Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37158597

RESUMO

The amoeba-resistant bacterium Legionella pneumophila causes Legionnaires' disease and employs a type IV secretion system (T4SS) to replicate in the unique, ER-associated Legionella-containing vacuole (LCV). The large fusion GTPase Sey1/atlastin is implicated in ER dynamics, ER-derived lipid droplet (LD) formation, and LCV maturation. Here, we employ cryo-electron tomography, confocal microscopy, proteomics, and isotopologue profiling to analyze LCV-LD interactions in the genetically tractable amoeba Dictyostelium discoideum. Dually fluorescence-labeled D. discoideum producing LCV and LD markers revealed that Sey1 as well as the L. pneumophila T4SS and the Ran GTPase activator LegG1 promote LCV-LD interactions. In vitro reconstitution using purified LCVs and LDs from parental or Δsey1 mutant D. discoideum indicated that Sey1 and GTP promote this process. Sey1 and the L. pneumophila fatty acid transporter FadL were implicated in palmitate catabolism and palmitate-dependent intracellular growth. Taken together, our results reveal that Sey1 and LegG1 mediate LD- and FadL-dependent fatty acid metabolism of intracellular L. pneumophila.


Assuntos
Dictyostelium , Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Legionella pneumophila/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Macrófagos/metabolismo , Dictyostelium/metabolismo , Gotículas Lipídicas/metabolismo , Vacúolos/metabolismo , Legionella/metabolismo , Doença dos Legionários/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
2.
Microbiol Spectr ; 11(3): e0106623, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37036353

RESUMO

Host metabolism reprogramming is a key feature of Mycobacterium tuberculosis (Mtb) infection that enables the survival of this pathogen within phagocytic cells and modulates the immune response facilitating the spread of the tuberculosis disease. Here, we demonstrate that a previously uncharacterized secreted protein from Mtb, Rv1813c, manipulates the host metabolism by targeting mitochondria. When expressed in eukaryotic cells, the protein is delivered to the mitochondrial intermembrane space and promotes the enhancement of host ATP production by boosting the oxidative phosphorylation metabolic pathway. Furthermore, the release of cytochrome c from mitochondria, an early apoptotic event in response to short-term oxidative stress, is delayed in Rv1813c-expressing cells. This study reveals a novel class of mitochondria targeting effectors from Mtb that might participate in host cell metabolic reprogramming and apoptosis control during Mtb infections. IMPORTANCE In this article, using a combination of techniques (bioinformatics, structural biology, and cell biology), we identified and characterized a new class of effectors present only in intracellular mycobacteria. These proteins specifically target host cell mitochondria when ectopically expressed in cells. We showed that one member of this family (Rv1813c) affects mitochondria metabolism in a way that might twist the immune response. This effector also inhibits the cytochrome c exit from mitochondria, suggesting that it might alter normal host cell apoptotic capacities, one of the first defenses of immune cells against Mtb infection.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Mycobacterium tuberculosis/metabolismo , Citocromos c/metabolismo , Tuberculose/microbiologia , Metabolismo Energético , Mitocôndrias/metabolismo , Interações Hospedeiro-Patógeno
3.
EMBO Rep ; 24(3): e56007, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36588479

RESUMO

Legionella pneumophila replicates in macrophages and amoeba within a unique compartment, the Legionella-containing vacuole (LCV). Hallmarks of LCV formation are the phosphoinositide lipid conversion from PtdIns(3)P to PtdIns(4)P, fusion with ER-derived vesicles and a tight association with the ER. Proteomics of purified LCVs indicate the presence of membrane contact sites (MCS) proteins possibly implicated in lipid exchange. Using dually fluorescence-labeled Dictyostelium discoideum amoeba, we reveal that VAMP-associated protein (Vap) and the PtdIns(4)P 4-phosphatase Sac1 localize to the ER, and Vap also localizes to the LCV membrane. Furthermore, Vap as well as Sac1 promote intracellular replication of L. pneumophila and LCV remodeling. Oxysterol binding proteins (OSBPs) preferentially localize to the ER (OSBP8) or the LCV membrane (OSBP11), respectively, and restrict (OSBP8) or promote (OSBP11) bacterial replication and LCV expansion. The sterol probes GFP-D4H* and filipin indicate that sterols are rapidly depleted from LCVs, while PtdIns(4)P accumulates. In addition to Sac1, the PtdIns(4)P-subverting L. pneumophila effector proteins LepB and SidC also support LCV remodeling. Taken together, the Legionella- and host cell-driven PtdIns(4)P gradient at LCV-ER MCSs promotes Vap-, OSBP- and Sac1-dependent pathogen vacuole maturation.


