Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 574, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750105

ABSTRACT

Metastases are the major cause of cancer-related death, yet, molecular weaknesses that could be exploited to prevent tumor cells spreading are poorly known. Here, we found that perturbing hydrolase transport to lysosomes by blocking either the expression of IGF2R, the main receptor responsible for their trafficking, or GNPT, a transferase involved in the addition of the specific tag recognized by IGF2R, reduces melanoma invasiveness potential. Mechanistically, we demonstrate that the perturbation of this traffic, leads to a compensatory lysosome neo-biogenesis devoided of degradative enzymes. This regulatory loop relies on the stimulation of TFEB transcription factor expression. Interestingly, the inhibition of this transcription factor playing a key role of lysosome production, restores melanomas' invasive potential in the absence of hydrolase transport. These data implicate that targeting hydrolase transport in melanoma could serve to develop new therapies aiming to prevent metastasis by triggering a physiological response stimulating TFEB expression in melanoma.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Hydrolases , Lysosomes , Melanoma , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Lysosomes/metabolism , Hydrolases/metabolism , Hydrolases/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Receptor, IGF Type 2/metabolism , Receptor, IGF Type 2/genetics , Neoplasm Metastasis , Protein Transport , Gene Expression Regulation, Neoplastic
2.
J Biomed Mater Res A ; 112(7): 1107-1123, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38433552

ABSTRACT

The ever-growing need for new tissue and organ replacement approaches paved the way for tissue engineering. Successful tissue regeneration requires an appropriate scaffold, which allows cell adhesion and provides mechanical support during tissue repair. In this light, an interpenetrating polymer network (IPN) system based on biocompatible polysaccharides, dextran (Dex) and gellan (Ge), was designed and proposed as a surface that facilitates cell adhesion in tissue engineering applications. The new matrix was developed in glycerol, an unconventional solvent, before the chemical functionalization of the polymer backbone, which provides the system with enhanced properties, such as increased stiffness and bioadhesiveness. Dex was modified introducing methacrylic groups, which are known to be sensitive to UV light. At the same time, Ge was functionalized with RGD moieties, known as promoters for cell adhesion. The printability of the systems was evaluated by exploiting the ability of glycerol to act as a co-initiator in the process, speeding up the kinetics of crosslinking. Following semi-IPNs formation, the solvent was removed by extensive solvent exchange with HEPES and CaCl2, leading to conversion into IPNs due to the ionic gelation of Ge chains. Mechanical properties were investigated and IPNs ability to promote osteoblasts adhesion was evaluated on thin-layer, 3D-printed disk films. Our results show a significant increase in adhesion on hydrogels decorated with RGD moieties, where osteoblasts adopted the spindle-shaped morphology typical of adherent mesenchymal cells. Our findings support the use of RGD-decorated Ge/Dex IPNs as new matrices able to support and facilitate cell adhesion in the perspective of bone tissue regeneration.


Subject(s)
Cell Adhesion , Dextrans , Glycerol , Methacrylates , Oligopeptides , Polysaccharides, Bacterial , Printing, Three-Dimensional , Oligopeptides/chemistry , Oligopeptides/pharmacology , Glycerol/chemistry , Glycerol/pharmacology , Methacrylates/chemistry , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/pharmacology , Dextrans/chemistry , Cell Adhesion/drug effects , Animals , Mice , Humans
3.
Sci Adv ; 9(33): eade2120, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37585527

ABSTRACT

Fibroblasts play a fundamental role in tumor development. Among other functions, they regulate cancer cells' migration through rearranging the extracellular matrix, secreting soluble factors, and establishing direct physical contacts with cancer cells. Here, we report that migrating fibroblasts deposit on the substrate a network of tubular structures that serves as a guidance cue for cancer cell migration. Such membranous tubular network, hereafter called tracks, is stably anchored to the substrate in a ß5-integrin-dependent manner. We found that cancer cells specifically adhere to tracks by using clathrin-coated structures that pinch and engulf tracks. Tracks thus represent a spatial memory of fibroblast migration paths that is read and erased by cancer cells directionally migrating along them. We propose that fibroblast tracks represent a topography-based intercellular communication system capable of steering cancer cell migration.


