ERK activity in each cell. In conclusion, NuCyM is a versatile cell morphological marker that enables us to grasp histological information as with H&E staining.Key words: in vivo imaging, histology, machine learning, molecular activity.


Subject(s)
Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer/methods , Imaging, Three-Dimensional/methods , MAP Kinase Signaling System , Machine Learning , Single-Cell Analysis/methods , Animals , Dogs , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence/methods
12.
Development ; 142(3): 497-509, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25564648

ABSTRACT

ß-catenin regulates the transcription of genes involved in diverse biological processes, including embryogenesis, tissue homeostasis and regeneration. Endothelial cell (EC)-specific gene-targeting analyses in mice have revealed that ß-catenin is required for vascular development. However, the precise function of ß-catenin-mediated gene regulation in vascular development is not well understood, since ß-catenin regulates not only gene expression but also the formation of cell-cell junctions. To address this question, we have developed a novel transgenic zebrafish line that allows the visualization of ß-catenin transcriptional activity specifically in ECs and discovered that ß-catenin-dependent transcription is central to the bone morphogenetic protein (Bmp)-mediated formation of venous vessels. During caudal vein (CV) formation, Bmp induces the expression of aggf1, a putative causative gene for Klippel-Trenaunay syndrome, which is characterized by venous malformation and hypertrophy of bones and soft tissues. Subsequently, Aggf1 potentiates ß-catenin transcriptional activity by acting as a transcriptional co-factor, suggesting that Bmp evokes ß-catenin-mediated gene expression through Aggf1 expression. Bmp-mediated activation of ß-catenin induces the expression of Nr2f2 (also known as Coup-TFII), a member of the nuclear receptor superfamily, to promote the differentiation of venous ECs, thereby contributing to CV formation. Furthermore, ß-catenin stimulated by Bmp promotes the survival of venous ECs, but not that of arterial ECs. Collectively, these results indicate that Bmp-induced activation of ß-catenin through Aggf1 regulates CV development by promoting the Nr2f2-dependent differentiation of venous ECs and their survival. This study demonstrates, for the first time, a crucial role of ß-catenin-mediated gene expression in the development of venous vessels.


Subject(s)
Endothelial Cells/physiology , Gene Expression Regulation, Developmental/physiology , Veins/embryology , beta Catenin/metabolism , Angiogenic Proteins/metabolism , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/metabolism , COUP Transcription Factor II/metabolism , DNA, Complementary/genetics , Endothelial Cells/ultrastructure , HEK293 Cells , Humans , In Situ Nick-End Labeling , Luciferases , Luminescent Proteins , Microscopy, Fluorescence , Morpholinos/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Veins/cytology , Zebrafish , Zebrafish Proteins/metabolism , Red Fluorescent Protein
13.
Mol Cell ; 37(1): 143-9, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20129063

ABSTRACT

Monoubiquitination of proliferating cell nuclear antigen (PCNA) is a critical posttranslational modification essential for DNA repair by translesion DNA synthesis (TLS). The Rad18 E3 ubiquitin ligase cooperates with the E2 Rad6 to monoubiquitinate PCNA in response to DNA damage. How PCNA is monoubiquitinated in unperturbed cells and whether this plays a role in the repair of DNA associated with replication is not known. We show that the CRL4(Cdt2) E3 ubiquitin ligase complex promotes PCNA monoubiqutination in proliferating cells in the absence of external DNA damage independent of Rad18. PCNA monoubiquitination via CRL4(Cdt2) is constitutively antagonized by the action of the ubiquitin-specific protease 1 (USP1). In vitro, CRL4(Cdt2) monoubiquitinates PCNA at Lys164, the same residue that is monoubiquitinated by Rad18. Significantly, CRL4(Cdt2) is required for TLS in nondamaged cells via a mechanism that is dependent on PCNA monoubiquitination. We propose that CRL4(Cdt2) regulates PCNA-dependent TLS associated with stresses accompanying DNA replication.


