Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(12): 3254-3266, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38640438

ABSTRACT

ABSTRACT: von Willebrand factor (VWF) undergoes complex posttranslational modification within endothelial cells (ECs) before secretion. This includes significant N- and O-linked glycosylation. Previous studies have demonstrated that changes in N-linked glycan structures significantly influence VWF biosynthesis. In contrast, although abnormalities in VWF O-linked glycans (OLGs) have been associated with enhanced VWF clearance, their effect on VWF biosynthesis remains poorly explored. Herein, we report a novel role for OLG determinants in regulating VWF biosynthesis and trafficking within ECs. We demonstrate that alterations in OLGs (notably reduced terminal sialylation) lead to activation of the A1 domain of VWF within EC. In the presence of altered OLG, VWF multimerization is reduced and Weibel-Palade body (WPB) formation significantly impaired. Consistently, the amount of VWF secreted from WPB after EC activation was significantly reduced in the context of O-glycosylation inhibition. Finally, altered OLG on VWF not only reduced the amount of VWF secreted after EC activation but also affected its hemostatic efficacy. Notably, VWF secreted after WPB exocytosis consisted predominantly of low molecular weight multimers, and the length of tethered VWF string formation on the surface of activated ECs was significantly reduced. In conclusion, our data therefore support the hypothesis that alterations in O-glycosylation pathways directly affect VWF trafficking within human EC. These findings are interesting given that previous studies have reported altered OLG on plasma VWF (notably increased T-antigen expression) in patients with von Willebrand disease.


Subject(s)
Polysaccharides , Protein Transport , Weibel-Palade Bodies , von Willebrand Factor , von Willebrand Factor/metabolism , Weibel-Palade Bodies/metabolism , Humans , Polysaccharides/metabolism , Glycosylation , Endothelial Cells/metabolism , Protein Processing, Post-Translational , Protein Multimerization
2.
Nat Commun ; 14(1): 7086, 2023 11 04.
Article in English | MEDLINE | ID: mdl-37925537

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomere maintenance mechanism activated in ~10-15% of cancers, characterized by telomeric damage. Telomeric damage-induced long non-coding RNAs (dilncRNAs) are transcribed at dysfunctional telomeres and contribute to telomeric DNA damage response (DDR) activation and repair. Here we observed that telomeric dilncRNAs are preferentially elevated in ALT cells. Inhibition of C-rich (teloC) dilncRNAs with antisense oligonucleotides leads to DNA replication stress responses, increased genomic instability, and apoptosis induction selectively in ALT cells. Cell death is dependent on DNA replication and is increased by DNA replication stress. Mechanistically, teloC dilncRNA inhibition reduces RAD51 and 53BP1 recruitment to telomeres, boosts the engagement of BIR machinery, and increases C-circles and telomeric sister chromatid exchanges, without increasing telomeric non-S phase synthesis. These results indicate that teloC dilncRNA is necessary for a coordinated recruitment of DDR factors to ALT telomeres and it is essential for ALT cancer cells survival.


Subject(s)
Telomerase , Telomere Homeostasis , Telomere Homeostasis/genetics , DNA Replication , RNA , Cell Survival/genetics , Telomere/genetics , Telomere/metabolism , Telomerase/genetics , Telomerase/metabolism
3.
ACS Omega ; 8(28): 25279-25287, 2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37483254

ABSTRACT

Insulin undergoes agglomeration with (subtle) changes in its biochemical environment, including acidity, application of heat, ionic imbalance, and exposure to hydrophobic surfaces. The therapeutic impact of such unwarranted insulin agglomeration is unclear and needs further evaluation. A systematic investigation was conducted on recombinant human insulin-with or without labeling with fluorescein isothiocyanate-while preparing insulin suspensions (0.125, 0.25, and 0.5 mg/mL) at pH 3. The suspensions were incubated (37 °C) and analyzed at different time points (t = 2, 4, 24, 48, and 72 h). Transmission electron microscopy and nanoparticle tracking analysis identified colloidally stable (zeta potential 15 ± 5 mV) spherical agglomerates of unlabeled insulin (100-500 nm). Circular dichroism established the preservation of insulin's secondary structure rich in α-helices despite exposure to an acidic environment (pH 3) for 72 h. Furthermore, fluorescence lifetime imaging microscopy illustrated an acidic core inside these spherical agglomerates, while the acidity gradually lessened toward the periphery. Some of these smaller agglomerates fused to form larger chunks with discrete zones of acidity. The data indicated a primary nucleation-driven mechanism of acid-induced insulin agglomeration under physiologically relevant conditions.