Subject(s)
Cues , Neoplasms , Humans , Cell Movement/physiology , Fibroblasts/physiology , Extracellular Matrix
4.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36250940

ABSTRACT

Integrin endocytosis is essential for many fundamental cellular processes. Whether and how the internalization impacts cellular mechanics remains elusive. Whereas previous studies reported the contribution of the integrin activator, talin, in force development, the involvement of inhibitors is less documented. We identified ICAP-1 as an integrin inhibitor involved in mechanotransduction by co-working with NME2 to control clathrin-mediated endocytosis of integrins at the edge of focal adhesions (FA). Loss of ICAP-1 enables ß3-integrin-mediated force generation independently of ß1 integrin. ß3-integrin-mediated forces were associated with a decrease in ß3 integrin dynamics stemming from their reduced diffusion within adhesion sites and slow turnover of FA. The decrease in ß3 integrin dynamics correlated with a defect in integrin endocytosis. ICAP-1 acts as an adaptor for clathrin-dependent endocytosis of integrins. ICAP-1 controls integrin endocytosis by interacting with NME2, a key regulator of dynamin-dependent clathrin-coated pits fission. Control of clathrin-mediated integrin endocytosis by an inhibitor is an unprecedented mechanism to tune forces at FA.


Subject(s)
Clathrin , Endocytosis , Focal Adhesions , Integrin beta1 , Integrin beta3 , Clathrin/metabolism , Endocytosis/physiology , Integrin beta1/genetics , Mechanotransduction, Cellular , Talin/genetics
5.
Eur J Med Chem ; 240: 114573, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35797900

ABSTRACT

A series of quinoline and quinazoline analogs were designed and synthesized as new tubulin polymerization (TP) and histone deacetylases (HDAC) inhibitors. Compounds 12a and 12d showed the best cytotoxicity activities against a panel of human cancer cell lines with an averaged IC50 value of 0.6 and 0.7 nM, respectively. Furthermore, these lead compounds showed good activities against CA-4-resistant colon-carcinoma and multidrug-resistant leukemia cells. In addition, compounds 12a and 12d induced HT29 cell cycle arrest in the G2/M phase and produced caspase-induced apoptosis of HT29 cells through mitochondrial dysfunction. Also, 12a and 12d inhibited HDAC8, 6, and 11 activities. Furthermore, lead compound 12a exhibited higher metabolic stability than isoCA-4 and was highly potent in suppressing tumor growth in the fibrosarcoma MCA205 tumor model. Collectively, these studies suggest that 12a represents a new dual inhibitor of TP and HDAC activities, which makes it a suitable candidate for further investigations in clinical development.


Subject(s)
Antineoplastic Agents , Quinolines , Cell Line, Tumor , Cell Proliferation , Drug Design , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Polymerization , Quinolines/pharmacology , Repressor Proteins , Tubulin/metabolism
6.
Sci Adv ; 7(45): eabf4647, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34739323

ABSTRACT

Migrating cells navigate in complex environments through sensing and interpreting biochemical and/or mechanical cues. Here, we report that recently identified tubular clathrin/AP-2 lattices (TCALs), a subset of clathrin-coated structures (CCSs) that pinch collagen fibers, mechanically control directed migration along fibers decorated with ligands of CCS cargoes in three-dimensional (3D) environments. We observed that epidermal growth factor or low-density lipoprotein bound to collagen fibers leads to increased local nucleation and accumulation of TCALs. By using engineered, mixed collagen networks, we demonstrate that this mechanism selectively increases local forces applied on ligand-decorated fibers. We show that these effects depend on the ligand's receptors but do not rely on their ability to trigger signaling events. We propose that the preferential accumulation of TCALs along ligand-decorated fibers steers migration in 3D environments. We conclude that ligand-regulated, local TCAL accumulation results in asymmetric force distribution that orients cell migration in 3D environments.