Subject(s)
DNA Damage , Nuclear Proteins/physiology , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases/physiology , Cell Line , DNA Replication , DNA-Binding Proteins/physiology , HeLa Cells , Humans , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
14.
Cell Struct Funct ; 42(1): 1-13, 2017 Jan 11.
Article in English | MEDLINE | ID: mdl-27885213

ABSTRACT

Genetically-encoded biosensors based on Förster/fluorescence resonance energy transfer (FRET) are versatile tools for studying the spatio-temporal regulation of signaling molecules within not only the cells but also tissues. Perhaps the hardest task in the development of a FRET biosensor for protein kinases is to identify the kinase-specific substrate peptide to be used in the FRET biosensor. To solve this problem, we took advantage of kinase-interacting substrate screening (KISS) technology, which deduces a consensus substrate sequence for the protein kinase of interest. Here, we show that a consensus substrate sequence for ROCK identified by KISS yielded a FRET biosensor for ROCK, named Eevee-ROCK, with high sensitivity and specificity. By treating HeLa cells with inhibitors or siRNAs against ROCK, we show that a substantial part of the basal FRET signal of Eevee-ROCK was derived from the activities of ROCK1 and ROCK2. Eevee-ROCK readily detected ROCK activation by epidermal growth factor, lysophosphatidic acid, and serum. When cells stably-expressing Eevee-ROCK were time-lapse imaged for three days, ROCK activity was found to increase after the completion of cytokinesis, concomitant with the spreading of cells. Eevee-ROCK also revealed a gradual increase in ROCK activity during apoptosis. Thus, Eevee-ROCK, which was developed from a substrate sequence predicted by the KISS technology, will pave the way to a better understanding of the function of ROCK in a physiological context.


Subject(s)
Biosensing Techniques , rho-Associated Kinases/metabolism , Amino Acid Sequence , Blotting, Western , Fluorescence Resonance Energy Transfer , Gene Expression/drug effects , HeLa Cells , Humans , Microscopy, Fluorescence , Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Substrate Specificity , Time-Lapse Imaging , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
15.
Genes Cells ; 21(11): 1164-1175, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27696620

ABSTRACT

Vasculogenesis is essential during early development to construct networks transporting oxygen, blood and nutrients. Tip and stalk cells are specialized endothelial cells involved in novel vessel formation because of their behavior such as sprouting as a leading cell and following tip cell. However, the spatiotemporal details determining the emergence of these cells are unknown. Here, we first show that the ERK activity in endothelial cells represents the precursor of tip and stalk cells for vasculogenesis in zebrafish. We identified that tip and stalk cells for intersegmental vessel (ISV) formation were already specialized in the dorsal aorta (DA) before sprouting. Furthermore, similar specialization was observed in tip cells during parachordal vessel (PAV) formation in lymphangiogenesis. We also identified that the ERK activity was required for specialized cells to emerge from existing blood vessels. Our data show that the ERK activity is a novel marker for determining the emergence of cells in both angiogenesis and lymphangiogenesis.


Subject(s)
Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neovascularization, Physiologic , Zebrafish Proteins/metabolism , Animals , Aorta/cytology , Aorta/embryology , Enzyme Activation , Veins/cytology , Veins/embryology , Zebrafish/embryology
16.
J Biol Chem ; 288(42): 30509-30514, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24022480

ABSTRACT

After acute DNA damage, the cell arrests S-phase progression by inhibiting origin initiation and fork progression to repair damaged DNA. The intra-S-phase checkpoint kinase Chk1 phosphorylates Cdc25A to target the latter for degradation by CRL1(ß-TrCP) and so inhibit origin firing. The mechanism for inhibiting fork progression, however, has not been identified. Here, we show that degradation of p12, the fourth subunit of DNA polymerase δ, is critical for inhibiting fork progression. CRL4(Cdt2) is an E3 ligase that ubiquitinates and degrades p12 after UV treatment. Cells expressing a stable form of p12 exhibit UV-resistant DNA synthesis. DNA fiber assay and alkaline-sucrose gradient assay demonstrate that the impairment of fork progression after DNA damage requires p12 degradation. These results suggest that ubiquitination of p12 through CRL4(Cdt2) and subsequent degradation form one mechanism by which a cell responds to DNA damage to inhibit fork progression.


Subject(s)
DNA Damage , DNA Polymerase III/metabolism , DNA/biosynthesis , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , DNA/genetics , DNA Polymerase III/genetics , Enzyme Stability/genetics , Enzyme Stability/radiation effects , HeLa Cells , Humans , Nuclear Proteins/genetics , Protein Subunits/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/radiation effects , Ultraviolet Rays
17.
Biochem Biophys Res Commun ; 444(1): 13-8, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24406169

