Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
1.
Commun Biol ; 7(1): 286, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38454140

ABSTRACT

Through its involvement in gene transcription and heterochromatin formation, DNA methylation regulates how cells interact with their environment. Nevertheless, the extracellular signaling cues that modulate the distribution of this central chromatin modification are largely unclear. DNA methylation is highly abundant at repetitive elements, but its investigation in live cells has been complicated by methodological challenges. Utilizing a CRISPR/dCas9 biosensor that reads DNA methylation of human α-satellite repeats in live cells, we here uncover a signaling pathway linking the chromatin and transcriptional state of repetitive elements to epithelial adherens junction integrity. Specifically, we find that in confluent breast epithelial cell monolayers, α-satellite repeat methylation is reduced by comparison to low density cultures. This is coupled with increased transcriptional activity at repeats. Through comprehensive perturbation experiments, we identify the junctional protein E-cadherin, which links to the actin cytoskeleton, as a central molecular player for signal relay into the nucleus. Furthermore, we find that this pathway is impaired in cancer cells that lack E-cadherin and are not contact-inhibited. This suggests that the molecular connection between cell density and repetitive element methylation could play a role in the maintenance of epithelial tissue homeostasis.


Subject(s)
Adherens Junctions , DNA Methylation , Humans , Adherens Junctions/genetics , Adherens Junctions/metabolism , Cadherins/genetics , Cadherins/metabolism , Signal Transduction , Chromatin/metabolism
2.
Cancer Gene Ther ; 31(2): 322-333, 2024 02.
Article in English | MEDLINE | ID: mdl-38057358

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is a subtype of CCA and has a high mortality rate and a relatively poor prognosis. However, studies focusing on increased cell motility and loss of epithelial integrity during iCCA progression remain relatively scarce. We collected seven fresh tumor samples from four patients to perform RNA sequencing (RNA-seq) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) to determine the transcriptome profile and chromatin accessibility of iCCA. The increased expression of cell cycle regulators, including PLK1 and its substrate MISP, was identified. Ninety-one iCCA patients were used to validate the clinical significance of PLK1 and MISP. The upregulation of PLK1 and MISP was determined in iCCA tissues. Increased expression of PLK1 and MISP was significantly correlated with tumor number, N stage, and lymphatic invasion in an iCCA cohort. Knockdown of PLK1 or MISP reduced trans-lymphatic endothelial migration and wound healing and affected focal adhesions in vitro. In cell‒cell junctions, MISP localized to adherens junctions and suppressed E-cadherin dimerization. PLK1 disrupted adherens junctions in a myosin-dependent manner. Furthermore, PLK1 and MISP promoted cell proliferation in vitro and tumorigenesis in vivo. In iCCA, PLK1 and MISP promote aggressiveness by increasing lymphatic invasion, tumor growth, and motility through the repression of E-cadherin adherens junctions.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Adherens Junctions/genetics , Adherens Junctions/metabolism , Adherens Junctions/pathology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Cadherins/genetics , Cadherins/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism
3.
J Cell Biol ; 222(12)2023 12 04.
Article in English | MEDLINE | ID: mdl-37796194

ABSTRACT

Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here, we employ a 3D model of a human ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from the loss of a transcription-independent, Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover that a Notch1-FAM83H interaction underlies control of epithelial adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.


Subject(s)
Actins , Adherens Junctions , Cell Adhesion , Receptor, Notch1 , Humans , Adherens Junctions/genetics , Cell Proliferation , Epithelial Cells , Proteins , Receptor, Notch1/genetics , Signal Transduction
4.
PLoS Genet ; 18(6): e1010292, 2022 06.
Article in English | MEDLINE | ID: mdl-35759519

