Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Colloids Surf B Biointerfaces ; 239: 113931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38718475

ABSTRACT

This study revealed the potential of magnesium whitlockite [WH: Ca18Mg2(HPO4)2(PO4)12] nanoparticles (WH NPs) for anti-inflammatory and anti-cancer therapies. Although magnesium whitlockite possesses promising biological properties, its effects on inflammation and cancer remain unexplored. In this study, we address this gap by synthesizing WH NPs and demonstrating their multifaceted functionalities. Through detailed characterization, we revealed the synthesis pathway involving brushite as a precursor, with magnesium ions incorporated during hydrothermal treatment. WH NPs exhibited anti-inflammatory properties by significantly reducing the production of key inflammatory markers (NO, TNF-α, and IL-6). Furthermore, they display promising anti-cancer activity by inhibiting the proliferation of MDA-MB-231 breast cancer cells. Our findings not only establish a deeper understanding of WH NP synthesis but also highlight their potential for the development of innovative cancer and inflammatory treatments.


Subject(s)
Anti-Inflammatory Agents , Antineoplastic Agents , Cell Proliferation , Magnesium , Nanoparticles , Humans , Magnesium/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Cell Proliferation/drug effects , Nanoparticles/chemistry , Cell Line, Tumor , Animals , Particle Size , Interleukin-6/metabolism , Mice , Drug Screening Assays, Antitumor , Tumor Necrosis Factor-alpha/metabolism , Cell Survival/drug effects , Surface Properties , Nitric Oxide/metabolism , Nitric Oxide/biosynthesis , Calcium Phosphates
2.
Nat Commun ; 12(1): 2198, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33850160

ABSTRACT

Cancer is initiated by somatic mutations in oncogenes or tumor suppressor genes. However, additional alterations provide selective advantages to the tumor cells to resist treatment and develop metastases. Their identification is of paramount importance. Reduced expression of EFA6B (Exchange Factor for ARF6, B) is associated with breast cancer of poor prognosis. Here, we report that loss of EFA6B triggers a transcriptional reprogramming of the cell-to-ECM interaction machinery and unleashes CDC42-dependent collective invasion in collagen. In xenograft experiments, MCF10 DCIS.com cells, a DCIS-to-IDC transition model, invades faster when knocked-out for EFA6B. In addition, invasive and metastatic tumors isolated from patients have lower expression of EFA6B and display gene ontology signatures identical to those of EFA6B knock-out cells. Thus, we reveal an EFA6B-regulated molecular mechanism that controls the invasive potential of mammary cells; this finding opens up avenues for the treatment of invasive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Animals , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Knockout Techniques , Humans , Mice , Mice, Nude , Transcriptome , cdc42 GTP-Binding Protein
3.
Molecules ; 26(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673086

ABSTRACT

Breast cancer is a major disease for women worldwide, where mortality is associated with tumour cell dissemination to distant organs. While the number of efficient anticancer therapies increased in the past 20 years, treatments targeting the invasive properties of metastatic tumour cells are still awaited. Various studies analysing invasive breast cancer cell lines have demonstrated that Arf6 is an important player of the migratory and invasive processes. These observations make Arf6 and its regulators potential therapeutic targets. As of today, no drug effective against Arf6 has been identified, with one explanation being that the activation of Arf6 is dependent on the presence of lipid membranes that are rarely included in drug screening. To overcome this issue we have set up a fluorescence-based high throughput screening that follows overtime the activation of Arf6 at the surface of lipid membranes. Using this unique screening assay, we isolated several compounds that affect Arf6 activation, among which the antibiotic chlortetracycline (CTC) appeared to be the most promising. In this report, we describe CTC in vitro biochemical characterization and show that it blocks both the Arf6-stimulated collective migration and cell invasion in a 3D collagen I gel of the invasive breast cancer cell line MDA-MB-231. Thus, CTC appears as a promising hit to target deadly metastatic dissemination and a powerful tool to unravel the molecular mechanisms of Arf6-mediated invasive processes.