ABSTRACT

Protocadherin10 (PCDH10)/OL-protocadherin is a cadherin-related transmembrane protein that has multiple roles in the brain, including facilitating specific cell-cell connections, cell migration and axon guidance. It has recently been reported that PCDH10 functions as a tumor suppressor and that its overexpression inhibits proliferation or invasion of multiple tumor cells. However, the function of PCDH10 in glioblastoma cells has not been elucidated. In contrast to previous reports on other tumors, we show here that suppression of the expression of PCDH10 by RNA interference (RNAi) induces the growth arrest and apoptosis of glioblastoma cells in vitro. Furthermore, we demonstrate that knockdown of PCDH10 inhibits the growth of glioblastoma cells xenografted into immunocompromised mice. These results suggest that PCDH10 is required for the proliferation and tumorigenicity of glioblastoma cells. We speculate that PCDH10 may be a promising target for the therapy of glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cadherins/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Apoptosis , Brain Neoplasms/genetics , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Glioblastoma/genetics , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protocadherins , RNA Interference
18.
Elife ; 122024 Jan 26.
Article in English | MEDLINE | ID: mdl-38276879

ABSTRACT

Prostaglandin E2 (PGE2) is a key player in a plethora of physiological and pathological events. Nevertheless, little is known about the dynamics of PGE2 secretion from a single cell and its effect on the neighboring cells. Here, by observing confluent Madin-Darby canine kidney (MDCK) epithelial cells expressing fluorescent biosensors, we demonstrate that calcium transients in a single cell cause PGE2-mediated radial spread of PKA activation (RSPA) in neighboring cells. By in vivo imaging, RSPA was also observed in the basal layer of the mouse epidermis. Experiments with an optogenetic tool revealed a switch-like PGE2 discharge in response to the increasing cytoplasmic Ca2+ concentrations. The cell density of MDCK cells correlated with the frequencies of calcium transients and the following RSPA. The extracellular signal-regulated kinase (ERK) activation also enhanced the frequency of RSPA in MDCK and in vivo. Thus, the PGE2 discharge is regulated temporally by calcium transients and ERK activity.


Subject(s)
Calcium , Extracellular Signal-Regulated MAP Kinases , Mice , Animals , Dogs , Dinoprostone , Kidney , Phosphorylation
19.
Nat Cell Biol ; 8(6): 615-22, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16715077

ABSTRACT

Semaphorins and their receptors have diverse functions in axon guidance, organogenesis, vascularization and/or angiogenesis, oncogenesis and regulation of immune responses. The primary receptors for semaphorins are members of the plexin family. In particular, plexin-A1, together with ligand-binding neuropilins, transduces repulsive axon guidance signals for soluble class III semaphorins, whereas plexin-A1 has multiple functions in chick cardiogenesis as a receptor for the transmembrane semaphorin, Sema6D, independent of neuropilins. Additionally, plexin-A1 has been implicated in dendritic cell function in the immune system. However, the role of plexin-A1 in vivo, and the mechanisms underlying its pleiotropic functions, remain unclear. Here, we generated plexin-A1-deficient (plexin-A1(-/-)) mice and identified its important roles, not only in immune responses, but also in bone homeostasis. Furthermore, we show that plexin-A1 associates with the triggering receptor expressed on myeloid cells-2 (Trem-2), linking semaphorin-signalling to the immuno-receptor tyrosine-based activation motif (ITAM)-bearing adaptor protein, DAP12. These findings reveal an unexpected role for plexin-A1 and present a novel signalling mechanism for exerting the pleiotropic functions of semaphorins.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Bone and Bones/physiology , Immunity , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Homeostasis , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Signal Transduction
20.
Inflamm Regen ; 43(1): 10, 2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36750856

ABSTRACT

Inflammation can contribute to the development and progression of cancer. The inflammatory responses in the tumor microenvironment are shaped by complex sequences of dynamic intercellular cross-talks among diverse types of cells, and recapitulation of these dynamic events in vitro has yet to be achieved. Today, intravital microscopy with two-photon excitation microscopes (2P-IVM) is the mainstay technique for observing intercellular cross-talks in situ, unraveling cellular and molecular mechanisms in the context of their spatiotemporal dynamics. In this review, we summarize the current state of 2P-IVM with fluorescent indicators of signal transduction to reveal the cross-talks between cancer cells and surrounding cells including both immune and non-immune cells. We also discuss the potential application of red-shifted indicators along with optogenetic tools to 2P-IVM. In an era of single-cell transcriptomics and data-driven research, 2P-IVM will remain a key advantage in delivering the missing spatiotemporal context in the field of cancer research.

SELECTION OF CITATIONS
SEARCH DETAIL