7.
Front Bioeng Biotechnol ; 8: 597721, 2020.
Article in English | MEDLINE | ID: mdl-33195167

ABSTRACT

Mechanical stimuli have profound effects on the cellular architecture and functions. Over the past two decades, considerable progress has been made in unraveling the molecular machineries that confer cells the ability to sense and transduce mechanical input into biochemical signals. This has resulted in the identification of several force-sensing proteins or mechanically activated ion channels distributed throughout most cell types, whereby the plasma membrane, cytoskeleton, and the nucleus have garnered much attention. Although organelles from the endomembrane system make up significant portion of cell volume and play pivotal roles in the spatiotemporal distribution of signaling molecules, they have received surprisingly little attention in mechanobiology. In this mini-review, we summarize results that document participation of the endomembrane system in sensing and responding to mechanical cues.

8.
J Cell Sci ; 133(17)2020 09 01.
Article in English | MEDLINE | ID: mdl-32788230

ABSTRACT

Cells experience mechanical stresses in different physiological and pathological settings. Clathrin-coated structures (CCSs) are sensitive to such perturbations in a way that often results in a mechanical impairment of endocytic budding. Compressive stress is a mechanical perturbation that leads to increased membrane tension and promotes proliferative signals. Here, we report that compression leads to frustration of CCSs and that CCSs are required to potentiate receptor-mediated signaling in these conditions. We show that cell compression stalled CCS dynamics and slowed down the dynamic exchange of CCS components. As previously reported, compression-induced paracrine activation of the epidermal growth factor receptor (EGFR) was the primary cause of ERK (ERK1 and ERK2, also known as MAPK3 and MAPK1, respectively) activation in these conditions. We observed that EGFR was efficiently recruited at CCSs upon compression and that CCSs were required for full ERK activation. In addition, we demonstrated that compression-induced frustrated CCSs could also increase ligand-dependent signaling of other receptors. We thus propose that CCS frustration resulting from mechanical perturbations can potentiate signaling through different receptors, with potential important consequences for the adaptation of the cell to its environment.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Clathrin , Frustration , Endocytosis , Ligands , Signal Transduction
9.
J Cell Sci ; 133(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32499318

ABSTRACT

Clathrin-mediated endocytosis is the main entry route for most cell surface receptors and their ligands. It is regulated by clathrin-coated structures that are endowed with the ability to cluster receptors and to locally bend the plasma membrane, resulting in the formation of receptor-containing vesicles that bud into the cytoplasm. This canonical role of clathrin-coated structures has been shown to play a fundamental part in many different aspects of cell physiology. However, it has recently become clear that the ability of clathrin-coated structures to deform membranes can be perturbed. In addition to chemical or genetic alterations, numerous environmental conditions can physically prevent or slow down membrane bending and/or budding at clathrin-coated structures. The resulting 'frustrated endocytosis' is emerging as not merely a passive consequence, but one that actually fulfils some very specific and important cellular functions. In this Review, we provide an historical and defining perspective on frustrated endocytosis in the clathrin pathway of mammalian cells, before discussing its causes and highlighting the possible functional consequences in physiology and diseases.