Assuntos
Dictyostelium , Legionella pneumophila , Legionella , Vacúolos/metabolismo , Legionella/metabolismo , Dictyostelium/microbiologia , Fosfatidilinositóis/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Bactérias/metabolismo
4.
Front Cell Dev Biol ; 9: 731964, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34746129

RESUMO

The haploid social amoeba Dictyostelium discoideum is a powerful model organism to study vesicle trafficking, motility and migration, cell division, developmental processes, and host cell-pathogen interactions. Dynamin superfamily proteins (DSPs) are large GTPases, which promote membrane fission and fusion, as well as membrane-independent cellular processes. Accordingly, DSPs play crucial roles for vesicle biogenesis and transport, organelle homeostasis, cytokinesis and cell-autonomous immunity. Major progress has been made over the last years in elucidating the function and structure of mammalian DSPs. D. discoideum produces at least eight DSPs, which are involved in membrane dynamics and other processes. The function and structure of these large GTPases has not been fully explored, despite the elaborate genetic and cell biological tools available for D. discoideum. In this review, we focus on the current knowledge about mammalian and D. discoideum DSPs, and we advocate the use of the genetically tractable amoeba to further study the role of DSPs in cell and infection biology. Particular emphasis is put on the virulence mechanisms of the facultative intracellular bacterium Legionella pneumophila.

5.
Cell Microbiol ; 23(5): e13318, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33583106

RESUMO

Dictyostelium discoideum Sey1 is the single ortholog of mammalian atlastin 1-3 (ATL1-3), which are large homodimeric GTPases mediating homotypic fusion of endoplasmic reticulum (ER) tubules. In this study, we generated a D. discoideum mutant strain lacking the sey1 gene and found that amoebae deleted for sey1 are enlarged, but grow and develop similarly to the parental strain. The ∆sey1 mutant amoebae showed an altered ER architecture, and the tubular ER network was partially disrupted without any major consequences for other organelles or the architecture of the secretory and endocytic pathways. Macropinocytic and phagocytic functions were preserved; however, the mutant amoebae exhibited cumulative defects in lysosomal enzymes exocytosis, intracellular proteolysis, and cell motility, resulting in impaired growth on bacterial lawns. Moreover, ∆sey1 mutant cells showed a constitutive activation of the unfolded protein response pathway (UPR), but they still readily adapted to moderate levels of ER stress, while unable to cope with prolonged stress. In D. discoideum ∆sey1 the formation of the ER-associated compartment harbouring the bacterial pathogen Legionella pneumophila was also impaired. In the mutant amoebae, the ER was less efficiently recruited to the "Legionella-containing vacuole" (LCV), the expansion of the pathogen vacuole was inhibited at early stages of infection and intracellular bacterial growth was reduced. In summary, our study establishes a role of D. discoideum Sey1 in ER architecture, proteolysis, cell motility and intracellular replication of L. pneumophila.