4.
RSC Adv ; 13(22): 14963-14973, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37200702

ABSTRACT

Three bis(anilino)-substituted NIR-AZA fluorophores have been designed, synthesized and tested to bridge the availability gap of molecular fluorophores for live-cell microscopy imaging in the 800-850 nm spectral range. The concise synthetic route allows for the later stage introduction of three tailored peripheral substituents which guides the sub-cellular localization and imaging. Live-cell fluorescence imaging of lipid droplets, plasma membrane and cytosolic vacuoles was successfully achieved. Photophysical and internal charge transfer (ICT) properties of each fluorophore were examined through solvent studies and analyte responses.

5.
Molecules ; 28(3)2023 Feb 02.
Article in English | MEDLINE | ID: mdl-36771082

ABSTRACT

BF2-azadipyrromethenes are highly versatile fluorophores used for cellular and in vivo imaging in the near-infrared and far-red regions of the spectrum. As of yet, their use in conjunction with super-resolution imaging methodologies has not been explored. In this report, a series of structurally related BF2-azadipyrromethenes has been examined for their suitability for use with stimulated emission depletion (STED) nanoscopy. The potential for STED imaging was initially evaluated using aqueous solutions of fluorophores as an effective predictor of fluorophore suitability. For live cell STED imaging in both 2D and 3D, several far-red emitting BF2-azadipyrromethenes were successfully employed. Image resolution below the diffraction limit of a confocal microscope was demonstrated through measurement of distinct intracellular features including the nuclear membrane, nuclear lamina invaginations, the endoplasmic reticulum, and vacuoles. As the STED ability of BF2-azadipyrromethene fluorophores has now been established, their use with this super-resolution method may be expected to increase in the future.


Subject(s)
Fluorescent Dyes , Vacuoles , Microscopy, Fluorescence/methods
6.
Chem Commun (Camb) ; 59(14): 1951-1954, 2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36722871

ABSTRACT

Sequential azide/diyne cycloadditions proved highly effective for the macrocyclization of a bis-azido aza-dipyrrin. Macrocyclic aza-dipyrrin could be produced in 30 min at rt in water with changes in fluorescence intensity and lifetimes measurable upon reaction. Live cell microscopy showed that aza-dipyrrins were suitable for confocal and STED super-resolution imaging and a bioorthogonal response to macrocyclization could be detected in cellular compartments. These results will encourage a broader examination of the sensing and imaging uses of aza-dipyrrins.


Subject(s)
Diynes , Microscopy, Fluorescence
7.
RSC Adv ; 12(55): 35655-35665, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36545082

ABSTRACT

A bio-responsive nanoparticle was formed by the directed self-assembly (DSA) of a hydrophobic NIR-fluorophore with poloxamer P188. Fluorophore emission was switched off when part of the nanoparticle, however upon stimulus induced nanoparticle dis-assembly the emission switched on. The emission quenching was shown to be due to fluorophore hydration and aggregation within the nanoparticle and the turn on response attributable to nanoparticle disassembly with embedding of the fluorophore within lipophilic environments. This was exploited for temporal and spatial live cell imaging with a measurable fluorescence response seen upon intracellular delivery of the fluorophore. The first dynamic response, seen within minutes, was from lipid droplets with other lipophilic regions such as the endoplasmic reticulum, nuclear membranes and secretory vacuoles imageable after hours. The high degree of fluorophore photostability facilitated continuous imaging for extended periods and the off to on switching facilitated the real-time observation of lipid droplet biogenesis as they emerged from the endoplasmic reticulum. With an in-depth understanding of the principles involved, further assembly controlling functional responses could be anticipated.