Subject(s)
Clathrin , Endocytosis , Animals , Cell Membrane , Clathrin-Coated Vesicles , Coated Pits, Cell-Membrane
10.
J Cell Biol ; 218(7): 2086-2095, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31208994

ABSTRACT

An understanding of the mechanisms whereby cell adhesion complexes (ACs) relay signals bidirectionally across the plasma membrane is necessary to interpret the role of adhesion in regulating migration, differentiation, and growth. A range of AC types has been defined, but to date all have similar compositions and are dependent on a connection to the actin cytoskeleton. Recently, a new class of AC has been reported that normally lacks association with both the cytoskeleton and integrin-associated adhesome components, but is rich in components of the clathrin-mediated endocytosis machinery. The characterization of this new type of adhesion structure, which is emphasized by mitotic cells and cells in long-term culture, identifies a hitherto underappreciated link between the adhesion machinery and clathrin structures at the plasma membrane. While this discovery has implications for how ACs are assembled and disassembled, it raises many other issues. Consequently, to increase awareness within the field, and stimulate research, we explore a number of the most significant questions below.


Subject(s)
Actin Cytoskeleton/genetics , Cell Adhesion/genetics , Cell Membrane/genetics , Clathrin/genetics , Actin Cytoskeleton/chemistry , Animals , Cell Differentiation/genetics , Cell Membrane/chemistry , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/ultrastructure , Mitosis/genetics
11.
Blood ; 133(16): 1778-1788, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30602618

ABSTRACT

Filamin A (FLNa) links the cell membrane with the cytoskeleton and is central in several cellular processes. Heterozygous mutations in the X-linked FLNA gene are associated with a large spectrum of conditions, including macrothrombocytopenia, called filaminopathies. Using an isogenic pluripotent stem cell model derived from patients, we show that the absence of the FLNa protein in megakaryocytes (MKs) leads to their incomplete maturation, particularly the inability to produce proplatelets. Reduction in proplatelet formation potential is associated with a defect in actomyosin contractility, which results from inappropriate RhoA activation. This dysregulated RhoA activation was observed when MKs were plated on fibrinogen but not on other matrices (fibronectin, vitronectin, collagen 1, and von Willebrand factor), strongly suggesting a role for FLNa/αIIbß3 interaction in the downregulation of RhoA activity. This was confirmed by experiments based on the overexpression of FLNa mutants deleted in the αIIbß3-binding domain and the RhoA-interacting domain, respectively. Finally, pharmacological inhibition of the RhoA-associated kinase ROCK1/2 restored a normal phenotype and proplatelet formation. Overall, this work suggests a new etiology for macrothrombocytopenia, in which increased RhoA activity is associated with disrupted FLNa/αIIbß3 interaction.


Subject(s)
Filamins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombocytopenia/etiology , Female , Fibrinogen/metabolism , Filamins/genetics , Humans , Megakaryocytes/chemistry , Megakaryocytes/pathology , Mutation , Protein Binding/physiology , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
12.
Nat Commun ; 9(1): 3825, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30237420

ABSTRACT

It is generally assumed that cells interrogate the mechanical properties of their environment by pushing and pulling on the extracellular matrix (ECM). For instance, acto-myosin-dependent contraction forces exerted at focal adhesions (FAs) allow the cell to actively probe substrate elasticity. Here, we report that a subset of long-lived and flat clathrin-coated structures (CCSs), also termed plaques, are contractility-independent mechanosensitive signaling platforms. We observed that plaques assemble in response to increasing substrate rigidity and that this is independent of FAs, actin and myosin-II activity. We show that plaque assembly depends on αvß5 integrin, and is a consequence of frustrated endocytosis whereby αvß5 tightly engaged with the stiff substrate locally stalls CCS dynamics. We also report that plaques serve as platforms for receptor-dependent signaling and are required for increased Erk activation and cell proliferation on stiff environments. We conclude that CCSs are mechanotransduction structures that sense substrate rigidity independently of cell contractility.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Endocytosis , Mechanotransduction, Cellular , Cell Line , Cell Proliferation , Clathrin-Coated Vesicles/ultrastructure , Humans , MAP Kinase Signaling System , Receptors, Vitronectin/metabolism
13.
J Pathol ; 246(3): 352-365, 2018 11.
Article in English | MEDLINE | ID: mdl-30058725

