Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Gut Microbes ; 16(1): 2320291, 2024.
Article in English | MEDLINE | ID: mdl-38417029

ABSTRACT

Intratumoral bacteria flexibly contribute to cellular and molecular tumor heterogeneity for supporting cancer recurrence through poorly understood mechanisms. Using spatial metabolomic profiling technologies and 16SrRNA sequencing, we herein report that right-sided colorectal tumors are predominantly populated with Colibactin-producing Escherichia coli (CoPEC) that are locally establishing a high-glycerophospholipid microenvironment with lowered immunogenicity. It coincided with a reduced infiltration of CD8+ T lymphocytes that produce the cytotoxic cytokines IFN-γ where invading bacteria have been geolocated. Mechanistically, the accumulation of lipid droplets in infected cancer cells relied on the production of colibactin as a measure to limit genotoxic stress to some extent. Such heightened phosphatidylcholine remodeling by the enzyme of the Land's cycle supplied CoPEC-infected cancer cells with sufficient energy for sustaining cell survival in response to chemotherapies. This accords with the lowered overall survival of colorectal patients at stage III-IV who were colonized by CoPEC when compared to patients at stage I-II. Accordingly, the sensitivity of CoPEC-infected cancer cells to chemotherapies was restored upon treatment with an acyl-CoA synthetase inhibitor. By contrast, such metabolic dysregulation leading to chemoresistance was not observed in human colon cancer cells that were infected with the mutant strain that did not produce colibactin (11G5∆ClbQ). This work revealed that CoPEC locally supports an energy trade-off lipid overload within tumors for lowering tumor immunogenicity. This may pave the way for improving chemoresistance and subsequently outcome of CRC patients who are colonized by CoPEC.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Peptides , Polyketides , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Tumor Microenvironment , Drug Resistance, Neoplasm , Mutagens/metabolism , Neoplasm Recurrence, Local , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology , Polyketides/metabolism , Lipids
2.
Gut Microbes ; 15(2): 2265138, 2023 12.
Article in English | MEDLINE | ID: mdl-37842920

ABSTRACT

Recently, an intestinal dysbiotic microbiota with enrichment in oral cavity bacteria has been described in colorectal cancer (CRC) patients. Here, we characterize and investigate one of these oral pathobionts, the Gram-positive anaerobic coccus Parvimonas micra. We identified two phylotypes (A and B) exhibiting different phenotypes and adhesion capabilities. We observed a strong association of phylotype A with CRC, with its higher abundance in feces and in tumoral tissue compared with the normal homologous colonic mucosa, which was associated with a distinct methylation status of patients. By developing an in vitro hypoxic co-culture system of human primary colonic cells with anaerobic bacteria, we show that P. micra phylotype A alters the DNA methylation profile promoters of key tumor-suppressor genes, oncogenes, and genes involved in epithelial-mesenchymal transition. In colonic mucosa of CRC patients carrying P. micra phylotype A, we found similar DNA methylation alterations, together with significant enrichment of differentially expressed genes in pathways involved in inflammation, cell adhesion, and regulation of actin cytoskeleton, providing evidence of P. micra's possible role in the carcinogenic process.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/genetics , Firmicutes/genetics , Bacteria , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology
3.
Gut Microbes ; 15(1): 2229569, 2023.
Article in English | MEDLINE | ID: mdl-37417545