Assuntos
Dictyostelium/fisiologia , Retículo Endoplasmático/ultraestrutura , GTP Fosfo-Hidrolases/metabolismo , Legionella pneumophila/fisiologia , Proteínas de Protozoários/metabolismo , Vacúolos/microbiologia , Dictyostelium/crescimento & desenvolvimento , Dictyostelium/microbiologia , Dictyostelium/ultraestrutura , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático Rugoso/microbiologia , Retículo Endoplasmático Rugoso/fisiologia , GTP Fosfo-Hidrolases/genética , Homeostase , Interações Hospedeiro-Patógeno , Legionella pneumophila/crescimento & desenvolvimento , Movimento , Muramidase/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Protozoários/genética , Vacúolos/fisiologia
6.
J Biol Chem ; 295(21): 7391-7403, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32303638

RESUMO

The intracellular bacterial pathogen Coxiella burnetii is the etiological agent of the emerging zoonosis Q fever. Crucial to its pathogenesis is type 4b secretion system-mediated secretion of bacterial effectors into host cells that subvert host cell membrane trafficking, leading to the biogenesis of a parasitophorous vacuole for intracellular replication. The characterization of prokaryotic serine/threonine protein kinases in bacterial pathogens is emerging as an important strategy to better understand host-pathogen interactions. In this study, we investigated CstK (for Coxiella Ser/Thr kinase), a protein kinase identified in C. burnetii by in silico analysis. We demonstrate that this putative protein kinase undergoes autophosphorylation on Thr and Tyr residues and phosphorylates a classical eukaryotic protein kinase substrate in vitro This dual Thr-Tyr kinase activity is also observed for a eukaryotic dual-specificity Tyr phosphorylation-regulated kinase class. We found that CstK is translocated during infections and localizes to Coxiella-containing vacuoles (CCVs). Moreover, a CstK-overexpressing C. burnetii strain displayed a severe CCV development phenotype, suggesting that CstK fine-tunes CCV biogenesis during the infection. Protein-protein interaction experiments identified the Rab7 GTPase-activating protein TBC1D5 as a candidate CstK-specific target, suggesting a role for this host GTPase-activating protein in Coxiella infections. Indeed, CstK co-localized with TBC1D5 in noninfected cells, and TBC1D5 was recruited to CCVs in infected cells. Accordingly, TBC1D5 depletion from infected cells significantly affected CCV development. Our results indicate that CstK functions as a bacterial effector protein that interacts with the host protein TBC1D5 during vacuole biogenesis and intracellular replication.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/enzimologia , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Quinases/metabolismo , Febre Q/metabolismo , Vacúolos/metabolismo , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Coxiella burnetii/genética , Proteínas Ativadoras de GTPase/genética , Humanos , Fosforilação , Proteínas Quinases/genética , Febre Q/genética , Vacúolos/genética , Vacúolos/microbiologia
7.
J Biol Chem ; 293(40): 15569-15580, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30131335

RESUMO

Secretion of bacterial signaling proteins and adaptation to the host, especially during infection, are processes that are often linked in pathogenic bacteria. The human pathogen Staphylococcus aureus is equipped with a large arsenal of immune-modulating factors, allowing it to either subvert the host immune response or to create permissive niches for its survival. Recently, we showed that one of the low-molecular-weight protein tyrosine phosphatases produced by S. aureus, PtpA, is secreted during growth. Here, we report that deletion of ptpA in S. aureus affects intramacrophage survival and infectivity. We also observed that PtpA is secreted during macrophage infection. Immunoprecipitation assays identified several host proteins as putative intracellular binding partners for PtpA, including coronin-1A, a cytoskeleton-associated protein that is implicated in a variety of cellular processes. Of note, we demonstrated that coronin-1A is phosphorylated on tyrosine residues upon S. aureus infection and that its phosphorylation profile is linked to PtpA expression. Our results confirm that PtpA has a critical role during infection as a bacterial effector protein that counteracts host defenses.