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), which is the primary cause of pancreatic cancer mortality, is poorly responsive to currently available interventions. Identifying new targets that drive PDAC formation and progression is critical for developing alternative therapeutic strategies to treat this lethal malignancy. Using genetic and pharmacological approaches, we investigated in vivo and in vitro whether uptake of the monoamine serotonin [5-hydroxytryptamine (5-HT)] is required for PDAC development. We demonstrated that pancreatic acinar cells have the ability to readily take up 5-HT in a transport-mediated manner. 5-HT uptake promoted activation of the small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1), which is required for transdifferentiation of acinar cells into acinar-to-ductal metaplasia (ADM), a key determinant in PDAC development. Consistent with the central role played by Rac1 in ADM formation, inhibition of the 5-HT transporter Sert (Slc6a4) with fluoxetine reduced ADM formation both in vitro and in vivo in a cell-autonomous manner. In addition, fluoxetine treatment profoundly compromised the stromal reaction and affected the proliferation and lipid metabolism of malignant PDAC cells. We propose that Sert is a promising therapeutic target to counteract the early event of ADM, with the potential to stall the initiation and progression of pancreatic carcinogenesis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Proliferation , Genes, ras , Neuropeptides/metabolism , Pancreas/enzymology , Pancreatic Neoplasms/enzymology , Serotonin/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transdifferentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chick Embryo , Disease Models, Animal , Enzyme Activation , Fluoxetine/pharmacology , Genetic Predisposition to Disease , Humans , Metaplasia , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Phenotype , Rats , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
14.
Science ; 356(6343)2017 06 16.
Article in English | MEDLINE | ID: mdl-28619886

ABSTRACT

Migrating cells often use focal adhesions in order to move. Focal adhesions are less prominent in cells migrating in three-dimensional (3D) as compared with 2D environments. We looked for alternative adhesion structures supporting cell migration. We analyzed the dynamics of clathrin-coated pits in cells migrating in a 3D environment of collagen fibers. Both topological cues and local engagement of integrins triggered the accumulation of clathrin-coated structures on fibers. Clathrin/adaptor protein 2 (AP-2) lattices pinched collagen fibers by adopting a tube-like morphology and regulated adhesion to fibers in an endocytosis-independent manner. During migration, tubular clathrin/AP-2 lattices stabilized cellular protrusions by providing anchoring points to collagen fibers. Thus, tubular clathrin/AP-2 lattices promote cell adhesion that, in coordination with focal adhesions, supports cell migration in 3D.


Subject(s)
Adaptor Protein Complex 2/metabolism , Cell Movement/physiology , Clathrin/metabolism , Collagen/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Humans , Integrins/metabolism , Protein Transport
15.
Small GTPases ; 6(2): 104-7, 2015.
Article in English | MEDLINE | ID: mdl-26156751

ABSTRACT

The role of polarity in cancer is an emerging research area and loss of polarity is widely considered an important event in cancer. Among the polarity regulating molecules, the small GTPase Cdc42 was extensively studied. Most attention was given to Cdc42 signaling at the plasma membrane, but whether and how Cdc42 is regulated at endomembranes remained poorly understood. Moreover, whether the endomembrane pool of Cdc42 is of any relevance to cell polarity was unknown. In our recent work, we identified a complex between the Golgi matrix protein GM130 and RasGRF and showed that it is responsible for regulating the Golgi pool of Cdc42, but had no effect on the plasma membrane pool of Cdc42. Depletion of GM130 disrupted apico-basal polarity as well as front-rear polarity, indicating that the spatial pool of Cdc42 is functionally relevant. The biomedical relevance of this finding was supported by the observation than GM130 is progressively lost in colorectal cancer. These findings support a role of the endomembrane pool of Cdc42 in cell polarity and point to a potential role of alterations of this pool in cancer.