ABSTRACT

Juvenile hormone (JH) acts as a gonadotrophic hormone stimulating insect vitellogenesis and oogenesis. Paracellular transport of yolk proteins through intercellular channels (patency) in the follicular epithelium is a developmentally regulated and evolutionarily conserved process during vitellogenesis. However, the mechanisms underlying patency opening are poorly understood. Using the migratory locust Locusta migratoria as a model system, we report here that JH-regulated remodeling of zonula adherens (ZA), the belt-like adherens junction maintaining physical linking between follicle cells controlled the opening of patency. JH triggered phosphorylation of Partitioning defective protein 3 (Par3) via a signaling cascade including G protein-coupled receptor (GPCR), small GTPase Cell division cycle 42 (Cdc42) and atypical Protein kinase C (aPKC). Par3 phosphorylation resulted in its disassociation from ß-Catenin, the cytoplasmic partner of ZA core component E-Cadherin. Release of Par3 from the ß-Catenin/E-Cadherin complex caused ZA disassembly at tricellular contacts, consequently leading to patency enlargement. This study provides new insight into how JH stimulates insect vitellogenesis and egg production via inducing the opening of paracellular route for vitellogenin transport crossing the follicular epithelium barrier.


Subject(s)
Adherens Junctions , Juvenile Hormones , Adherens Junctions/genetics , Adherens Junctions/metabolism , Cadherins/genetics , Epithelium/metabolism , Juvenile Hormones/genetics , Juvenile Hormones/metabolism , Vitellogenins/genetics , beta Catenin
5.
Arch Oral Biol ; 136: 105384, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35218988

ABSTRACT

OBJECTIVES: To study the association between the seven cell-cell adherens junctions (AJs) -related genes (CDH1, CTNND1, CTNNA1, ESRP1, ESRP2, PLEKHA5, and PLEKHA7) and non-syndromic cleft lip with or without cleft palate (NSCL/P) in the Asian population. DESIGN: The study included 895 NSCL/P case-parent trios of Asian ethnicity drawn from an international consortium established for a genome-wide association study. We performed association analysis by applying the genotypic transmission disequilibrium test for each of the 144 single nucleotide polymorphisms (SNPs), the versatile gene-based association study for the combined effects of all SNPs, all in or around the seven target genes, and the versatile pathway-based approach for the combined effects of these seven target genes, consecutively. RESULTS: In our analysis, we found neither a significant SNP-based nor gene-based association with NSCL/P for any of the 144 relevant SNPs or the seven target genes. However, novel evidence of a significant association (P = 6.00 × 10-6) with NSCL/P was observed in the combined effects of these seven target genes in the versatile pathway-based analysis. CONCLUSIONS: Our results were consistent with the findings of previously published human sequencing studies and reinforced the role of these cell-cell AJs-related genes in the pathogenesis of NSCL/P that may be replicated using data from other genome-wide association studies. This pathway approach better reflects our current understanding of the biological mechanisms underlying the aetiology of NSCL/P.


Subject(s)
Cleft Lip , Cleft Palate , Adherens Junctions/genetics , Case-Control Studies , Cleft Lip/genetics , Cleft Palate/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Parents , Polymorphism, Single Nucleotide
6.
Mol Biol Cell ; 33(2): ar14, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34851720

ABSTRACT

Distant organ metastasis is linked to poor prognosis during cancer progression. The expression level of the focal adhesion adapter protein paxillin varies among different human cancers, but its role in tumor progression is unclear. Herein we utilize a newly generated PyMT mammary tumor mouse model with conditional paxillin ablation in breast tumor epithelial cells, combined with in vitro three-dimensional (3D) tumor organoids invasion analysis and 2D calcium switch assays, to assess the roles for paxillin in breast tumor cell invasion. Paxillin had little effect on primary tumor initiation and growth but is critical for the formation of distant lung metastasis. In paxillin-depleted 3D tumor organoids, collective cell invasion was substantially perturbed. The 2D cell culture revealed paxillin-dependent stabilization of adherens junctions (AJ). Mechanistically, paxillin is required for AJ assembly through facilitating E-cadherin endocytosis and recycling and HDAC6-mediated microtubule acetylation. Furthermore, Rho GTPase activity analysis and rescue experiments with a RhoA activator or Rac1 inhibitor suggest paxillin is potentially regulating the E-cadherin-dependent junction integrity and contractility through control of the balance of RhoA and Rac1 activities. Together, these data highlight new roles for paxillin in the regulation of cell-cell adhesion and collective tumor cell migration to promote the formation of distance organ metastases.