Assuntos
Proteínas de Bactérias/genética , Interações Hospedeiro-Patógeno , Proteínas dos Microfilamentos/genética , Proteínas Tirosina Fosfatases/genética , Infecções Estafilocócicas/genética , Staphylococcus aureus/genética , Animais , Proteínas de Bactérias/metabolismo , Clonagem Molecular , Dictyostelium/genética , Dictyostelium/metabolismo , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Fosforilação , Ligação Proteica , Proteínas Tirosina Fosfatases/metabolismo , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/enzimologia , Staphylococcus aureus/patogenicidade , Tirosina/metabolismo , Virulência
8.
Nat Commun ; 8(1): 1543, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29146912

RESUMO

Legionella pneumophila can cause Legionnaires' disease and replicates intracellularly in a distinct Legionella-containing vacuole (LCV). LCV formation is a complex process that involves a plethora of type IV-secreted effector proteins. The effector RidL binds the Vps29 retromer subunit, blocks retrograde vesicle trafficking, and promotes intracellular bacterial replication. Here, we reveal that the 29-kDa N-terminal domain of RidL (RidL2-281) adopts a "foot-like" fold comprising a protruding ß-hairpin at its "heel". The deletion of the ß-hairpin, the exchange to Glu of Ile170 in the ß-hairpin, or Leu152 in Vps29 abolishes the interaction in eukaryotic cells and in vitro. RidL2-281 or RidL displace the Rab7 GTPase-activating protein (GAP) TBC1D5 from the retromer and LCVs, respectively, and TBC1D5 promotes the intracellular growth of L. pneumophila. Thus, the hydrophobic ß-hairpin of RidL is critical for binding of the L. pneumophila effector to the Vps29 retromer subunit and displacement of the regulator TBC1D5.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Legionella pneumophila/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Proteínas de Bactérias/química , Dictyostelium , Proteínas Ativadoras de GTPase/química , Células HeLa , Humanos , Legionella pneumophila/fisiologia , Camundongos , Microscopia Confocal , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Células RAW 264.7 , Vacúolos/metabolismo , Vacúolos/microbiologia , Proteínas de Transporte Vesicular/química
9.
J Cell Sci ; 129(12): 2354-67, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27170354

RESUMO

Proteins that contain Eps15 homology domains (EHDs) in their C-terminus are newly identified key regulators of endosomal membrane trafficking. Here, we show that D. discoideum contains a single EHD protein (referred to as EHD) that localizes to endosomal compartments and newly formed phagosomes. We provide the first evidence that EHD regulates phagosome maturation. Deletion of EHD results in defects in intraphagosomal proteolysis and acidification. These defects are linked to early delivery of lysosomal enzymes and fast retrieval of the vacuolar H(+)-ATPase in maturing phagosomes. We also demonstrate that EHD physically interacts with DymA. Our results indicate that EHD and DymA can associate independently with endomembranes, and yet they share identical kinetics in recruitment to phagosomes and release during phagosome maturation. Functional analysis of ehd(-), dymA(-) and double dymA(-)ehd(-) knockout strains indicate that DymA and EHD play non-redundant and independent functions in phagosome maturation. Finally, we show that the absence of EHD leads to increased tubulation of endosomes, indicating that EHD participates in the scission of endosomal tubules, as reported for DymA.


Assuntos
Dictyostelium/metabolismo , Dinaminas/metabolismo , Fagossomos/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Endossomos/metabolismo , Concentração de Íons de Hidrogênio , Mutação/genética , Ligação Proteica , Proteólise , Proteínas de Protozoários/química , Imagem com Lapso de Tempo
10.
J Cell Sci ; 127(Pt 21): 4702-13, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25189617

RESUMO

Dictyostelium discoideum ACAP-A is an Arf GTPase-activating protein (GAP) involved in cytokinesis, cell migration and actin cytoskeleton dynamics. In mammalian cells, ACAP family members regulate endocytic protein trafficking. Here, we explored the function of ACAP-A in the endocytic pathway of D. discoideum. In the absence of ACAP-A, the efficiency of fusion between post-lysosomes and the plasma membrane was reduced, resulting in the accumulation of post-lysosomes. Moreover, internalized fluid-phase markers showed extended intracellular transit times, and the transfer kinetics of phagocyted particles from lysosomes to post-lysosomes was reduced. Neutralization of lysosomal pH, one essential step in lysosome maturation, was also delayed. Whereas expression of ACAP-A-GFP in acapA(-) cells restored normal particle transport kinetics, a mutant ACAP-A protein with no GAP activity towards the small GTPase ArfA failed to complement this defect. Taken together, these data support a role for ACAP-A in maturation of lysosomes into post-lysosomes through an ArfA-dependent mechanism. In addition, we reveal that ACAP-A is required for efficient intracellular growth of Legionella pneumophila, a pathogen known to subvert the endocytic host cell machinery for replication. This further emphasizes the role of ACAP-A in the endocytic pathway.