9.
Nat Commun ; 12(1): 3937, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168151

ABSTRACT

Although human nucleoporin Tpr is frequently deregulated in cancer, its roles are poorly understood. Here we show that Tpr depletion generates transcription-dependent replication stress, DNA breaks, and genomic instability. DNA fiber assays and electron microscopy visualization of replication intermediates show that Tpr deficient cells exhibit slow and asymmetric replication forks under replication stress. Tpr deficiency evokes enhanced levels of DNA-RNA hybrids. Additionally, complementary proteomic strategies identify a network of Tpr-interacting proteins mediating RNA processing, such as MATR3 and SUGP2, and functional experiments confirm that their depletion trigger cellular phenotypes shared with Tpr deficiency. Mechanistic studies reveal the interplay of Tpr with GANP, a component of the TREX-2 complex. The Tpr-GANP interaction is supported by their shared protein level alterations in a cohort of ovarian carcinomas. Our results reveal links between nucleoporins, DNA transcription and replication, and the existence of a network physically connecting replication forks with transcription, splicing, and mRNA export machinery.


Subject(s)
DNA Replication , Nuclear Pore Complex Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Acetyltransferases/genetics , Acetyltransferases/metabolism , Cell Survival , DNA Damage , Genomic Instability , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/genetics , Nuclear Pore Complex Proteins/genetics , Protein Interaction Maps , Proto-Oncogene Proteins/genetics , RNA Transport
10.
Sci Signal ; 14(676)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785611

ABSTRACT

Innate immune responses to Gram-negative bacteria depend on the recognition of lipopolysaccharide (LPS) by a receptor complex that includes CD14 and TLR4. In dendritic cells (DCs), CD14 enhances the activation not only of TLR4 but also that of the NFAT family of transcription factors, which suppresses cell survival and promotes the production of inflammatory mediators. NFAT activation requires Ca2+ mobilization. In DCs, Ca2+ mobilization in response to LPS depends on phospholipase C γ2 (PLCγ2), which produces inositol 1,4,5-trisphosphate (IP3). Here, we showed that the IP3 receptor 3 (IP3R3) and ITPKB, a kinase that converts IP3 to inositol 1,3,4,5-tetrakisphosphate (IP4), were both necessary for Ca2+ mobilization and NFAT activation in mouse and human DCs. A pool of IP3R3 was located on the plasma membrane of DCs, where it colocalized with CD14 and ITPKB. Upon LPS binding to CD14, ITPKB was required for Ca2+ mobilization through plasma membrane-localized IP3R3 and for NFAT nuclear translocation. Pharmacological inhibition of ITPKB in mice reduced both LPS-induced tissue swelling and the severity of inflammatory arthritis to a similar extent as that induced by the inhibition of NFAT using nanoparticles that delivered an NFAT-inhibiting peptide specifically to phagocytic cells. Our results suggest that ITPKB may represent a promising target for anti-inflammatory therapies that aim to inhibit specific DC functions.


Subject(s)
Calcium/metabolism , Dendritic Cells , Phosphotransferases (Alcohol Group Acceptor) , Animals , Lipopolysaccharides , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics
11.
Front Cell Dev Biol ; 9: 637565, 2021.
Article in English | MEDLINE | ID: mdl-33718375

ABSTRACT

Snap29 is a conserved regulator of membrane fusion essential to complete autophagy and to support other cellular processes, including cell division. In humans, inactivating SNAP29 mutations causes CEDNIK syndrome, a rare multi-systemic disorder characterized by congenital neuro-cutaneous alterations. The fibroblasts of CEDNIK patients show alterations of the Golgi apparatus (GA). However, whether and how Snap29 acts at the GA is unclear. Here we investigate SNAP29 function at the GA and endoplasmic reticulum (ER). As part of the elongated structures in proximity to these membrane compartments, a pool of SNAP29 forms a complex with Syntaxin18, or with Syntaxin5, which we find is required to engage SEC22B-loaded vesicles. Consistent with this, in HeLa cells, in neuroepithelial stem cells, and in vivo, decreased SNAP29 activity alters GA architecture and reduces ER to GA trafficking. Our data reveal a new regulatory function of Snap29 in promoting secretory trafficking.