Subject(s)
Adherens Junctions/physiology , Neoplasm Invasiveness/genetics , Paxillin/metabolism , Adherens Junctions/genetics , Animals , Breast/metabolism , Breast Neoplasms/metabolism , Cadherins/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Epithelial Cells/metabolism , Female , Focal Adhesions/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Paxillin/genetics , rho GTP-Binding Proteins/metabolism
7.
Shock ; 57(2): 309-315, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34907119

ABSTRACT

ABSTRACT: ERG (ETS-related gene) is a member of the ETS (Erythroblast-transformation specific) family of transcription factors abundantly present in vascular endothelial cells. Recent studies demonstrate that ERG has important roles in blood vessel stability and angiogenesis. However, it is unclear how ERG is potentially involved in microvascular barrier functions and permeability. A wide variety of diseases and clinical conditions including trauma-hemorrhagic shock and burn injury are associated with microvascular dysfunctions, which causes excessive microvascular permeability, tissue edema and eventually, multiple organ dysfunction and death. The main purpose of this study was to determine the specific role of ERG in regulating microvascular permeability in human lung microvascular endothelial cells (HLMEC) and to evaluate if exogenous ERG will protect the barrier. The HLMECs were grown on Transwell inserts as monolayers and were transfected with ERG CRISPR/cas9 knockdown plasmid, ERG CRISPR activation plasmid, recombinant ERG protein or their respective controls. Recombinant vascular endothelial growth factor (VEGF) was used as an inducer of permeability for evaluating the effect of ERG activation on permeability. Changes in barrier integrity and permeability were studied using monolayer permeability assay and immunofluorescence of adherens junction proteins (VE-cadherin and ß-catenin) respectively. CRISPR/cas9-based ERG knockdown as well as VEGF treatment induced monolayer hyperpermeability, VE-cadherin, and ß-catenin junctional relocation and cytoskeletal F-actin stress fiber formation. CRISPR based ERG activation and recombinant ERG transfection attenuated VEGF-induced monolayer hyperpermeability. ERG activation preserved the adherens junctions and cytoskeleton. These results demonstrate that ERG is a potent regulator of barrier integrity and permeability in human lung microvascular endothelial cells and endogenously or exogenously enhancing ERG provides protection against barrier dysfunction and hyperpermeability.


Subject(s)
Adherens Junctions/genetics , Capillary Permeability/genetics , Endothelial Cells , Endothelium, Vascular/cytology , Microvessels , Transcriptional Activation , Cells, Cultured , Humans , Transcriptional Regulator ERG/genetics
8.
Nat Commun ; 12(1): 6974, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34848713

ABSTRACT

The phenomenon of tissue fluidity-cells' ability to rearrange relative to each other in confluent tissues-has been linked to several morphogenetic processes and diseases, yet few molecular regulators of tissue fluidity are known. Ommatidial rotation (OR), directed by planar cell polarity signaling, occurs during Drosophila eye morphogenesis and shares many features with polarized cellular migration in vertebrates. We utilize in vivo live imaging analysis tools to quantify dynamic cellular morphologies during OR, revealing that OR is driven autonomously by ommatidial cell clusters rotating in successive pulses within a permissive substrate. Through analysis of a rotation-specific nemo mutant, we demonstrate that precise regulation of junctional E-cadherin levels is critical for modulating the mechanical properties of the tissue to allow rotation to progress. Our study defines Nemo as a molecular tool to induce a transition from solid-like tissues to more viscoelastic tissues broadening our molecular understanding of tissue fluidity.