Assuntos
Dictyostelium/metabolismo , Dictyostelium/microbiologia , Legionella pneumophila/fisiologia , Lisossomos/metabolismo , Interações Hospedeiro-Patógeno
11.
J Cell Sci ; 126(Pt 3): 756-66, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264736

RESUMO

ACAPs and ASAPs are Arf-GTPase-activating proteins with BAR, PH, GAP and ankyrin repeat domains and are known to regulate vesicular traffic and actin cytoskeleton dynamics in mammalian cells. The amoeba Dictyostelium has only two proteins with this domain organization, instead of the six in human, enabling a more precise functional analysis. Genetic invalidation of acapA resulted in multinucleated cells with cytokinesis defects. Mutant acapA(-) cells were hardly motile and their multicellular development was significantly delayed. In addition, formation of filopodial protrusions was deficient in these cells. Conversely, re-expression of ACAP-A-GFP resulted in numerous and long filopodia-like protrusions. Mutagenesis studies showed that the ACAP-A actin remodeling function was dependent on its ability to activate its substrate, the small GTPase ArfA. Likewise, the expression of a constitutively active ArfA•GTP mutant in wild-type cells led to a significant reduction in filopodia length. Together, our data support a role for ACAP-A in the control of the actin cytoskeleton organization and dynamics through an ArfA-dependent mechanism.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Dictyostelium/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Pseudópodes/metabolismo , Animais , Movimento Celular/genética , Citocinese/genética , Dictyostelium/enzimologia , Proteínas Ativadoras de GTPase/genética , Humanos , Mutação/genética , Pseudópodes/patologia , RNA Interferente Pequeno/genética , Transgenes/genética
12.
ScientificWorldJournal ; 9: 629-32, 2009 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-19618090

RESUMO

Syndecans are transmembrane proteoglycan receptors that interact with a wide variety of extracellular molecules such as adhesion receptors and extracellular matrix components. There are four syndecans in mammals, which are expressed in a development-, cell-type-, and tissue-specific manner, and function either as coreceptors that cooperate with other cell surface receptors or as cell adhesion receptors that independently mediate cell signaling. Cell signaling is supported through their short cytoplasmic tail that contains four tyrosine residues, which are conserved among all syndecan family members. In this commentary, we report and discuss data showing that the receptor syndecan-1 interacts with the carboxy-terminal LG4/5 domain in laminin-332 to participate in cell adhesion and spreading. Remarkably, cell adhesion to LG4/5 is associated with a rapid dephosphorylation of tyrosine in syndecan-1. These results unveil for the first time that one "turn on" signal for syndecan-1 upon LG4/5 recognition may not be phosphorylation, but tyrosine dephosphorylation, an unexpected outcome. How this regulatory event may take place and which tyrosine residues are concerned are questions tackled in this report.


Assuntos
Sindecana-1/metabolismo , Tirosina/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Fosforilação , Ligação Proteica , Transdução de Sinais , Calinina
13.
J Biol Chem ; 284(16): 10659-71, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19228696