12.
Cell Death Differ ; 27(8): 2383-2401, 2020 08.
Article in English | MEDLINE | ID: mdl-32042098

ABSTRACT

Mitochondria change distribution across cells following a variety of pathophysiological stimuli. The mechanisms presiding over this redistribution are yet undefined. In a murine model overexpressing Drp1 specifically in skeletal muscle, we find marked mitochondria repositioning in muscle fibres and we demonstrate that Drp1 is involved in this process. Drp1 binds KLC1 and enhances microtubule-dependent transport of mitochondria. Drp1-KLC1 coupling triggers the displacement of KIF5B from kinesin-1 complex increasing its binding to microtubule tracks and mitochondrial transport. High levels of Drp1 exacerbate this mechanism leading to the repositioning of mitochondria closer to nuclei. The reduction of Drp1 levels decreases kinesin-1 activation and induces the partial recovery of mitochondrial distribution. Drp1 overexpression is also associated with higher cyclin-dependent kinase-1 (Cdk-1) activation that promotes the persistent phosphorylation of desmin at Ser-31 and its disassembling. Fission inhibition has a positive effect on desmin Ser-31 phosphorylation, regardless of Cdk-1 activation, suggesting that induction of both fission and Cdk-1 are required for desmin collapse. This altered desmin architecture impairs mechanotransduction and compromises mitochondrial network stability priming mitochondria transport through microtubule-dependent trafficking with a mechanism that involves the Drp1-dependent regulation of kinesin-1 complex.


Subject(s)
Desmin/metabolism , Dynamins/metabolism , Kinesins/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Animals , CDC2 Protein Kinase/metabolism , Enzyme Activation , Humans , Mice, Inbred C57BL , Microtubules/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Quinazolinones/metabolism , Succinate Dehydrogenase/metabolism
13.
Nat Cell Biol ; 21(10): 1286-1299, 2019 10.
Article in English | MEDLINE | ID: mdl-31570834

ABSTRACT

Damage-induced long non-coding RNAs (dilncRNA) synthesized at DNA double-strand breaks (DSBs) by RNA polymerase II are necessary for DNA-damage-response (DDR) focus formation. We demonstrate that induction of DSBs results in the assembly of functional promoters that include a complete RNA polymerase II preinitiation complex, MED1 and CDK9. Absence or inactivation of these factors causes a reduction in DDR foci both in vivo and in an in vitro system that reconstitutes DDR events on nucleosomes. We also show that dilncRNAs drive molecular crowding of DDR proteins, such as 53BP1, into foci that exhibit liquid-liquid phase-separation condensate properties. We propose that the assembly of DSB-induced transcriptional promoters drives RNA synthesis, which stimulates phase separation of DDR factors in the shape of foci.


Subject(s)
Cyclin-Dependent Kinase 9/genetics , DNA Repair , DNA/genetics , Mediator Complex Subunit 1/metabolism , Transcription, Genetic , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Mediator Complex Subunit 1/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
14.
Nat Mater ; 18(11): 1252-1263, 2019 11.
Article in English | MEDLINE | ID: mdl-31332337

ABSTRACT

During wound repair, branching morphogenesis and carcinoma dissemination, cellular rearrangements are fostered by a solid-to-liquid transition, known as unjamming. The biomolecular machinery behind unjamming and its pathophysiological relevance remain, however, unclear. Here, we study unjamming in a variety of normal and tumorigenic epithelial two-dimensional (2D) and 3D collectives. Biologically, the increased level of the small GTPase RAB5A sparks unjamming by promoting non-clathrin-dependent internalization of epidermal growth factor receptor that leads to hyperactivation of the kinase ERK1/2 and phosphorylation of the actin nucleator WAVE2. This cascade triggers collective motility effects with striking biophysical consequences. Specifically, unjamming in tumour spheroids is accompanied by persistent and coordinated rotations that progressively remodel the extracellular matrix, while simultaneously fluidizing cells at the periphery. This concurrent action results in collective invasion, supporting the concept that the endo-ERK1/2 pathway is a physicochemical switch to initiate collective invasion and dissemination of otherwise jammed carcinoma.