Subject(s)
Adherens Junctions , Cell Polarity , Extracellular Fluid , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Cadherins , Cell Polarity/genetics , Cell Polarity/physiology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Ectoderm , Eye/cytology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis , Wings, Animal/cytology
9.
Development ; 148(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34846063

ABSTRACT

mRNA localization is an evolutionarily widespread phenomenon that can facilitate subcellular protein targeting. Extensive work has focused on mRNA targeting through 'zip-codes' within untranslated regions (UTRs), whereas much less is known about translation-dependent cues. Here, we examine mRNA localization in Caenorhabditis elegans embryonic epithelia. From an smFISH-based survey, we identified mRNAs associated with the cell membrane or cortex, and with apical junctions in a stage- and cell type-specific manner. Mutational analyses for one of these transcripts, dlg-1/discs large, revealed that it relied on a translation-dependent process and did not require its 5' or 3' UTRs. We suggest a model in which dlg-1 transcripts are co-translationally localized with the nascent protein: first the translating complex goes to the cell membrane using sequences located at the C-terminal/3' end, and then apically using N-terminal/5' sequences. These studies identify a translation-based process for mRNA localization within developing epithelia and determine the necessary cis-acting sequences for dlg-1 mRNA targeting.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Cell Membrane/genetics , Embryonic Development/genetics , Guanylate Kinases/genetics , Protein Biosynthesis/genetics , Adherens Junctions/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Cell Polarity/genetics , Embryo, Nonmammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Intercellular Junctions/genetics , Membrane Proteins/genetics , Protein Transport/genetics , RNA, Messenger/genetics , Sequence Homology, Amino Acid
10.
Mol Biol Cell ; 32(19): 1824-1837, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34260281

ABSTRACT

Desmosomes (DSMs), together with adherens junctions (AJs) and tight junctions (TJs), constitute the apical cell junctional complex (AJC). While the importance of the apical and basolateral polarity machinery in the organization of AJs and TJs is well established, how DSMs are positioned within the AJC is not understood. Here we use highly polarized DLD1 cells as a model to address how DSMs integrate into the AJC. We found that knockout (KO) of the desmosomal ARM protein Pkp3, but not other major DSM proteins, uncouples DSMs from the AJC without blocking DSM assembly. DLD1 cells also exhibit a prominent extraDSM pool of Pkp3, concentrated in tricellular (tC) contacts. Probing distinct apicobasal polarity pathways revealed that neither the DSM's association with AJC nor the extraDSM pool of Pkp3 are abolished in cells with defects in Scrib module proteins responsible for basolateral membrane development. However, a loss of the apical polarity protein, Par3, completely eliminates the extraDSM pool of Pkp3 and disrupts AJC localization of desmosomes, dispersing these junctions along the entire length of cell-cell contacts. Our data are consistent with a model whereby Par3 facilitates DSM assembly within the AJC, controlling the availability of an assembly competent pool of Pkp3 stored in tC contacts.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Cell Cycle Proteins/metabolism , Desmosomes/metabolism , Plakophilins/metabolism , Tight Junctions/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adherens Junctions/genetics , Animals , Caco-2 Cells , Cell Communication/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Polarity/genetics , Cells, Cultured , Desmosomes/genetics , Dogs , Epithelial Cells/metabolism , Gene Knockout Techniques , Humans , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence/methods , Plakophilins/genetics , Tight Junctions/genetics
11.
J Biol Chem ; 297(1): 100848, 2021 07.
Article in English | MEDLINE | ID: mdl-34058200