ABSTRACT

Colorectal cancer (CRC) patients are frequently colonized by colibactin-producing Escherichia coli (CoPEC) (>40%), which enhances tumorigenesis in mouse models of CRC. We observed that 50% of CoPEC also contains the cnf1 gene, which encodes cytotoxic necrotizing factor-1 (CNF1), an enhancer of the eukaryotic cell cycle. The impact of its co-occurrence with colibactin (Clb) has not yet been investigated. We evaluated the impact of CNF1 on colorectal tumorigenesis using human colonic epithelial HT-29 cells and CRC-susceptible ApcMin/+ mice inoculated with the CoPEC 21F8 clinical strain (Clb+Cnf+) or 21F8 isogenic mutants (Clb+Cnf-, Clb-Cnf+ and Clb-Cnf-). Infection with the Clb+Cnf- strain induced higher levels of inflammatory cytokines and senescence markers both in vitro and in vivo compared to those induced by infection with the Clb+Cnf+ strain. In contrast, the Clb+Cnf- and Clb+Cnf+ strains generated similar levels of DNA damage in HT-29 cells and in colonic murine tissues. Furthermore, the ApcMin/+ mice inoculated with the Clb+Cnf- strain developed significantly more tumors than the mice inoculated with the Clb+Cnf+ strain or the isogenic mutants, and the composition of their microbiota was changed. Finally, rectal administration of the CNF1 protein in ApcMin/+ mice inoculated with the Clb+Cnf- strain significantly decreased tumorigenesis and inflammation. Overall, this study provides evidence that CNF1 decreases the carcinogenic effects of CoPEC in ApcMin/+ mice by decreasing CoPEC-induced cellular senescence and inflammation.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Gastrointestinal Microbiome , Mice , Humans , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Colon , Carcinogenesis , Cell Transformation, Neoplastic , Inflammation
4.
Nat Commun ; 13(1): 6059, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229487

ABSTRACT

Extracellular matrix (ECM) elasticity is perceived by cells via focal adhesion structures, which transduce mechanical cues into chemical signalling to conform cell behavior. Although the contribution of ECM compliance to the control of cell migration or division is extensively studied, little is reported regarding infectious processes. We study this phenomenon with the extraintestinal Escherichia coli pathogen UTI89. We show that UTI89 takes advantage, via its CNF1 toxin, of integrin mechanoactivation to trigger its invasion into cells. We identify the HACE1 E3 ligase-interacting protein Optineurin (OPTN) as a protein regulated by ECM stiffness. Functional analysis establishes a role of OPTN in bacterial invasion and integrin mechanical coupling and for stimulation of HACE1 E3 ligase activity towards the Rac1 GTPase. Consistent with a role of OPTN in cell mechanics, OPTN knockdown cells display defective integrin-mediated traction force buildup, associated with limited cellular invasion by UTI89. Nevertheless, OPTN knockdown cells display strong mechanochemical adhesion signalling, enhanced Rac1 activation and increased cyclin D1 translation, together with enhanced cell proliferation independent of ECM stiffness. Together, our data ascribe a new function to OPTN in mechanobiology.


Subject(s)
Cyclin D1 , Integrins , Cell Division , Cyclin D1/metabolism , Integrins/metabolism , Mechanotransduction, Cellular/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , rac1 GTP-Binding Protein/metabolism
5.
Gut Microbes ; 14(1): 2121577, 2022.
Article in English | MEDLINE | ID: mdl-36154446