Subject(s)
Cell Differentiation , Cell Movement , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/metabolism , Humans , Kinetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , rab5 GTP-Binding Proteins/metabolism
15.
Autophagy ; 15(4): 631-651, 2019 04.
Article in English | MEDLINE | ID: mdl-30335591

ABSTRACT

Macroautophagy/autophagy, a defense mechanism against aberrant stresses, in neurons counteracts aggregate-prone misfolded protein toxicity. Autophagy induction might be beneficial in neurodegenerative diseases (NDs). The natural compound trehalose promotes autophagy via TFEB (transcription factor EB), ameliorating disease phenotype in multiple ND models, but its mechanism is still obscure. We demonstrated that trehalose regulates autophagy by inducing rapid and transient lysosomal enlargement and membrane permeabilization (LMP). This effect correlated with the calcium-dependent phosphatase PPP3/calcineurin activation, TFEB dephosphorylation and nuclear translocation. Trehalose upregulated genes for the TFEB target and regulator Ppargc1a, lysosomal hydrolases and membrane proteins (Ctsb, Gla, Lamp2a, Mcoln1, Tpp1) and several autophagy-related components (Becn1, Atg10, Atg12, Sqstm1/p62, Map1lc3b, Hspb8 and Bag3) mostly in a PPP3- and TFEB-dependent manner. TFEB silencing counteracted the trehalose pro-degradative activity on misfolded protein causative of motoneuron diseases. Similar effects were exerted by trehalase-resistant trehalose analogs, melibiose and lactulose. Thus, limited lysosomal damage might induce autophagy, perhaps as a compensatory mechanism, a process that is beneficial to counteract neurodegeneration. Abbreviations: ALS: amyotrophic lateral sclerosis; AR: androgen receptor; ATG: autophagy related; AV: autophagic vacuole; BAG3: BCL2-associated athanogene 3; BECN1: beclin 1, autophagy related; CASA: chaperone-assisted selective autophagy; CTSB: cathepsin b; DAPI: 4',6-diamidino-2-phenylindole; DMEM: Dulbecco's modified Eagle's medium; EGFP: enhanced green fluorescent protein; fALS, familial amyotrophic lateral sclerosis; FRA: filter retardation assay; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GLA: galactosidase, alpha; HD: Huntington disease; hIPSCs: human induced pluripotent stem cells; HSPA8: heat shock protein A8; HSPB8: heat shock protein B8; IF: immunofluorescence analysis; LAMP1: lysosomal-associated membrane protein 1; LAMP2A: lysosomal-associated membrane protein 2A; LGALS3: lectin, galactose binding, soluble 3; LLOMe: L-leucyl-L-leucine methyl ester; LMP: lysosomal membrane permeabilization; Lys: lysosomes; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MCOLN1: mucolipin 1; mRNA: messenger RNA; MTOR: mechanistic target of rapamycin kinase; NDs: neurodegenerative diseases; NSC34: neuroblastoma x spinal cord 34; PBS: phosphate-buffered saline; PD: Parkinson disease; polyQ: polyglutamine; PPARGC1A: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PPP3CB: protein phosphatase 3, catalytic subunit, beta isoform; RT-qPCR: real-time quantitative polymerase chain reaction; SBMA: spinal and bulbar muscular atrophy; SCAs: spinocerebellar ataxias; siRNA: small interfering RNA; SLC2A8: solute carrier family 2, (facilitated glucose transporter), member 8; smNPCs: small molecules neural progenitors cells; SOD1: superoxide dismutase 1; SQSTM1/p62: sequestosome 1; STED: stimulated emission depletion; STUB1: STIP1 homology and U-box containing protein 1; TARDBP/TDP-43: TAR DNA binding protein; TFEB: transcription factor EB; TPP1: tripeptidyl peptidase I; TREH: trehalase (brush-border membrane glycoprotein); WB: western blotting; ZKSCAN3: zinc finger with KRAB and SCAN domains 3.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcineurin/metabolism , Lysosomes/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Trehalose/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/enzymology , Autophagosomes/metabolism , Autophagy/genetics , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Bulbo-Spinal Atrophy, X-Linked/drug therapy , Bulbo-Spinal Atrophy, X-Linked/metabolism , Calcineurin/genetics , Calcium/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Down-Regulation/genetics , Humans , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/ultrastructure , Lysosomes/drug effects , Lysosomes/enzymology , Lysosomes/ultrastructure , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Motor Neurons/enzymology , Motor Neurons/ultrastructure , Neuroprotection/drug effects , Neuroprotection/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Trehalose/analogs & derivatives , Tripeptidyl-Peptidase 1 , Unfolded Protein Response/genetics
16.
Mol Cancer Ther ; 17(11): 2451-2461, 2018 11.
Article in English | MEDLINE | ID: mdl-30135216