Subject(s)
ADP-Ribosylation Factors/genetics , Breast Neoplasms/drug therapy , Chlortetracycline/pharmacology , ADP-Ribosylation Factor 6 , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Signal Transduction/drug effects
4.
J Cell Sci ; 134(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33483367

ABSTRACT

Ciliogenesis is a coordinated process initiated by the recruitment and fusion of pre-ciliary vesicles at the distal appendages of the mother centriole through mechanisms that remain unclear. Here, we report that EFA6A (also known as PSD), an exchange factor for the small G protein Arf6, is involved in early stage of ciliogenesis by promoting the fusion of distal appendage vesicles forming the ciliary vesicle. EFA6A is present in the vicinity of the mother centriole before primary cilium assembly and prior to the arrival of Arl13B-containing vesicles. During ciliogenesis, EFA6A initially accumulates at the mother centriole and later colocalizes with Arl13B along the ciliary membrane. EFA6A depletion leads to the inhibition of ciliogenesis, the absence of centrosomal Rab8-positive structures and the accumulation of Arl13B-positive vesicles around the distal appendages. Our results uncover a novel fusion machinery, comprising EFA6A, Arf6 and Arl13B, that controls the coordinated fusion of ciliary vesicles docked at the distal appendages of the mother centriole.


Subject(s)
ADP-Ribosylation Factors , Centrioles , Cilia , Guanine Nucleotide Exchange Factors , Animals , Cell Line , Cytoplasmic Vesicles
5.
J Am Coll Emerg Physicians Open ; 1(6): 1418-1426, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33392546

ABSTRACT

Coronavirus disease 2019 (COVID-19) is associated with a severe acute respiratory condition requiring respiratory support and mechanical ventilation. Based on the pathophysiology and clinical course of the disease, a therapeutic approach can be adapted. Three phases have been identified, in which different strategies are recommended in a stepwise invasiveness approach. In the second or acute phase, patients are frequently admitted to the ICU for severe pneumonia and hypoxemia with evidence of a proinflammatory and hypercoagulable state. This stage is an opportunity to intervene early in the disease. Medical strategies and mechanical ventilation should be individualized to improve outcomes.

6.
Sci Rep ; 9(1): 19209, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844082

ABSTRACT

The Arf6-specific exchange factor EFA6 is involved in the endocytic/recycling pathway for different cargos. In addition EFA6 acts as a powerful actin cytoskeleton organizer, a function required for its role in the establishment of the epithelial cell polarity and in neuronal morphogenesis. We previously showed that the C-terminus of EFA6 (EFA6-Ct) is the main domain which contributes to actin reorganization. Here, by in vitro and in vivo experiments, we sought to decipher, at the molecular level, how EFA6 controls the dynamic and structuring of actin filaments. We showed that EFA6-Ct interferes with actin polymerization by interacting with and capping actin filament barbed ends. Further, in the presence of actin mono-filaments, the addition of EFA6-Ct triggered the formation of actin bundles. In cells, when the EFA6-Ct was directed to the plasma membrane, as is the case for the full-length protein, its expression induced the formation of membrane protrusions enriched in actin cables. Collectively our data explain, at least in part, how EFA6 plays an essential role in actin organization by interacting with and bundling F-actin.


Subject(s)
Actins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Cell Polarity/physiology , Cytoskeleton/metabolism , Humans , Neurons/metabolism , Protein Domains
7.
J Cell Biol ; 217(9): 3161-3182, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30061108