RESUMO

Heparan sulfate proteoglycan receptor syndecan-1 interacts with the carboxyl-terminal LG4/5 domain in laminin 332 (alpha3LG4/5) and participates in cell adhesion and spreading. To dissect the function of syndecan-1 in these processes, we made use of a cell adhesion model in which syndecan-1 exclusively interacts with a recombinantly expressed alpha3LG4/5 fragment. Plating HT1080 cells on this fragment induces the formation of actin-containing protrusive structures in an integrin-independent manner. Here we show that syndecan-1-mediated formation of membrane protrusions requires dephosphorylation of tyrosine residues in syndecan-1. Accordingly, inhibition of phosphatases with orthovanadate decreases cell adhesion to the alpha3LG4/5 fragment. We demonstrate that the PDZ-containing protein syntenin-1, known to connect cytoskeletal proteins, binds to syndecan-1 in cells plated on the alpha3LG4/5 fragment and participates in the formation of membrane protrusions. We further show that syntenin-1 recruitment depends on the dephosphorylation of Tyr-309 located within syndecan-1 PDZ binding domain EFYA. We propose that tyrosine dephosphorylation of syndecan-1 may regulate its association with cytoskeleton components.


Assuntos
Sindecana-1/metabolismo , Sinteninas/metabolismo , Tirosina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Extensões da Superfície Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Sindecana-1/genética , Sinteninas/genética
14.
Traffic ; 10(2): 161-71, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19054384

RESUMO

Sorting of ubiquitinated proteins to multivesicular bodies (MVBs) in mammalian cells relies on proteins with a Vps27/Hrs/STAM (VHS) domain. Here, we show that the amoeba Dictyostelium presents only one protein with a VHS domain: DdTom1. We demonstrate that the VHS domain of DdTom1 is followed by a Golgi-localized, gamma-ear-containing, ADP-ribosylation-factor-binding and Tom1 (GAT) domain that binds ubiquitin, and by a non-conserved C-terminal domain that can recruit clathrin, EGFr pathway substrate 15 and tumor susceptibility gene 101, a component of the MVB biogenesis machinery [endosomal complexes required for transport (ESCRT) complexes]. Both VHS and GAT domains interact with phospholipids and therefore could ensure the recruitment of DdTom1 to endosomal membranes. We propose that DdTom1 participates in an ancestral ESCRT-0 complex implicated in the sorting of ubiquitinated proteins into MVBs.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Dictyostelium/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Motivos de Aminoácidos , Animais , Clatrina/metabolismo , Citoplasma/metabolismo , Dictyostelium/genética , Dictyostelium/ultraestrutura , Deleção de Genes , Humanos , Lisossomos/metabolismo , Microscopia Eletrônica , Ligação Proteica , Proteínas de Protozoários/genética
15.
Exp Cell Res ; 314(15): 2822-33, 2008 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-18634783

RESUMO

Soluble N-ethylmaleimide-sensitive-factor Attachment protein Receptors (SNAREs) participate in the specificity of membrane fusions in the cell. The mechanisms of specific SNARE sorting are still however poorly documented. We investigated the possible role of Adaptor Protein (AP) complexes in sorting of the Dictyostelium discoideum v-SNARE VAMP7. In live cells, GFP-VAMP7 is observed in the membrane of endocytic compartments. It is also observed in the plasma membrane of a small proportion of the cells. Mutation of a potential dileucine motif dramatically increases the proportion of cells with GFP-VAMP7 in their plasma membrane, strongly supporting the participation of an AP complex in VAMP7 sorting to the endocytic pathway. A partial increase occurs in knockout cells for the medium subunits of AP-2 and AP-3 complexes, indicating a role for both AP-2 and AP-3. VAMP7, as well as its t-SNAREs partners syntaxin 8 and Vti1, are co-immunoprecipitated with each of the medium subunits of the AP-1, AP-2, AP-3 and AP-4 complexes. This result supports the conclusion that VAMP7 directly interacts with both AP-2 and AP-3. It also raises the hypothesis of an interaction with AP-1 and AP-4. GFP-VAMP7 is retrieved from the endocytic pathway at and/or before the late post-lysosomal stage through an AP-independent mechanism.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Dictyostelium/metabolismo , Endocitose/fisiologia , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Vesículas Transportadoras/metabolismo , Complexo 2 de Proteínas Adaptadoras/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Proteínas de Fluorescência Verde/genética , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Substâncias Macromoleculares/metabolismo , Transporte Proteico/fisiologia , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Vesículas Transportadoras/ultraestrutura
16.
J Biol Chem ; 283(11): 6764-72, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18178563