Subject(s)
Cell Polarity , Golgi Apparatus/metabolism , Neoplasms/metabolism , Second Messenger Systems , cdc42 GTP-Binding Protein/metabolism , ras-GRF1/metabolism , Animals , Autoantigens/metabolism , Humans , Membrane Proteins/metabolism , Neoplasms/pathology
16.
Cell Cycle ; 14(8): 1139-47, 2015.
Article in English | MEDLINE | ID: mdl-25892554

ABSTRACT

Spatially distinct pools of the small GTPase Cdc42 were observed, but the major focus of research so far has been to investigate its signaling at the plasma membrane. We recently showed that the Golgi pool of Cdc42 is relevant for cell polarity and that it is regulated by GM130, a Golgi matrix protein. Loss of GM130 abrogated cell polarity and consistent with the notion that polarity is frequently impaired in cancer, we found that GM130 is downregulated in colorectal cancer. Whether the loss of GM130 solely affects polarity, or whether it affects other processes relevant for tumorigenesis remains unclear. In a panel of breast cancer cells lines, we investigated the consequences of GM130 depletion on traits of relevance for tumor progression, such as survival, proliferation, adhesion, migration and invasion. We show that cellular assays that depend on polarity, such as chemotaxis and wound scratch assays, are only of limited use to investigate the role of polarity modulators in cancer. Depletion of GM130 increases cellular velocity and increases the invasiveness of breast cancer cells, therefore supporting the view that alterations of polarity contribute to tumor progression.


Subject(s)
Autoantigens/genetics , Autoantigens/metabolism , Breast Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Polarity/genetics , Cell Proliferation , Doxorubicin/pharmacology , Female , Fluorescence Resonance Energy Transfer , Golgi Apparatus/metabolism , Humans , MCF-7 Cells , Membrane Proteins/antagonists & inhibitors , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , cdc42 GTP-Binding Protein/metabolism
17.
Mol Cell ; 57(6): 995-1010, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25684205

ABSTRACT

The small Rho GTPase RAC1 is an essential regulator of cellular signaling that controls actin rearrangements and cell motility. Here, we identify a novel CUL3 RING ubiquitin ligase complex, containing the substrate adaptors KBTBD6 and KBTBD7, that mediates ubiquitylation and proteasomal degradation of TIAM1, a RAC1-specific GEF. Increasing the abundance of TIAM1 by depletion of KBTBD6 and/or KBTBD7 leads to elevated RAC1 activity, changes in actin morphology, loss of focal adhesions, reduced proliferation, and enhanced invasion. KBTBD6 and KBTBD7 employ ATG8 family-interacting motifs to bind preferentially to GABARAP proteins. Surprisingly, ubiquitylation and degradation of TIAM1 by CUL3(KBTBD6/KBTBD7) depends on its binding to GABARAP proteins. Our study reveals that recruitment of CUL3(KBTBD6/KBTBD7) to GABARAP-containing vesicles regulates the abundance of membrane-associated TIAM1 and subsequently spatially restricted RAC1 signaling. Besides their role in autophagy and trafficking, we uncovered a previously unknown function of GABARAP proteins as membrane-localized signaling scaffolds.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cullin Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubule-Associated Proteins/metabolism , Trans-Activators/metabolism , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Apoptosis Regulatory Proteins , Autophagy-Related Protein 8 Family , Cullin Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Multimerization , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Trans-Activators/genetics , Ubiquitination , rac1 GTP-Binding Protein/genetics
18.
J Cell Sci ; 128(4): 670-82, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25526736

ABSTRACT

We currently lack a broader mechanistic understanding of the integration of the early secretory pathway with other homeostatic processes such as cell growth. Here, we explore the possibility that Sec16A, a major constituent of endoplasmic reticulum exit sites (ERES), acts as an integrator of growth factor signaling. Surprisingly, we find that Sec16A is a short-lived protein that is regulated by growth factors in a manner dependent on Egr family transcription factors. We hypothesize that Sec16A acts as a central node in a coherent feed-forward loop that detects persistent growth factor stimuli to increase ERES number. Consistent with this notion, Sec16A is also regulated by short-term growth factor treatment that leads to increased turnover of Sec16A at ERES. Finally, we demonstrate that Sec16A depletion reduces proliferation, whereas its overexpression increases proliferation. Together with our finding that growth factors regulate Sec16A levels and its dynamics on ERES, we propose that this protein acts as an integrator linking growth factor signaling and secretion. This provides a mechanistic basis for the previously proposed link between secretion and proliferation.