ABSTRACT

The endocytic protein NUMB has been implicated in the control of various polarized cellular processes, including the acquisition of mesenchymal migratory traits through molecular mechanisms that have only been partially defined. Here, we report that NUMB is a negative regulator of a specialized set of understudied, apically restricted, actin-based protrusions, the circular dorsal ruffles (CDRs), induced by either PDGF or HGF stimulation. Through its PTB domain, NUMB binds directly to an N-terminal NPLF motif of the ARF6 guanine nucleotide exchange factor, EFA6B, and promotes its exchange activity in vitro. In cells, a NUMB-EFA6B-ARF6 axis regulates the recycling of the actin regulatory cargo RAC1 and is critical for the formation of CDRs that mark the acquisition of a mesenchymal mode of motility. Consistently, loss of NUMB promotes HGF-induced cell migration and invasion. Thus, NUMB negatively controls membrane protrusions and the acquisition of mesenchymal migratory traits by modulating EFA6B-ARF6 activity.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Movement/physiology , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , Mesoderm/metabolism , Nerve Tissue Proteins/metabolism , ADP-Ribosylation Factor 6 , Cell Line, Tumor , Cell Polarity , HeLa Cells , Hepatocyte Growth Factor/metabolism , Humans , Membrane Proteins/genetics , Mesoderm/cytology , Nerve Tissue Proteins/genetics , Platelet-Derived Growth Factor/metabolism , Protein Binding , Protein Domains , RNA Interference , RNA, Small Interfering/genetics , rac1 GTP-Binding Protein/metabolism
8.
J Integr Neurosci ; 17(3-4): 679-693, 2018.
Article in English | MEDLINE | ID: mdl-30103346

ABSTRACT

Cognitive processing is needed to elicit emotional responses. At the same time, emotional responses modulate and guide cognition to enable adaptive responses to the environment. However, most empirical studies and theoretical models of cognitive functions have been investigated without taking into account emotion, which is considered interference that is counterproductive to the correct functioning of the cognitive system. To understand how complex behaviors are carried out in the brain, an understanding of the interactions between emotion and cognition may be indispensable. Given the enormous scope of this topic for both cognition and emotion, these concepts will not be further defined here; instead, this review will be relatively narrow in scope and will emphasize several brain systems involved in the interactions between emotion and working memory because an important dimension of cognition involves working memory function. In attempting to understand the relationship between emotion and working memory, we will describe the projections of a set of brain structures that support our emotional life and the neuromodulator dopamine (which is also involved in emotion processing and incentive motivational behavior) in the prefrontal cortex. According to the literature, working memory engages the cortical regions. Thus, the prefrontal cortex, particularly the dorsolateral prefrontal cortex (DLPFC), although commonly viewed as a purely cognitive area, provides a test for the hypothesis that working memory and emotion are strongly integrated in the brain. In this review, we provide an overview of neuropsychological, neuroanatomical and molecular evidence, with the aim of establishing the extent to which working memory and emotion are related.


Subject(s)
Brain/physiology , Cognition/physiology , Emotions/physiology , Memory, Short-Term/physiology , Animals , Brain/anatomy & histology , Humans
9.
J Cell Sci ; 131(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29246944

ABSTRACT

A key step of epithelial morphogenesis is the creation of the lumen. Luminogenesis by hollowing proceeds through the fusion of apical vesicles at cell-cell contacts. The small nascent lumens grow through extension, coalescence and enlargement, coordinated with cell division, to give rise to a single central lumen. Here, by using MDCK cells grown in 3D-culture, we show that EFA6A (also known as PSD) participates in luminogenesis. EFA6A recruits α-actinin 1 (ACTN1) through direct binding. In polarized cells, ACTN1 was found to be enriched at the tight junction where it acts as a primary effector of EFA6A for normal luminogenesis. Both proteins are essential for the lumen extension and enlargement, where they mediate their effect by regulating the cortical acto-myosin contractility. Finally, ACTN1 was also found to act as an effector for the isoform EFA6B (also known as PSD4) in the human mammary tumoral MCF7 cell line. EFA6B restored the glandular morphology of this tumoral cell line in an ACTN1-dependent manner. Thus, we identified new regulators of cyst luminogenesis essential for the proper maturation of a newly-formed lumen into a single central lumen.