ABSTRACT

Epidemiological projections point to acquisition of ever-expanding multidrug resistance (MDR) by Escherichia coli, a commensal of the digestive tract and a source of urinary tract pathogens. Bioinformatics analyses of a large collection of E. coli genomes from EnteroBase, enriched in clinical isolates of worldwide origins, suggest the Cytotoxic Necrotizing Factor 1 (CNF1)-toxin encoding gene, cnf1, is preferentially distributed in four common sequence types (ST) encompassing the pandemic E. coli MDR lineage ST131. This lineage is responsible for a majority of extraintestinal infections that escape first-line antibiotic treatment, with known enhanced capacities to colonize the gastrointestinal tract. Statistical projections based on this dataset point to a global expansion of cnf1-positive multidrug-resistant ST131 strains from subclade H30Rx/C2, accounting for a rising prevalence of cnf1-positive strains in ST131. Despite the absence of phylogeographical signals, cnf1-positive isolates segregated into clusters in the ST131-H30Rx/C2 phylogeny, sharing a similar profile of virulence factors and the same cnf1 allele. The suggested dominant expansion of cnf1-positive strains in ST131-H30Rx/C2 led us to uncover the competitive advantage conferred by cnf1 for gut colonization to the clinical strain EC131GY ST131-H30Rx/C2 versus cnf1-deleted isogenic strain. Complementation experiments showed that colon tissue invasion was compromised in the absence of deamidase activity on Rho GTPases by CNF1. Hence, gut colonization factor function of cnf1 was confirmed for another clinical strain ST131-H30Rx/C2. In addition, functional analysis of the cnf1-positive clinical strain EC131GY ST131-H30Rx/C2 and a cnf1-deleted isogenic strain showed no detectable impact of the CNF1 gene on bacterial fitness and inflammation during the acute phase of bladder monoinfection. Together these data argue for an absence of role of CNF1 in virulence during UTI, while enhancing gut colonization capacities of ST131-H30Rx/C2 and suggested expansion of cnf1-positive MDR isolates in subclade ST131-H30Rx/C2.


Subject(s)
Bacterial Toxins , Escherichia coli Infections , Escherichia coli Proteins , Gastrointestinal Microbiome , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Humans , Virulence Factors/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism , rho GTP-Binding Proteins
6.
iScience ; 25(7): 104537, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35769882

ABSTRACT

The development of anti-infectives against a large range of AB-like toxin-producing bacteria includes the identification of compounds disrupting toxin transport through both the endolysosomal and retrograde pathways. Here, we performed a high-throughput screening of compounds blocking Rac1 proteasomal degradation triggered by the Cytotoxic Necrotizing Factor-1 (CNF1) toxin, which was followed by orthogonal screens against two toxins that hijack the endolysosomal (diphtheria toxin) or retrograde (Shiga-like toxin 1) pathways to intoxicate cells. This led to the identification of the molecule C910 that induces the enlargement of EEA1-positive early endosomes associated with sorting defects of CNF1 and Shiga toxins to their trafficking pathways. C910 protects cells against eight bacterial AB toxins and the CNF1-mediated pathogenic Escherichia coli invasion. Interestingly, C910 reduces influenza A H1N1 and SARS-CoV-2 viral infection in vitro. Moreover, parenteral administration of C910 to mice resulted in its accumulation in lung tissues and a reduction in lethal influenza infection.

7.
J Cell Sci ; 135(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-34878112

ABSTRACT

Metabolic studies and animal knockout models point to the critical role of polyunsaturated docosahexaenoic acid (22:6, DHA)-containing phospholipids (DHA-PLs) in physiology. Here, we investigated the impact of DHA-PLs on the dynamics of transendothelial cell macroapertures (TEMs) triggered by RhoA inhibition-associated cell spreading. Lipidomic analyses showed that human umbilical vein endothelial cells (HUVECs) subjected to a DHA diet undergo a 6-fold enrichment in DHA-PLs at the plasma membrane (PM) at the expense of monounsaturated oleic acid-containing PLs (OA-PLs). Consequently, DHA-PL enrichment at the PM induces a reduction in cell thickness and shifts cellular membranes towards a permissive mode of membrane fusion for transcellular tunnel initiation. We provide evidence that a global homeostatic control of membrane tension and cell cortex rigidity minimizes overall changes of TEM area through a decrease of TEM size and lifetime. Conversely, low DHA-PL levels at the PM lead to the opening of unstable and wider TEMs. Together, this provides evidence that variations of DHA-PL levels in membranes affect cell biomechanical properties.