ABSTRACT

Within the intestinal epithelium, regulation of intracellular protein and vesicular trafficking is of utmost importance for barrier maintenance, immune responses, and tissue polarity. RAB11A is a small GTPase that mediates the anterograde transport of protein cargos to the plasma membrane. Loss of RAB11A-dependent trafficking in mature intestinal epithelial cells results in increased epithelial proliferation and nuclear accumulation of Yes-associated protein (YAP), a key Hippo-signaling transducer that senses cell-cell contacts and regulates tissue growth. However, it is unclear how RAB11A regulates YAP intracellular localizations. In this report, we examined the relationship of RAB11A to epithelial junctional complexes, YAP, and the associated consequences on colonic epithelial tissue repair. We found that RAB11A controls the biochemical associations of YAP with multiple components of adherens and tight junctions, including α-catenin, ß-catenin, and Merlin, a tumor suppressor. In the absence of RAB11A and Merlin, we observed enhanced YAP-ß-catenin complex formation and nuclear translocation. Upon chemical injury to the intestine, mice deficient in RAB11A were found to have reduced epithelial integrity, decreased YAP localization to adherens and tight junctions, and increased nuclear YAP accumulation in the colon epithelium. Thus, RAB11A-regulated trafficking regulates the Hippo-YAP signaling pathway for rapid reparative response after tissue injury.


Subject(s)
Cell Cycle Proteins/genetics , Colitis/genetics , Neurofibromin 2/genetics , Transcription Factors/genetics , beta Catenin/genetics , rab GTP-Binding Proteins/genetics , Adherens Junctions/genetics , Animals , Caco-2 Cells , Cell Proliferation/genetics , Colitis/chemically induced , Colitis/pathology , Colon/growth & development , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelium/growth & development , Epithelium/pathology , Humans , Mice , Tight Junctions/genetics , alpha Catenin/genetics
12.
Nat Commun ; 12(1): 2610, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972531

ABSTRACT

Angiogenic sprouting relies on collective migration and coordinated rearrangements of endothelial leader and follower cells. VE-cadherin-based adherens junctions have emerged as key cell-cell contacts that transmit forces between cells and trigger signals during collective cell migration in angiogenesis. However, the underlying molecular mechanisms that govern these processes and their functional importance for vascular development still remain unknown. We previously showed that the F-BAR protein PACSIN2 is recruited to tensile asymmetric adherens junctions between leader and follower cells. Here we report that PACSIN2 mediates the formation of endothelial sprouts during angiogenesis by coordinating collective migration. We show that PACSIN2 recruits the trafficking regulators EHD4 and MICAL-L1 to the rear end of asymmetric adherens junctions to form a recycling endosome-like tubular structure. The junctional PACSIN2/EHD4/MICAL-L1 complex controls local VE-cadherin trafficking and thereby coordinates polarized endothelial migration and angiogenesis. Our findings reveal a molecular event at force-dependent asymmetric adherens junctions that occurs during the tug-of-war between endothelial leader and follower cells, and allows for junction-based guidance during collective migration in angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , DNA-Binding Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Microfilament Proteins/metabolism , Mixed Function Oxygenases/metabolism , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Catenins/metabolism , Cell Movement/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/genetics , Signal Transduction/genetics , Spheroids, Cellular/metabolism
13.
Dev Comp Immunol ; 122: 104111, 2021 09.
Article in English | MEDLINE | ID: mdl-33933535

ABSTRACT

Molecular underpinning of mycobacteria-induced CNS-pathology is not well understood. In the present study, zebrafish were infected with Mycobacterium fortuitum and the prognosis of CNS-pathogenesis studied. We observed M. fortuitum triggers extensive brain-pathology. Evans blue extravasation demonstrated compromised blood-brain barrier (BBB) integrity. Further, decreased expression in tight-junction (TJ) and adherens junction complex (AJC) genes were noted in infected brain. Wnt-signaling has emerged as a major player in host-mycobacterial immunity but its involvement/role in brain-infection is not well studied. Sustained expression of wnt2, wnt3a, fzd5, lrp5/6 and ß-catenin, with concordant decline in degradation complex components axin, gsk3ß and ß-catenin regulator capn2a were observed. The surge in ifng1 and tnfa expression preceding il10 and il4 suggested cytokine-interplay critical in M. fortuitum-induced brain-pathology. Therefore, we suggest adult zebrafish as a viable model for studying CNS-pathology and using the same, conclude that M. fortuitum infection is associated with repressed TJ-AJC gene expression and compromised BBB permeability. Our results implicate Wnt/ß-catenin pathway in M. fortuitum-induced CNS-pathology wherein Th1-type signals facilitate bacterial clearance and Th2-type signals prevent the disease sequel.