Subject(s)
Actinin/metabolism , Morphogenesis , Nerve Tissue Proteins/metabolism , Animals , Dogs , Guanine Nucleotide Exchange Factors , Humans , MCF-7 Cells , Madin Darby Canine Kidney Cells , Protein Binding
10.
Crit Care Med ; 44(10): 1861-70, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27359085

ABSTRACT

OBJECTIVES: The 2009-2010 influenza A (H1N1pdm09) pandemic caused substantial morbidity and mortality among young patients; however, mortality estimates have been confounded by regional differences in eligibility criteria and inclusion of selected populations. In 2013-2014, H1N1pdm09 became North America's dominant seasonal influenza strain. Our objective was to compare the baseline characteristics, resources, and treatments with outcomes among critically ill patients with influenza A (H1N1pdm09) in Mexican and Canadian hospitals in 2014 using consistent eligibility criteria. DESIGN: Observational study and a survey of available healthcare setting resources. SETTING: Twenty-one hospitals, 13 in Mexico and eight in Canada. PATIENTS: Critically ill patients with confirmed H1N1pdm09 during 2013-2014 influenza season. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: The main outcome measures were 90-day mortality and independent predictors of mortality. Among 165 adult patients with H1N1pdm09-related critical illness between September 2013 and March 2014, mean age was 48.3 years, 64% were males, and nearly all influenza was community acquired. Patients were severely hypoxic (median PaO2-to-FIO2 ratio, 83 mm Hg), 97% received mechanical ventilation, with mean positive end-expiratory pressure of 14 cm H2O at the onset of critical illness and 26.7% received rescue oxygenation therapy with prone ventilation, extracorporeal life support, high-frequency oscillatory ventilation, or inhaled nitric oxide. At 90 days, mortality was 34.6% (13.9% in Canada vs 50.5% in Mexico, p < 0.0001). Independent predictors of mortality included lower presenting PaO2-to-FIO2 ratio (odds ratio, 0.89 per 10-point increase [95% CI, 0.80-0.99]), age (odds ratio, 1.49 per 10 yr increment [95% CI, 1.10-2.02]), and requiring critical care in Mexico (odds ratio, 7.76 [95% CI, 2.02-27.35]). ICUs in Canada generally had more beds, ventilators, healthcare personnel, and rescue oxygenation therapies. CONCLUSIONS: Influenza A (H1N1pdm09)-related critical illness still predominantly affects relatively young to middle-aged patients and is associated with severe hypoxemic respiratory failure. The local critical care system and available resources may be influential determinants of patient outcome.


Subject(s)
Critical Illness/therapy , Influenza A Virus, H1N1 Subtype , Influenza, Human/physiopathology , Influenza, Human/therapy , Intensive Care Units/statistics & numerical data , Adrenal Cortex Hormones/economics , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Antiviral Agents/economics , Antiviral Agents/therapeutic use , Canada/epidemiology , Critical Illness/epidemiology , Extracorporeal Membrane Oxygenation/economics , Extracorporeal Membrane Oxygenation/methods , Female , Health Expenditures , Humans , Influenza, Human/economics , Influenza, Human/epidemiology , Male , Mexico/epidemiology , Middle Aged , Respiration, Artificial/economics , Respiration, Artificial/methods , Respiratory Insufficiency/physiopathology , Respiratory Insufficiency/therapy
11.
J Cell Biol ; 210(2): 243-56, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26169353

ABSTRACT

In Myxococcus xanthus the gliding motility machinery is assembled at the leading cell pole to form focal adhesions, translocated rearward to propel the cell, and disassembled at the lagging pole. We show that MglA, a Ras-like small G-protein, is an integral part of this machinery. In this function, MglA stimulates the assembly of the motility complex by directly connecting it to the MreB actin cytoskeleton. Because the nucleotide state of MglA is regulated spatially and MglA only binds MreB in the guanosine triphosphate-bound form, the motility complexes are assembled at the leading pole and dispersed at the lagging pole where the guanosine triphosphatase activating protein MglB disrupts the MglA-MreB interaction. Thus, MglA acts as a nucleotide-dependent molecular switch to regulate the motility machinery spatially. The function of MreB in motility is independent of its function in peptidoglycan synthesis, representing a coopted function. Our findings highlight a new function for the MreB cytoskeleton and suggest that G-protein-cytoskeleton interactions are a universally conserved feature.