Subject(s)
Docosahexaenoic Acids , Phospholipids , Animals , Cell Membrane/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Endothelial Cells/metabolism , Humans , Membrane Fusion , Phospholipids/metabolism
8.
EMBO Rep ; 20(4)2019 04.
Article in English | MEDLINE | ID: mdl-30804013

ABSTRACT

Ubiquitin domain-containing protein 1 (UBTD1) is highly evolutionary conserved and has been described to interact with E2 enzymes of the ubiquitin-proteasome system. However, its biological role and the functional significance of this interaction remain largely unknown. Here, we demonstrate that depletion of UBTD1 drastically affects the mechanical properties of epithelial cancer cells via RhoA activation and strongly promotes their aggressiveness. On a stiff matrix, UBTD1 expression is regulated by cell-cell contacts, and the protein is associated with ß-catenin at cell junctions. Yes-associated protein (YAP) is a major cell mechano-transducer, and we show that UBTD1 is associated with components of the YAP degradation complex. Interestingly, UBTD1 promotes the interaction of YAP with its E3 ubiquitin ligase ß-TrCP Consequently, in cancer cells, UBTD1 depletion decreases YAP ubiquitylation and triggers robust ROCK2-dependent YAP activation and downstream signaling. Data from lung and prostate cancer patients further corroborate the in cellulo results, confirming that low levels of UBTD1 are associated with poor patient survival, suggesting that biological functions of UBTD1 could be beneficial in limiting cancer progression.


Subject(s)
Disease Susceptibility , Insulin-Like Growth Factor I/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Ubiquitins/metabolism , Cell Adhesion , Cell Cycle Proteins/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Mechanotransduction, Cellular , Models, Biological , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
9.
Nat Commun ; 10(1): 52, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604763

ABSTRACT

The presence of aberrant number of centrioles is a recognized cause of aneuploidy and hallmark of cancer. Hence, centriole duplication needs to be tightly regulated. It has been proposed that centriole separation limits centrosome duplication. The mechanism driving centriole separation is poorly understood and little is known on how this is linked to centriole duplication. Here, we propose that actin-generated forces regulate centriole separation. By imposing geometric constraints via micropatterns, we were able to prove that precise acto-myosin force arrangements control direction, distance and time of centriole separation. Accordingly, inhibition of acto-myosin contractility impairs centriole separation. Alongside, we observed that organization of acto-myosin force modulates specifically the length of S-G2 phases of the cell cycle, PLK4 recruitment at the centrosome and centriole fidelity. These discoveries led us to suggest that acto-myosin forces might act in fundamental mechanisms of aneuploidy prevention.


Subject(s)
Actins/metabolism , Cell Cycle/physiology , Centrioles/metabolism , Myosins/metabolism , Protein Serine-Threonine Kinases/metabolism , Actins/physiology , Aneuploidy , Cell Cycle/drug effects , Centrioles/physiology , HeLa Cells , Humans , Intravital Microscopy/methods , Microscopy, Confocal , Myosins/physiology , Protein Serine-Threonine Kinases/physiology , Thymidine/pharmacology , Time-Lapse Imaging/methods
10.
J Cell Biol ; 217(11): 3839-3852, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30242034

ABSTRACT

Finely tuned regulation of epithelial cell death maintains tissue integrity and homeostasis. At the cellular level, life and death decisions are controlled by environmental stimuli such as the activation of death receptors. We show that cell polarity and adherens junction formation prevent proapoptotic signals emanating from the Fas death receptor. Fas is sequestered in E-cadherin actin-based adhesion structures that are less able to induce downstream apoptosis signaling. Using a proteomic-based approach, we find that the polarity molecule Dlg1 interacts with the C-terminal PDZ-binding site in Fas and that this interaction decreases formation of the death-inducing complex upon engagement with Fas ligand (FasL), thus acting as an additional cell death protection mechanism. We propose that E-cadherin and Dlg1 inhibit FasL-induced cell death by two complementary but partially independent mechanisms that help to maintain epithelial homeostasis by protecting normal polarized epithelia from apoptosis. When polarity is lost, the Fas-cadherin-Dlg1 antiapoptotic complex is disrupted, and FasL can promote the elimination of compromised nonpolarized cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Epithelial Cells/metabolism , Fas Ligand Protein/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , fas Receptor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adherens Junctions/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Polarity , Discs Large Homolog 1 Protein , Epithelial Cells/cytology , Fas Ligand Protein/genetics , Humans , Membrane Proteins/genetics , Multiprotein Complexes/genetics , Protein Domains , Proteomics , fas Receptor/genetics
11.
Pathog Dis ; 76(5)2018 07 01.
Article in English | MEDLINE | ID: mdl-29733372