Subject(s)
COP-Coated Vesicles/metabolism , Cell Proliferation/physiology , Endoplasmic Reticulum/metabolism , Secretory Pathway/physiology , Vesicular Transport Proteins/metabolism , Cell Line , Cell Proliferation/genetics , Early Growth Response Protein 1/genetics , Early Growth Response Protein 3/genetics , Early Growth Response Transcription Factors/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Hep G2 Cells , Humans , Monomeric GTP-Binding Proteins/genetics , NM23 Nucleoside Diphosphate Kinases/genetics , Nucleoside-Diphosphate Kinase/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Signal Transduction , Vesicular Transport Proteins/genetics
19.
Nat Commun ; 5: 4839, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25208761

ABSTRACT

The small GTPase Cdc42 is a key regulator of polarity, but little is known in mammals about its spatial regulation and the relevance of spatial Cdc42 pools for polarity. Here we report the identification of a GM130-RasGRF complex as a regulator of Cdc42 at the Golgi. Silencing GM130 results in RasGRF-dependent inhibition of the Golgi pool of Cdc42, but does not affect Cdc42 at the cell surface. Furthermore, active Cdc42 at the Golgi is important to sustain asymmetric front-rear Cdc42-GTP distribution in directionally migrating cells. Concurrent to Cdc42 inhibition, silencing GM130 also results in RasGRF-dependent Ras-ERK pathway activation. Moreover, depletion of GM130 is sufficient to induce E-cadherin downregulation, indicative of a loss in cell polarity and epithelial identity. Accordingly, GM130 expression is frequently lost in colorectal and breast cancer patients. These findings establish a previously unrecognized role for a GM130-RasGRF-Cdc42 connection in regulating polarity and tumorigenesis.


Subject(s)
Autoantigens/genetics , Carcinogenesis/genetics , Cell Polarity/genetics , Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/genetics , cdc42 GTP-Binding Protein/genetics , ras Guanine Nucleotide Exchange Factors/genetics , ras-GRF1/genetics , Antigens, CD , Autoantigens/metabolism , Cadherins/metabolism , Cell Line, Tumor , Gene Silencing , Humans , MAP Kinase Signaling System/physiology , MCF-7 Cells , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , ras-GRF1/metabolism
20.
Methods Cell Biol ; 118: 345-58, 2013.
Article in English | MEDLINE | ID: mdl-24295317

ABSTRACT

Research on the secretory pathway in the past three decades accounts for our known knowledge about the composition and architecture of organelles and about the machinery that regulates membrane transport. An emerging topic in the past few years was the discovery that the secretory pathway is regulated by signaling, and in this regard, the Golgi apparatus received major attention. In the current chapter, we will highlight various techniques that are used by us and others to study signaling at the Golgi. We describe methods to study lipid and protein phosphorylation at the Golgi and various techniques for studying spatial activation of GTPases at this organelle. We also discuss how combining these techniques and improving their limitations is important for gaining a better understanding of how the Golgi intersects with various signal transduction pathways.


Subject(s)
Golgi Apparatus/metabolism , Signal Transduction , Diglycerides/metabolism , Enzyme Activation , Fluorescence Resonance Energy Transfer , GTP Phosphohydrolases/metabolism , HeLa Cells , Humans , Phosphorylation , Photobleaching , Protein Kinase C/metabolism , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...