Subject(s)
Bacterial Proteins/metabolism , Cytoskeletal Proteins/metabolism , Myxococcus xanthus/metabolism , Bacterial Adhesion , Focal Adhesions/metabolism , Myxococcus xanthus/cytology , Peptidoglycan/biosynthesis , Protein Binding , Protein Interaction Mapping , Protein Transport
12.
Proc Natl Acad Sci U S A ; 111(34): 12378-83, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114232

ABSTRACT

Guanine nucleotide exchange factors (GEFs) of the exchange factor for Arf6 (EFA6), brefeldin A-resistant Arf guanine nucleotide exchange factor (BRAG), and cytohesin subfamilies activate small GTPases of the Arf family in endocytic events. These ArfGEFs carry a pleckstrin homology (PH) domain in tandem with their catalytic Sec7 domain, which is autoinhibitory and supports a positive feedback loop in cytohesins but not in BRAGs, and has an as-yet unknown role in EFA6 regulation. In this study, we analyzed how EFA6A is regulated by its PH and C terminus (Ct) domains by reconstituting its GDP/GTP exchange activity on membranes. We found that EFA6 has a previously unappreciated high efficiency toward Arf1 on membranes and that, similar to BRAGs, its PH domain is not autoinhibitory and strongly potentiates nucleotide exchange on anionic liposomes. However, in striking contrast to both cytohesins and BRAGs, EFA6 is regulated by a negative feedback loop, which is mediated by an allosteric interaction of Arf6-GTP with the PH-Ct domain of EFA6 and monitors the activation of Arf1 and Arf6 differentially. These observations reveal that EFA6, BRAG, and cytohesins have unanticipated commonalities associated with divergent regulatory regimes. An important implication is that EFA6 and cytohesins may combine in a mixed negative-positive feedback loop. By allowing EFA6 to sustain a pool of dormant Arf6-GTP, such a circuit would fulfill the absolute requirement of cytohesins for activation by Arf-GTP before amplification of their GEF activity by their positive feedback loop.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factors/metabolism , Nerve Tissue Proteins/metabolism , ADP-Ribosylation Factor 1/chemistry , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/genetics , Amino Acid Substitution , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cricetinae , Enzyme Activation , Feedback, Physiological , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Kinetics , Liposomes , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Models, Biological , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfotransferases/chemistry , Sulfotransferases/genetics , Sulfotransferases/metabolism
13.
Cancer Res ; 74(19): 5493-506, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25115298

ABSTRACT

One of the earliest events in epithelial carcinogenesis is the dissolution of tight junctions and cell polarity signals that are essential for normal epithelial barrier function. Here, we report that EFA6B, a guanine nucleotide exchange factor for the Ras superfamily protein Arf6 that helps assemble and stabilize tight junction, is required to maintain apico-basal cell polarity and mesenchymal phenotypes in mammary epithelial cells. In organotypic three-dimensional cell cultures, endogenous levels of EFA6B were critical to determine epithelial-mesenchymal status. EFA6B downregulation correlated with a mesenchymal phenotype and ectopic expression of EFA6B hampered TGFß-induced epithelial-to-mesenchymal transition (EMT). Transcriptomic and immunohistochemical analyses of human breast tumors revealed that the reduced expression of EFA6B was associated with loss of tight junction components and with increased signatures of EMT, cancer stemness, and poor prognosis. Accordingly, tumors with low levels of EFA6B were enriched in the aggressive triple-negative and claudin-low breast cancer subtypes. Our results identify EFA6B as a novel antagonist in breast cancer and they point to its regulatory and signaling pathways as rational therapeutic targets in aggressive forms of this disease.