ABSTRACT

Alterations of the cellular proteome over time due to spontaneous or toxin-mediated enzymatic deamidation of glutamine (Gln) and asparagine (Asn) residues contribute to bacterial infection and might represent a source of aging-related diseases. Here, we put into perspective what is known about the mode of action of the CNF1 toxin from pathogenic Escherichia coli, a paradigm of bacterial deamidases that activate Rho GTPases, to illustrate the importance of determining whether exposure to these factors are risk factors in the etiology age-related diseases, such as cancer. In particular, through in silico analysis of the distribution of the CNF1-like deamidase active site Gly-Cys-(Xaa)n-His sequence motif in bacterial genomes, we unveil the wide distribution of the super-family of CNF-like toxins and CNF-like deamidase domains among members of the Enterobacteriacae and in association with a large variety of toxin delivery systems. We extent our discussion with recent findings concerning cellular systems that control activated Rac1 GTPase stability and provide protection against cancer. These findings point to the urgency for developing holistic approaches toward personalized medicine that include monitoring for asymptomatic carriage of pathogenic toxin-producing bacteria and that ultimately might lead to improved public health and increased lifespans.


Subject(s)
Amidohydrolases/metabolism , Bacterial Toxins/metabolism , Enterobacteriaceae/enzymology , Escherichia coli Proteins/metabolism , Immunologic Factors/metabolism , Virulence Factors/metabolism , rac1 GTP-Binding Protein/metabolism , Amidohydrolases/genetics , Asparagine/metabolism , Bacterial Toxins/genetics , Computational Biology , Enterobacteriaceae Infections/complications , Enterobacteriaceae Infections/pathology , Escherichia coli Proteins/genetics , Glutamine/metabolism , Neoplasms/etiology , Neoplasms/physiopathology , Virulence Factors/genetics
12.
Sci Rep ; 8(1): 1410, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362425

ABSTRACT

The regulation of Rac1 by HACE1-mediated ubiquitination and proteasomal degradation is emerging as an essential element in the maintenance of cell homeostasis. However, how the E3 ubiquitin ligase activity of HACE1 is regulated remains undetermined. Using a proteomic approach, we identified serine 385 as a target of group-I PAK kinases downstream Rac1 activation by CNF1 toxin from pathogenic E. coli. Moreover, cell treatment with VEGF also promotes Ser-385 phosphorylation of HACE1. We have established in vitro that HACE1 is a direct target of PAK1 kinase activity. Mechanistically, we found that the phospho-mimetic mutant HACE1(S385E), as opposed to HACE1(S385A), displays a lower capacity to ubiquitinate Rac1 in cells. Concomitantly, phosphorylation of Ser-385 plays a pivotal role in controlling the oligomerization state of HACE1. Finally, Ser-385 phosphorylated form of HACE1 localizes in the cytosol away from its target Rac1. Together, our data point to a feedback inhibition of HACE1 ubiquitination activity on Rac1 by group-I PAK kinases.