Subject(s)
Blood-Brain Barrier/microbiology , Brain/pathology , Cytokines/metabolism , Fish Diseases/immunology , Mycobacterium fortuitum/immunology , Wnt Signaling Pathway/immunology , Zebrafish/immunology , Adherens Junctions/genetics , Animals , Axin Protein/metabolism , Blood-Brain Barrier/metabolism , Brain/microbiology , Calpain/metabolism , Fish Diseases/microbiology , Glycogen Synthase Kinase 3 beta/metabolism , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-4/immunology , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium Infections, Nontuberculous/veterinary , Mycobacterium fortuitum/pathogenicity , Receptors, Cell Surface/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Tight Junctions/genetics , Tumor Necrosis Factor-alpha/immunology , Wnt Proteins/metabolism , Wnt3A Protein/metabolism , Zebrafish/microbiology , Zebrafish Proteins/metabolism , beta Catenin/metabolism
14.
Elife ; 102021 04 30.
Article in English | MEDLINE | ID: mdl-33929320

ABSTRACT

Ras-responsive element-binding protein 1 (Rreb1) is a zinc-finger transcription factor acting downstream of RAS signaling. Rreb1 has been implicated in cancer and Noonan-like RASopathies. However, little is known about its role in mammalian non-disease states. Here, we show that Rreb1 is essential for mouse embryonic development. Loss of Rreb1 led to a reduction in the expression of vasculogenic factors, cardiovascular defects, and embryonic lethality. During gastrulation, the absence of Rreb1 also resulted in the upregulation of cytoskeleton-associated genes, a change in the organization of F-ACTIN and adherens junctions within the pluripotent epiblast, and perturbed epithelial architecture. Moreover, Rreb1 mutant cells ectopically exited the epiblast epithelium through the underlying basement membrane, paralleling cell behaviors observed during metastasis. Thus, disentangling the function of Rreb1 in development should shed light on its role in cancer and other diseases involving loss of epithelial integrity.


Subject(s)
Blood Vessels/embryology , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Mice/embryology , Neovascularization, Physiologic , Transcription Factors/metabolism , Actins/genetics , Actins/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Blood Vessels/metabolism , DNA-Binding Proteins/genetics , Embryonic Development , Mice/genetics , Mice/metabolism , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/genetics
15.
Elife ; 102021 04 28.
Article in English | MEDLINE | ID: mdl-33908348

ABSTRACT

Background: Hypoxia and consequent production of vascular endothelial growth factor A (VEGFA) promote blood vessel leakiness and edema in ocular diseases. Anti-VEGFA therapeutics may aggravate hypoxia; therefore, therapy development is needed. Methods: Oxygen-induced retinopathy was used as a model to test the role of nitric oxide (NO) in pathological neovascularization and vessel permeability. Suppression of NO formation was achieved chemically using L-NMMA, or genetically, in endothelial NO synthase serine to alanine (S1176A) mutant mice. Results: Suppression of NO formation resulted in reduced retinal neoangiogenesis. Remaining vascular tufts exhibited reduced vascular leakage through stabilized endothelial adherens junctions, manifested as reduced phosphorylation of vascular endothelial (VE)-cadherin Y685 in a c-Src-dependent manner. Treatment with a single dose of L-NMMA in established retinopathy restored the vascular barrier and prevented leakage. Conclusions: We conclude that NO destabilizes adheren junctions, resulting in vascular hyperpermeability, by converging with the VEGFA/VEGFR2/c-Src/VE-cadherin pathway. Funding: This study was supported by the Swedish Cancer foundation (19 0119 Pj ), the Swedish Research Council (2020-01349), the Knut and Alice Wallenberg foundation (KAW 2020.0057) and a Fondation Leducq Transatlantic Network of Excellence Grant in Neurovascular Disease (17 CVD 03). KAW also supported LCW with a Wallenberg Scholar grant (2015.0275). WCS was supported by Grants R35 HL139945, P01 HL1070205, AHA MERIT Award. DV was supported by grants from the Deutsche Forschungsgemeinschaft, SFB1450, B03, and CRU342, P2.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , CSK Tyrosine-Protein Kinase/metabolism , Cadherins/chemistry , Cadherins/metabolism , Nitric Oxide Synthase Type III/metabolism , Retinal Diseases/enzymology , Tyrosine/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Amino Acid Motifs , Animals , Antigens, CD/genetics , CSK Tyrosine-Protein Kinase/genetics , Cadherins/genetics , Capillary Permeability , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Retinal Diseases/genetics , Retinal Diseases/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Exp Cell Res ; 402(1): 112525, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33662366