Subject(s)
Breast Neoplasms/physiopathology , Guanine Nucleotide Exchange Factors/physiology , Breast Neoplasms/pathology , Cell Line, Tumor , Claudin-3/metabolism , Epithelial-Mesenchymal Transition , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Middle Aged , RNA, Messenger/genetics , Tight Junctions/physiology
14.
Proc Natl Acad Sci U S A ; 111(26): 9473-8, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24979773

ABSTRACT

Members of the Arf family of small G proteins are involved in membrane traffic and organelle structure. They control the recruitment of coat proteins, and modulate the structure of actin filaments and the lipid composition of membranes. The ADP-ribosylation factor 6 (Arf6) isoform and the exchange factor for Arf6 (EFA6) are known to regulate the endocytic pathway of many different receptors. To determine the molecular mechanism of the EFA6/Arf6 function in vesicular transport, we searched for new EFA6 partners. In a two-hybrid screening using the catalytic Sec7 domain as a bait, we identified endophilin as a new partner of EFA6. Endophilin contains a Bin/Amphiphysin/Rvs (BAR) domain responsible for membrane bending, and an SH3 domain responsible for the recruitment of dynamin and synaptojanin, two proteins involved, respectively, in the fission and uncoating of clathrin-coated vesicles. By using purified proteins, we confirmed the direct interaction, and identified the N-BAR domain as the binding motif to EFA6A. We showed that endophilin stimulates the catalytic activity of EFA6A on Arf6. In addition, we observed that the Sec7 domain competes with flat but not with highly curved lipid membranes to bind the N-BAR. In cells, expression of EFA6A recruits endophilin to EFA6A-positive plasma membrane ruffles, whereas expression of endophilin rescues the EFA6A-mediated inhibition of transferrin internalization. Overall, our results support a model whereby EFA6 recruits endophilin on flat areas of the plasma membrane to control Arf6 activation and clathrin-mediated endocytosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Endocytosis/physiology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Recombinant Proteins/metabolism , ADP-Ribosylation Factor 6 , Clathrin/metabolism , Cloning, Molecular , Polymerase Chain Reaction , Recombinant Proteins/genetics , Xanthenes
15.
Nature ; 502(7472): 567-70, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24097348

ABSTRACT

In most eukaryotic cells microtubules undergo post-translational modifications such as acetylation of α-tubulin on lysine 40, a widespread modification restricted to a subset of microtubules that turns over slowly. This subset of stable microtubules accumulates in cell protrusions and regulates cell polarization, migration and invasion. However, mechanisms restricting acetylation to these microtubules are unknown. Here we report that clathrin-coated pits (CCPs) control microtubule acetylation through a direct interaction of the α-tubulin acetyltransferase αTAT1 (refs 8, 9) with the clathrin adaptor AP2. We observe that about one-third of growing microtubule ends contact and pause at CCPs and that loss of CCPs decreases lysine 40 acetylation levels. We show that αTAT1 localizes to CCPs through a direct interaction with AP2 that is required for microtubule acetylation. In migrating cells, the polarized orientation of acetylated microtubules correlates with CCP accumulation at the leading edge, and interaction of αTAT1 with AP2 is required for directional migration. We conclude that microtubules contacting CCPs become acetylated by αTAT1. In migrating cells, this mechanism ensures the acetylation of microtubules oriented towards the leading edge, thus promoting directional cell locomotion and chemotaxis.