Subject(s)
Serine/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Bacterial Toxins/pharmacology , Cell Line , Escherichia coli Proteins/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Phosphorylation , Protein Multimerization , Proteomics , Ubiquitination , Vascular Endothelial Growth Factor A/pharmacology
13.
Sci Rep ; 7: 44779, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28317937

ABSTRACT

The E3 ubiquitin ligase HACE1 is a potent tumor suppressor that controls cell proliferation and ubiquitylates the small GTPase Rac1 to target it to proteasomal degradation. Whether and how the activity of HACE1 is regulated by the N-terminal ankyrin (ANK) and the middle (MID) domains is ill defined. Here, we identified in the version 64 of the Catalogue of Somatic Mutations in Cancer (COSMIC) 13 missense mutations of hace1 located outside the HECT domain, and found that all lead to defective control of cell proliferation. In addition, several mutations located in the ankyrin domain displayed a dramatic reduction in Rac1 ubiquitylation associated with a decrease of colony formation in soft agar. 3D structure modelling of the 7 ankyrin-repeats coupled to functional analysis identified a surface epitope centered on one of the mutated residue, Gly-175, which is critical for controlling Rac1 binding and ubiquitylation. We also identified a role for the MID domain in conferring the specificity of association of HACE1 to the active form of Rac1. Our study of the functional interplay between HACE1 and Rac1 in cancer thus sheds a new light on the molecular mechanism of Rac1 ubiquitylation by HACE1 and the impact of its cancer-associated mutations in cell proliferation.


Subject(s)
Mutation, Missense/genetics , Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , rac1 GTP-Binding Protein/metabolism , Amino Acid Sequence , Cell Line , Cell Proliferation , Humans , Models, Molecular , Mutant Proteins/chemistry , Protein Binding , Protein Domains , Structure-Activity Relationship , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
14.
Cytoskeleton (Hoboken) ; 72(10): 542-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26403219

ABSTRACT

It remains a challenge to decode the molecular basis of the long-term actin cytoskeleton rearrangements that are governed by the reprogramming of gene expression. Bacillus anthracis lethal toxin (LT) inhibits mitogen-activated protein kinase (MAPK) signaling, thereby modulating gene expression, with major consequences for actin cytoskeleton organization and the loss of endothelial barrier function. Using a laser ablation approach, we characterized the contractile and tensile mechanical properties of LT-induced stress fibers. These actin cables resist pulling forces that are transmitted at cell-matrix interfaces and at cell-cell discontinuous adherens junctions. We report that treating the cells with trichostatin A (TSA), a broad range inhibitor of histone deacetylases (HDACs), or with MS-275, which targets HDAC1, 2 and 3, induces stress fibers. LT decreased the cellular levels of HDAC1, 2 and 3 and reduced the global HDAC activity in the nucleus. Both the LT and TSA treatments induced Rnd3 expression, which is required for the LT-mediated induction of actin stress fibers. Furthermore, we reveal that treating the LT-intoxicated cells with garcinol, an inhibitor of histone acetyl-transferases (HATs), disrupts the stress fibers and limits the monolayer barrier dysfunctions. These data demonstrate the importance of modulating the flux of protein acetylation in order to control actin cytoskeleton organization and the endothelial cell monolayer barrier.


Subject(s)
Actins/chemistry , Antigens, Bacterial/chemistry , Bacillus anthracis/chemistry , Bacterial Toxins/chemistry , Histones/chemistry , Stress Fibers/chemistry , Acetylation , Adherens Junctions , Cell Communication , Cell Nucleus/metabolism , Endothelial Cells/cytology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/chemistry , Light , Microscopy, Fluorescence , Tensile Strength
15.
Mol Cancer Ther ; 14(2): 586-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25527635