ABSTRACT

Glioblastoma (GB) is the most lethal, aggressive, and diffuse brain tumor. The main challenge for successful treatment is targeting the cancer stem cell (CSC) subpopulation responsible for tumor origin, progression, and recurrence. Chloride Intracellular Channel 1 (CLIC1), highly expressed in CSCs, is constitutively present in the plasma membrane where it is associated with chloride ion permeability. In vitro, CLIC1 inhibition leads to a significant arrest of GB CSCs in G1 phase of the cell cycle. Furthermore, CLIC1 knockdown impairs tumor growth in vivo Here, we demonstrate that CLIC1 membrane localization and function is specific for GB CSCs. Mesenchymal stem cells (MSC) do not show CLIC1-associated chloride permeability, and inhibition of CLIC1 protein function has no influence on MSC cell-cycle progression. Investigation of the basic functions of GB CSCs reveals a constitutive state of oxidative stress and cytoplasmic alkalinization compared with MSCs. Both intracellular oxidation and cytoplasmic pH changes have been reported to affect CLIC1 membrane functional expression. We now report that in CSCs these three elements are temporally linked during CSC G1-S transition. Impeding CLIC1-mediated chloride current prevents both intracellular ROS accumulation and pH changes. CLIC1 membrane functional impairment results in GB CSCs resetting from an allostatic tumorigenic condition to a homeostatic steady state. In contrast, inhibiting NADPH oxidase and NHE1 proton pump results in cell death of both GB CSCs and MSCs. Our results show that CLIC1 membrane protein is crucial and specific for GB CSC proliferation, and is a promising pharmacologic target for successful brain tumor therapies. Mol Cancer Ther; 17(11); 2451-61. ©2018 AACR.


Subject(s)
Brain Neoplasms/pathology , Chloride Channels/metabolism , G1 Phase , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Reactive Oxygen Species/metabolism , S Phase , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cyclin D1/metabolism , Humans , Hydrogen-Ion Concentration , Middle Aged , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neoplastic Stem Cells/metabolism , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Sodium-Hydrogen Exchanger 1/metabolism , Time Factors
17.
Genes Dev ; 31(21): 2136-2150, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29196537

ABSTRACT

ESCO1/2 acetyltransferases mediating SMC3 acetylation and sister chromatid cohesion (SCC) are differentially required for genome integrity and development. Here we established chicken DT40 cell lines with mutations in ESCO1/2, SMC3 acetylation, and the cohesin remover WAPL. Both ESCO1 and ESCO2 promoted SCC, while ESCO2 was additionally and specifically required for proliferation and centromere integrity. ESCO1 overexpression fully suppressed the slow proliferation and centromeric separation phenotypes of esco2 cells but only partly suppressed its chromosome arm SCC defects. Concomitant inactivation of ESCO1 and ESCO2 caused lethality owing to compromised mitotic chromosome segregation. Neither wapl nor acetyl-mimicking smc3-QQ mutations rescued esco1 esco2 lethality. Notably, esco1 esco2 wapl conditional mutants showed very severe proliferation defects associated with catastrophic mitoses and also abnormal interphase chromatin organization patterns. The results indicate that cohesion establishment by vertebrate ESCO1/2 is linked to interphase chromatin architecture formation, a newly identified function of cohesin acetyltransferases that is both fundamentally and medically relevant.