ABSTRACT

Cells dividing in the plane of epithelial tissues proceed by polarized constriction of the actomyosin contractile ring, leading to asymmetric ingression of the plasma mem brane. Asymmetric cytokinesis results in the apical positioning of the actomyosin contractile ring and ultimately of the midbody. Studies have indicated that the contractile ring is associated with adherens junctions, whose role is to maintain epithelial tissue cohesion. However, it is yet unknown when the contractile ring becomes associated with adherens junctions in epithelial cells. Here, we examined contractile ring formation and activation in the epithelium of Xenopus embryos and explored the implication of adherens junctions in the contractile ring formation. We show that accumulation of proteins involved in contractile ring formation and activation is polarized, starting at apical cell-cell contacts at the presumptive division site and spreading within seconds towards the cell basal side. We also show that adherens junctions are involved in the kinetics of contractile ring formation. Our study reveals that the link between the adherens junctions and the contractile ring is established from the onset of cytokinesis.


Subject(s)
Adherens Junctions/genetics , Embryonic Development/genetics , Epithelial Cells/metabolism , Xenopus laevis/genetics , Actin Cytoskeleton/genetics , Actomyosin/genetics , Animals , Cell Division/genetics , Cell Polarity/genetics , Contractile Proteins/genetics , Cytokinesis/genetics , Embryo, Nonmammalian , Epithelial Cells/cytology , Xenopus laevis/growth & development
18.
Int J Mol Med ; 47(2): 621-632, 2021 02.
Article in English | MEDLINE | ID: mdl-33416101

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is highly expressed in mammalian intestines, and is essential for maintaining the homeostasis of the intestinal epithelium. EpCAM protein is localized at tight junctions and the basolateral membrane of the intestinal epithelium, where it interacts with many cell adhesion molecules. To explore the molecular functions of EpCAM in regulating adherens junctions in the intestinal epithelium, EpCAM knockout embryos and newborn pups were analyzed. Hematoxylin and eosin staining was used to assess the histology of the duodenum, jejunum, ileum and colon from wild-type and EpCAM­/­ mice at E18.5, P0 and P3. The expression and localization of adherens junction­associated genes and genes that encode the proteins that participate in the assembly of adherens junctions were measured at the mRNA and protein levels using qPCR, western blot analysis and immunofluorescence staining. The results showed that although there was no significant damage to the intestines of EpCAM­/­ mice at E18.5 and P0, they were significantly damaged at P3 in mutant mice. The expression of adherens junction­associated genes in EpCAM mutant mice was normal at the mRNA level from E18.5 to P3, but their protein levels were gradually reduced and mislocalized from E18.5 to P3. The expression of nectin 1, which can regulate the assembly and adhesion activity of E­cadherin, was also gradually reduced at both the mRNA and protein levels in the intestinal epithelium of EpCAM mutant mice from E18.5 to P3. In summary, the loss of EpCAM may cause the reduction and mislocalization of proteins that compose adherens junctions partly via the downregulation of nectin 1 in the intestines.