ABSTRACT

Cell migration is a critical step in the progression of prostate cancer to the metastatic state, the lethal form of the disease. The antidiabetic drug metformin has been shown to display antitumoral properties in prostate cancer cell and animal models; however, its role in the formation of metastases remains poorly documented. Here, we show that metformin reduces the formation of metastases to fewer solid organs in an orthotopic metastatic prostate cancer cell model established in nude mice. As predicted, metformin hampers cell motility in PC3 and DU145 prostate cancer cells and triggers a radical reorganization of the cell cytoskeleton. The small GTPase Rac1 is a master regulator of cytoskeleton organization and cell migration. We report that metformin leads to a major inhibition of Rac1 GTPase activity by interfering with some of its multiple upstream signaling pathways, namely P-Rex1 (a Guanine nucleotide exchange factor and activator of Rac1), cAMP, and CXCL12/CXCR4, resulting in decreased migration of prostate cancer cells. Importantly, overexpression of a constitutively active form of Rac1, or P-Rex, as well as the inhibition of the adenylate cyclase, was able to reverse the antimigratory effects of metformin. These results establish a novel mechanism of action for metformin and highlight its potential antimetastatic properties in prostate cancer.


Subject(s)
Cell Movement/drug effects , Metformin/pharmacology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , rac1 GTP-Binding Protein/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/metabolism
16.
Cell Host Microbe ; 16(3): 338-50, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25211076

ABSTRACT

Dugesia japonica planarian flatworms are naturally exposed to various microbes but typically survive this challenge. We show that planarians eliminate bacteria pathogenic to Homo sapiens, Caenorhabditis elegans, and/or Drosophila melanogaster and thus represent a model to identify innate resistance mechanisms. Whole-transcriptome analysis coupled with RNAi screening of worms infected with Staphylococcus aureus or Legionella pneumophila identified 18 resistance genes with nine human orthologs, of which we examined the function of MORN2. Human MORN2 facilitates phagocytosis-mediated restriction of Mycobacterium tuberculosis, L. pneumophila, and S. aureus in macrophages. MORN2 promotes the recruitment of LC3, an autophagy protein also involved in phagocytosis, to M. tuberculosis-containing phagosomes and subsequent maturation to degradative phagolysosomes. MORN2-driven trafficking of M. tuberculosis to single-membrane, LC3-positive compartments requires autophagy-related proteins Atg5 and Beclin-1, but not Ulk-1 and Atg13, highlighting the importance of MORN2 in LC3-associated phagocytosis. These findings underscore the value of studying planarian defenses to identify immune factors.


Subject(s)
Helminth Proteins/immunology , Legionella pneumophila/physiology , Microtubule-Associated Proteins/immunology , Phagocytosis , Planarians/immunology , Planarians/microbiology , Staphylococcus aureus/physiology , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/microbiology , Disease Models, Animal , Helminth Proteins/genetics , Humans , Legionella pneumophila/immunology , Microtubule-Associated Proteins/genetics , Planarians/genetics , Staphylococcus aureus/immunology
17.
FASEB J ; 28(2): 692-704, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24174428

ABSTRACT

The aim of this study was to identify the molecular signals produced in human endothelial cells (ECs) by the interaction of α5ß1 integrin with soluble vascular endothelial growth factor receptor-1 (sVEGFR-1) present in the extracellular matrix. We generated a gene expression profile of ECs adhering to sVEGFR-1 or to fibronectin, the classic extracellular matrix ligand for α5ß1 integrin or in a nonadhering condition. Several biological pathways were differently modulated, 3 protein kinase C substrates [adducin, myristoylated alanine-rich protein kinase C substrate (MARCKS), and radixin] were differently expressed and phosphorylated when cells adhering to sVEGFR-1 were compared with those adhering to fibronectin. Rac1 activation and Gα13 protein involvement through the interaction with radixin were also detected after attachment to sVEGFR-1, and these responses depended on active VEGFR-2 signaling. On sVEGFR-1, ECs exhibited a motile phenotype that was consistent with the abundant presence of MARCKS, a stabilizer of dynamic adhesions. Moreover, ECs silenced for radixin expression no longer responded to the proangiogenic VEGFR-1-derived peptide 12. We propose that the presence of sVEGFR-1 in the EC microenvironment directs α5ß1 integrin signaling to generate a dynamic, motile phenotype. Our findings also provide new insights into the mechanism of action of proangiogenic peptide 12, relevant to a therapeutic perspective.