Subject(s)
Acetyltransferases/metabolism , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Microtubules/metabolism , Acetylation , Adaptor Protein Complex 2/metabolism , Biocatalysis , Cell Movement , Coated Pits, Cell-Membrane/enzymology , HeLa Cells , Humans , Microtubules/chemistry , Protein Binding , Tubulin/metabolism
16.
J Cell Sci ; 125(Pt 17): 4026-35, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22611259

ABSTRACT

ß2-adrenergic receptor (ß2AR), a member of the GPCR (G-protein coupled receptor) family, is internalized in a ligand- and ß-arrestin-dependent manner into early endosomes, and subsequently recycled back to the plasma membrane. Here, we report that ß-arrestin promotes the activation of the small G protein Arf6, which regulates the recycling and degradation of ß2AR. We demonstrate in vitro that the C-terminal region of ß-arrestin1 interacts directly and simultaneously with Arf6GDP and its specific exchange factor EFA6, to promote Arf6 activation. Similarly, the ligand-mediated activation of ß2AR leads to the formation of Arf6GTP in vivo in a ß-arrestin-dependent manner. Expression of either EFA6 or an activated Arf6 mutant caused accumulation of ß2AR in the degradation pathway. This phenotype could be rescued by the expression of an activated mutant of Rab4, suggesting that Arf6 acts upstream of Rab4. We propose a model in which Arf6 plays an essential role in ß2AR desensitization. The ligand-mediated stimulation of ß2AR relocates ß-arrestin to the plasma membrane, and triggers the activation of Arf6 by EFA6. The activation of Arf6 leads to accumulation of ß2AR in the degradation pathway, and negatively controls Rab4-dependent fast recycling to prevent the re-sensitization of ß2AR.


Subject(s)
ADP-Ribosylation Factors/metabolism , Endocytosis , Receptors, Adrenergic, beta-2/metabolism , ADP-Ribosylation Factor 6 , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Arrestins/chemistry , Arrestins/metabolism , Biocatalysis/drug effects , Cattle , Cell Compartmentation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Exchange Factors , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Isoproterenol/pharmacology , Ligands , Lysosomes/drug effects , Lysosomes/metabolism , Models, Biological , Nerve Tissue Proteins/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , beta-Arrestins , rab4 GTP-Binding Proteins/metabolism
17.
PLoS Biol ; 8(7): e1000430, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20652021

ABSTRACT

Regulated cell polarity is central to many cellular processes. We investigated the mechanisms that govern the rapid switching of cell polarity (reversals) during motility of the bacterium Myxococcus xanthus. Cellular reversals are mediated by pole-to-pole oscillations of motility proteins and the frequency of the oscillations is under the control of the Frz chemosensory system. However, the molecular mechanism that creates dynamic polarity remained to be characterized. In this work, we establish that polarization is regulated by the GTP cycle of a Ras-like GTPase, MglA. We initially sought an MglA regulator and purified a protein, MglB, which was found to activate GTP hydrolysis by MglA. Using live fluorescence microscopy, we show that MglA and MglB localize at opposite poles and oscillate oppositely when cells reverse. In absence of MglB, MglA-YFP accumulates at the lagging cell end, leading to a strikingly aberrant reversal cycle. Spatial control of MglA is achieved through the GAP activity of MglB because an MglA mutant that cannot hydrolyze GTP accumulates at the lagging cell end, despite the presence of MglB. Genetic and cell biological studies show that the MglA-GTP cycle controls dynamic polarity and the reversal switch. The study supports a model wherein a chemosensory signal transduction system (Frz) activates reversals by relieving a spatial inhibition at the back pole of the cells: reversals are allowed by Frz-activated switching of MglB to the opposite pole, allowing MglA-GTP to accumulate at the back of the cells and create the polarity switch. In summary, our results provide insight into how bacteria regulate their polarity dynamically, revealing unsuspected conserved regulations with eukaryots.