Subject(s)
Adherens Junctions/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression Regulation , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Adherens Junctions/genetics , Animals , Epithelial Cell Adhesion Molecule/genetics , Mice , Mice, Knockout
19.
RNA Biol ; 18(3): 391-403, 2021 03.
Article in English | MEDLINE | ID: mdl-32876513

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a hallmark of aggressive, mesenchymal-like high-grade serous ovarian carcinoma (HGSOC). The SRC kinase is a key driver of cancer-associated EMT promoting adherens junction (AJ) disassembly by phosphorylation-driven internalization and degradation of AJ proteins. Here, we show that the IGF2 mRNA-binding protein 1 (IGF2BP1) is up-regulated in mesenchymal-like HGSOC and promotes SRC activation by a previously unknown protein-ligand-induced, but RNA-independent mechanism. IGF2BP1-driven invasive growth of ovarian cancer cells essentially relies on the SRC-dependent disassembly of AJs. Concomitantly, IGF2BP1 enhances ERK2 expression in an RNA-binding dependent manner. Together this reveals a post-transcriptional mechanism of interconnected stimulation of SRC/ERK signalling in ovarian cancer cells. The IGF2BP1-SRC/ERK2 axis is targetable by the SRC-inhibitor saracatinib and MEK-inhibitor selumetinib. However, due to IGF2BP1-directed stimulation, only combinatorial treatment effectively overcomes the IGF2BP1-promoted invasive growth in 3D culture conditions as well as intraperitoneal mouse models. In conclusion, we reveal an unexpected role of IGF2BP1 in enhancing SRC/MAPK-driven invasive growth of ovarian cancer cells. This provides a rationale for the therapeutic benefit of combinatorial SRC/MEK inhibition in mesenchymal-like HGSOC.


Subject(s)
Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinases/metabolism , Ovarian Neoplasms/etiology , Ovarian Neoplasms/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , src-Family Kinases/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , src Homology Domains , src-Family Kinases/antagonists & inhibitors
20.
Clin Transl Oncol ; 23(3): 591-600, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32720055

ABSTRACT

PURPOSE: Melanoma is a malignant skin tumor, and its incidence is rising. To explore the specific differences in benign and malignant melanoma at the genetic level, we performed a series of bioinformatics analyses, including differential gene analysis, co-expression analysis, enrichment analysis, and regulatory prediction. METHODS: The microarray data of benign and malignant melanocytes were downloaded from GEO, and 1917 differential genes were obtained by differential analysis (p < 0.05). Weighted gene co-expression network analysis obtained three functional barrier modules. The essential genes of each module are SMARTA4, HECA, and C1R. RESULTS: The results of the enrichment analysis showed that the dysfunctional module gene was mainly associated with RNA splicing and Adherens junction. Through the pivotal analysis of ncRNA, it was found that miR-448, miR-152-3p, and miR-302b-3p essentially regulate three modules, which we consider to be critical regulators. In the pivot analysis of TF, more control modules include ARID3A, E2F1, E2F3, and E2F8. CONCLUSIONS: We believe that the regulator (miR-448, miR-152-3p, miR-302b-3p) regulates the expression of the core gene SMARCA4, which in turn affects the signal transduction of the Adherens junction. It eventually leads to the deterioration of benign skin spasms into melanoma.


Subject(s)
Adherens Junctions/genetics , Cell Transformation, Neoplastic/genetics , DNA Helicases/genetics , Gene Regulatory Networks/genetics , Melanocytes , Melanoma/genetics , Nuclear Proteins/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics , Cell Transformation, Neoplastic/pathology , Computational Biology , DNA-Binding Proteins , Databases, Genetic , E2F1 Transcription Factor , E2F3 Transcription Factor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Melanocytes/pathology , Melanoma/pathology , MicroRNAs , Neoplasm Proteins/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Untranslated , Repressor Proteins , Signal Transduction , Skin Neoplasms/pathology , Tissue Array Analysis/methods , Melanoma, Cutaneous Malignant
SELECTION OF CITATIONS
SEARCH DETAIL
...