Subject(s)
Acetyltransferases/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Structures/genetics , Genomic Instability/genetics , Acetylation , Acetyltransferases/genetics , Animals , Cell Line , Cell Proliferation/genetics , Centromere/genetics , Chickens , Chromosomal Proteins, Non-Histone/genetics , Gene Knockout Techniques , Gene Silencing , Interphase/genetics , Nuclear Proteins/genetics
18.
EMBO Rep ; 17(7): 1061-80, 2016 07.
Article in English | MEDLINE | ID: mdl-27255086

ABSTRACT

The mechanisms of tumor cell dissemination and the contribution of membrane trafficking in this process are poorly understood. Through a functional siRNA screening of human RAB GTPases, we found that RAB2A, a protein essential for ER-to-Golgi transport, is critical in promoting proteolytic activity and 3D invasiveness of breast cancer (BC) cell lines. Remarkably, RAB2A is amplified and elevated in human BC and is a powerful and independent predictor of disease recurrence in BC patients. Mechanistically, RAB2A acts at two independent trafficking steps. Firstly, by interacting with VPS39, a key component of the late endosomal HOPS complex, it controls post-endocytic trafficking of membrane-bound MT1-MMP, an essential metalloprotease for matrix remodeling and invasion. Secondly, it further regulates Golgi transport of E-cadherin, ultimately controlling junctional stability, cell compaction, and tumor invasiveness. Thus, RAB2A is a novel trafficking determinant essential for regulation of a mesenchymal invasive program of BC dissemination.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Golgi Apparatus/metabolism , Matrix Metalloproteinase 14/metabolism , rab GTP-Binding Proteins/metabolism , Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Endosomes/metabolism , Exocytosis , Extracellular Matrix/metabolism , Female , Gene Expression , Gene Expression Profiling , Gene Silencing , Homeodomain Proteins/metabolism , Humans , Neoplasm Invasiveness , Prognosis , Protein Transport , Proteolysis , Recurrence , Tumor Suppressor Proteins/metabolism , rab GTP-Binding Proteins/genetics
19.
J Cell Sci ; 127(Pt 20): 4381-95, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25179599

ABSTRACT

Inflammatory cells acquire a polarized phenotype to migrate towards sites of infection or injury. A conserved polarity complex comprising PAR-3, PAR-6 and atypical protein kinase C (aPKC) relays extracellular polarizing cues to control cytoskeletal and signaling networks affecting morphological and functional polarization. However, there is no evidence that myeloid cells use PAR signaling to migrate vectorially in three-dimensional (3D) environments in vivo. Using genetically encoded bioprobes and high-resolution live imaging, we reveal the existence of F-actin oscillations in the trailing edge and constant repositioning of the microtubule organizing center (MTOC) to direct leukocyte migration in wounded medaka fish larvae (Oryzias latipes). Genetic manipulation in live myeloid cells demonstrates that the catalytic activity of aPKC and the regulated interaction with PAR-3 and PAR-6 are required for consistent F-actin oscillations, MTOC perinuclear mobility, aPKC repositioning and wound-directed migration upstream of Rho kinase (also known as ROCK or ROK) activation. We propose that the PAR complex coordinately controls cytoskeletal changes affecting both the generation of traction force and the directionality of leukocyte migration to sites of injury.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cell Movement , Leukocytes/physiology , Microtubule-Organizing Center/physiology , Protein Kinase C/metabolism , Zebrafish Proteins/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Cell Polarity/genetics , Cells, Cultured , Multiprotein Complexes/genetics , Mutation/genetics , Oryzias , Protein Kinase C/genetics , Protein Transport , Zebrafish , Zebrafish Proteins/genetics , rho-Associated Kinases/metabolism
20.
Cell ; 158(3): 633-46, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083873

ABSTRACT

ATR controls chromosome integrity and chromatin dynamics. We have previously shown that yeast Mec1/ATR promotes chromatin detachment from the nuclear envelope to counteract aberrant topological transitions during DNA replication. Here, we provide evidence that ATR activity at the nuclear envelope responds to mechanical stress. Human ATR associates with the nuclear envelope during S phase and prophase, and both osmotic stress and mechanical stretching relocalize ATR to nuclear membranes throughout the cell cycle. The ATR-mediated mechanical response occurs within the range of physiological forces, is reversible, and is independent of DNA damage signaling. ATR-defective cells exhibit aberrant chromatin condensation and nuclear envelope breakdown. We propose that mechanical forces derived from chromosome dynamics and torsional stress on nuclear membranes activate ATR to modulate nuclear envelope plasticity and chromatin association to the nuclear envelope, thus enabling cells to cope with the mechanical strain imposed by these molecular processes.


Subject(s)
Nuclear Envelope/metabolism , Stress, Mechanical , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Checkpoint Kinase 1 , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Osmosis , Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...