Subject(s)
Bacterial Proteins/metabolism , Cell Polarity , GTPase-Activating Proteins/metabolism , Myxococcus xanthus/cytology , Myxococcus xanthus/metabolism , ras Proteins/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , Models, Biological , Movement , Protein Transport , Recombinant Fusion Proteins/metabolism , Signal Transduction
18.
EMBO J ; 29(9): 1499-509, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20339350

ABSTRACT

In epithelial cells, the tight junction (TJ) functions as a permeability barrier and is involved in cellular differentiation and proliferation. Although many TJ proteins have been characterized, little is known about the sequence of events and temporal regulation of TJ assembly in response to adhesion cues. We report here that the deubiquitinating enzyme USP9x has a critical function in TJ biogenesis by controlling the levels of the exchange factor for Arf6 (EFA6), a protein shown to facilitate TJ formation, during a narrow temporal window preceding the establishment of cell polarity. At steady state, EFA6 is constitutively ubiquitinated and turned over by the proteasome. However, at newly forming contacts, USP9x-mediated deubiquitination protects EFA6 from proteasomal degradation, leading to a transient increase in EFA6 levels. Consistent with this model, USP9x and EFA6 transiently co-localize at primordial epithelial junctions. Furthermore, knockdown of either EFA6 or USP9x impairs TJ biogenesis and EFA6 overexpression rescues TJ biogenesis in USP9x-knockdown cells. As the loss of cell polarity is a critical event in the metastatic spread of cancer, these findings may help to understand the pathology of human carcinomas.


Subject(s)
Epithelial Cells/metabolism , Nerve Tissue Proteins/metabolism , Tight Junctions/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Cell Line , Dogs , Epithelial Cells/cytology , Gene Expression Regulation , Gene Knockdown Techniques , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Proteome/metabolism , Ubiquitin Thiolesterase/analysis , Ubiquitin Thiolesterase/genetics , Ubiquitination
19.
EMBO J ; 28(18): 2835-45, 2009 Sep 16.
Article in English | MEDLINE | ID: mdl-19644450

ABSTRACT

The JNK-interacting proteins, JIP3 and JIP4, are specific effectors of the small GTP-binding protein ARF6. The interaction of ARF6-GTP with the second leucine zipper (LZII) domains of JIP3/JIP4 regulates the binding of JIPs to kinesin-1 and dynactin. Here, we report the crystal structure of ARF6-GTP bound to the JIP4-LZII at 1.9 A resolution. The complex is a heterotetramer with dyad symmetry arranged in an ARF6-(JIP4)(2)-ARF6 configuration. Comparison of the ARF6-JIP4 interface with the equivalent region of ARF1 shows the structural basis of JIP4's specificity for ARF6. Using site-directed mutagenesis and surface plasmon resonance, we further show that non-conserved residues at the switch region borders are the key structural determinants of JIP4 specificity. A structure-derived model of the association of the ARF6-JIP3/JIP4 complex with membranes shows that the JIP4-LZII coiled-coil should lie along the membrane to prevent steric hindrances, resulting in only one ARF6 molecule bound. Such a heterotrimeric complex gives insights to better understand the ARF6-mediated motor switch regulatory function.


Subject(s)
ADP-Ribosylation Factors/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Kinesins/chemistry , Microtubule-Associated Proteins/chemistry , ADP-Ribosylation Factor 6 , Amino Acid Sequence , Dimerization , Dynactin Complex , Guanosine Triphosphate/metabolism , Models, Biological , Molecular Conformation , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Surface Plasmon Resonance
20.
Opt Express ; 17(14): 11450-6, 2009 Jul 06.
Article in English | MEDLINE | ID: mdl-19582060

ABSTRACT

Short lived plasma channels generated through filamentation of femtosecond laser pulses in air can be revived after several milliseconds by a delayed nanosecond pulse. Electrons initially ionized from oxygen molecules and subsequently captured by neutral oxygen molecules provide the long-lived reservoir of low affinity allowing this process. A Bessel-like nanosecond-duration laser beam can easily detach these weakly bound electrons and multiply them in an avalanche process. We have experimentally demonstrated such revivals over a channel length of 50 cm by focusing the nanosecond laser with an axicon.


Subject(s)
Lasers , Optics and Photonics , Air , Electrons , Equipment Design , Oxygen/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...