RESUMO

TM9 proteins constitute a well defined family, characterized by the presence of a large variable extracellular domain and nine putative transmembrane domains. This family is highly conserved throughout evolution and comprises three members in Dictyostelium discoideum and Saccharomyces cerevisiae and four in humans and mice. In Dictyostelium, previous analysis demonstrated that TM9 proteins are implicated in cellular adhesion. In this study, we generated TM9 mutants in S. cerevisiae and analyzed their phenotype with particular attention to cellular adhesion. S. cerevisiae strains lacking any one of the three TM9 proteins were severely suppressed for adhesive growth and filamentous growth under conditions of nitrogen starvation. In these mutants, expression of the FLO11-lacZ reporter gene was strongly reduced, whereas expression of FRE(Ty1)-lacZ was not, suggesting that TM9 proteins are implicated at a late stage of nutrient-controlled signaling pathways. We also reexamined the phenotype of Dictyostelium TM9 mutant cells, focusing on nutrient-controlled cellular functions. Although the initiation of multicellular development and autophagy was unaltered in Dictyostelium TM9 mutants, nutrient-controlled secretion of lysosomal enzymes was dysregulated in these cells. These results suggest that in both yeast and amoebae, TM9 proteins participate in the control of specific cellular functions in response to changing nutrient conditions.


Assuntos
Dictyostelium/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/fisiologia , Regulação Fúngica da Expressão Gênica , Regulação da Expressão Gênica , Proteínas de Membrana/fisiologia , Animais , Autofagia , Adesão Celular , Proteínas Fúngicas/genética , Lisossomos/metabolismo , Proteínas de Membrana/genética , Modelos Biológicos , Mutação , Plasmídeos/metabolismo , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Especificidade da Espécie
17.
J Cell Physiol ; 214(1): 238-49, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17579341

RESUMO

Laminin 5/laminin 332 (LN332) is an adhesion substrate for epithelial cells. After secretion of LN332, a regulated cleavage occurs at the carboxy-terminus of its alpha3 subunit, which releases a tandem of two globular modules named LG4/5. We show that the presence of the LG4/5 domain in precursor LN332 decreases its integrin-mediated cell adhesion properties in comparison with mature LN332. Whereas cell adhesion to the recombinant LG4/5 fragment relies solely on the heparan sulfate proteoglycan (HSPG) receptor syndecan-1, we reveal that both syndecan-1 and the alpha3beta1 integrin bind to precursor LN332. We further demonstrate that syndecan-1 mediated cell adhesion to the LG4/5 fragment and pre-LN332 allows the formation of fascin-containing protrusions, depending on the GTPases Rac and Cdc42 activation. Reducing syndecan-1 expression in normal keratinocytes prevents cell protrusions on pre-LN332 with subsequent failure of the peripheral localization of the alpha3beta1 integrin. We finally show that cell migration on pre-LN332 requires syndecan-1. Therefore, the LG4/5 domain in precursor LN332 appears to trigger intracellular signaling events, which participate in keratinocyte motility.


Assuntos
Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Queratinócitos/fisiologia , Sindecana-1/metabolismo , Adesão Celular , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/isolamento & purificação , Células Cultivadas , Ativação Enzimática , Fibrossarcoma/patologia , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Humanos , Integrina alfa3beta1/metabolismo , Masculino , Melanoma/patologia , Microscopia de Vídeo , Faloidina , Plasmídeos , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Rodaminas , Pele/citologia , Transfecção , Proteína cdc42 de Ligação ao GTP/análise , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/análise , Proteínas rac de Ligação ao GTP/metabolismo , Calinina
18.
Traffic ; 7(11): 1528-38, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17010123