Subject(s)
Cell Adhesion/physiology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Blotting, Western , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Fibronectins/metabolism , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
18.
Nat Commun ; 4: 2180, 2013.
Article in English | MEDLINE | ID: mdl-23864022

ABSTRACT

The Hace1-HECT E3 ligase is a tumor suppressor that ubiquitylates the activated GTP-bound form of the Rho family GTPase Rac1, leading to Rac1 proteasomal degradation. Here we show that, in vertebrates, Hace1 targets Rac1 for degradation when Rac1 is localized to the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase holoenzyme. This event blocks de novo reactive oxygen species generation by Rac1-dependent NADPH oxidases, and thereby confers cellular protection from reactive oxygen species-induced DNA damage and cyclin D1-driven hyper-proliferation. Genetic inactivation of Hace1 in mice or zebrafish, as well as Hace1 loss in human tumor cell lines or primary murine or human tumors, leads to chronic NADPH oxidase-dependent reactive oxygen species elevation, DNA damage responses and enhanced cyclin D1 expression. Our data reveal a conserved ubiquitin-dependent molecular mechanism that controls the activity of Rac1-dependent NADPH oxidase complexes, and thus constitutes the first known example of a tumor suppressor protein that directly regulates reactive oxygen species production in vertebrates.


Subject(s)
NADPH Oxidases/genetics , Neuropeptides/genetics , Protein Isoforms/genetics , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/genetics , rac1 GTP-Binding Protein/genetics , Animals , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , DNA Damage , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Neuropeptides/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/deficiency , Zebrafish , rac1 GTP-Binding Protein/metabolism
19.
Cell Adh Migr ; 6(6): 528-34, 2012.
Article in English | MEDLINE | ID: mdl-23257831

ABSTRACT

Angiogenesis requires the development of a hierarchically branched network of vessels, which undergoes radial expansion and anastomosis to form a close circuit. Branching is achieved by coordinated behavior of endothelial cells that organize into leading "tip" cells and trailing "stalk" cells. Such organization is under control of the Dll4-Notch signaling pathway, which sets a hierarchy in receptiveness of cells to VEGF-A. Recent studies have shed light on a control of the Notch pathway by basement membrane proteins and integrin signaling, disclosing that extracellular matrix exerts active control on vascular branching morphogenesis. We will survey in the present review how extracellular matrix is a multifaceted substrate, which behind a classical structural role hides a powerful conductor function to shape the branching pattern of vessels.


Subject(s)
Extracellular Matrix/metabolism , Morphogenesis , Retina/growth & development , Retinal Vessels/physiology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Extracellular Matrix/genetics , Fibronectins/genetics , Fibronectins/metabolism , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Phosphorylation , Pseudopodia/genetics , Pseudopodia/metabolism , Pseudopodia/physiology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Retina/cytology , Retina/metabolism , Retinal Vessels/embryology , Retinal Vessels/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Small GTPases ; 3(2): 102-6, 2012.
Article in English | MEDLINE | ID: mdl-22790197

ABSTRACT

Rho GTPases undergo ubiquitylation and degradation via the ubiquitin-proteasome pathway. We now report in the November issue of Developmental Cell that the E3 ubiquitin-ligase HACE1 catalyzes the ubiquitylation of GTP-bound Rac1. Depletion of HACE1 leads to an increase of Rac1 activity. We have proposed that HACE1 limits Rac1 activity in cells, a regulation that is usurped by some pathogenic bacteria for efficient invasion of host cell monolayers. We here review these findings in parallel with the regulation of RhoA by the ubiquitin and proteasome system (UPS) and discuss the impact of these regulations on the capacity of Rho GTPases to signal.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , Ubiquitination , rac1 GTP-Binding Protein/metabolism , Animals , Humans , Proteasome Endopeptidase Complex/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...