RESUMO

Dictyostelium discoideum cells continuously internalize extracellular material, which accumulates in well-characterized endocytic vacuoles. In this study, we describe a new endocytic compartment identified by the presence of a specific marker, the p25 protein. This compartment presents features reminiscent of mammalian recycling endosomes: it is localized in the pericentrosomal region but distinct from the Golgi apparatus. It specifically contains surface proteins that are continuously endocytosed but rapidly recycled to the cell surface and thus absent from maturing endocytic compartments. We evaluated the importance of each clathrin-associated adaptor complex in establishing a compartmentalized endocytic system by studying the phenotype of the corresponding mutants. In knockout cells for mu3, a subunit of the AP-3 clathrin-associated complex, membrane proteins normally restricted to p25-positive endosomes were mislocalized to late endocytic compartments. Our results suggest that AP-3 plays an essential role in the compartmentalization of the endocytic pathway in Dictyostelium.


Assuntos
Complexo 3 de Proteínas Adaptadoras/fisiologia , Dictyostelium/fisiologia , Endocitose/fisiologia , Subunidades mu do Complexo de Proteínas Adaptadoras/deficiência , Subunidades mu do Complexo de Proteínas Adaptadoras/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Dictyostelium/genética , Endossomos/metabolismo , Deleção de Genes , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Modelos Biológicos , Transporte Proteico/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
19.
Mol Biol Cell ; 17(12): 4982-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16987957

RESUMO

Dictyostelium amoebae grow as single cells but upon starvation they initiate multicellular development. Phg2 was characterized previously as a kinase controlling cellular adhesion and the organization of the actin cytoskeleton. Here we report that Phg2 also plays a role during the transition between growth and multicellular development, as evidenced by the fact that phg2 mutant cells can initiate development even in the presence of nutrients. Even at low cell density and in rich medium, phg2 mutant cells express discoidin, one of the earliest predevelopmental markers. Complementation studies indicate that, in addition to the kinase domain, the core region of Phg2 is involved in the initiation of development. In this region, a small domain contiguous with a previously described ras-binding domain was found to interact with the Dictyostelium ortholog of the mammalian adhesion-regulating molecule (ADRM1). In addition, adrm1 knockout cells also exhibit abnormal initiation of development. These results suggest that a Phg2-Adrm1 signaling pathway is involved in the control of the transition from growth to differentiation in Dictyostelium. Phg2 thus plays a dual role in the control of cellular adhesion and initiation of development.


Assuntos
Dictyostelium/crescimento & desenvolvimento , Alimentos , Glicoproteínas de Membrana/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Dictyostelium/citologia , Privação de Alimentos/fisiologia , Mutação/genética , Ligação Proteica , Estrutura Terciária de Proteína
20.
J Cell Sci ; 119(Pt 19): 4079-87, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16968738

RESUMO

Specialized eukaryotic cells can ingest large particles and sequester them within membrane-delimited phagosomes. Many studies have described the delivery of lysosomal proteins to the phagosome, but little is known about membrane sorting during the early stages of phagosome formation. Here we used Dictyostelium discoideum amoebae to analyze the membrane composition of newly formed phagosomes. The membrane delimiting the closing phagocytic cup was essentially derived from the plasma membrane, but a subgroup of proteins was specifically excluded. Interestingly the same phenomenon was observed during the formation of macropinosomes, suggesting that the same sorting mechanisms are at play during phagocytosis and macropinocytosis. Analysis of mutant strains revealed that clathrin-associated adaptor complexes AP-1, -2 and -3 were not necessary for this selective exclusion and, accordingly, ultrastructural analysis revealed no evidence for vesicular transport around phagocytic cups. Our results suggest the existence of a new, as yet uncharacterized, sorting mechanism in phagocytic and macropinocytic cups.


Assuntos
Membrana Celular/fisiologia , Fagocitose/fisiologia , Pinocitose/fisiologia , Proteínas Adaptadoras de Transporte Vesicular , Animais , Transporte Biológico , Membrana Celular/química , Células Cultivadas , Dictyostelium/fisiologia , Membranas Intracelulares/fisiologia , Modelos Biológicos , Miosinas/fisiologia , Fagossomos/química , Fagossomos/fisiologia , Transporte Proteico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...