Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Life Sci Alliance ; 5(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35086936

RESUMO

Unlike constitutively secreted proteins, peptide hormones are stored in densely packed secretory granules, before regulated release upon stimulation. Secretory granules are formed at the TGN by self-aggregation of prohormones as functional amyloids. The nonapeptide hormone vasopressin, which forms a small disulfide loop, was shown to be responsible for granule formation of its precursor in the TGN as well as for toxic fibrillar aggregation of unfolded mutants in the ER. Several other hormone precursors also contain similar small disulfide loops suggesting their function as a general device to mediate aggregation for granule sorting. To test this hypothesis, we studied the capacity of small disulfide loops of different hormone precursors to mediate aggregation in the ER and the TGN. They indeed induced ER aggregation in Neuro-2a and COS-1 cells. Fused to a constitutively secreted reporter protein, they also promoted sorting into secretory granules, enhanced stimulated secretion, and increased Lubrol insolubility in AtT20 cells. These results support the hypothesis that small disulfide loops act as novel signals for sorting into secretory granules by self-aggregation.


Assuntos
Hormônios Peptídicos/metabolismo , Vesículas Secretórias/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Grânulos Citoplasmáticos/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Complexo de Golgi/metabolismo , Hormônios/genética , Hormônios/metabolismo , Humanos , Hormônios Peptídicos/genética , Transporte Proteico , Vesículas Secretórias/fisiologia , Vasopressinas/metabolismo , Rede trans-Golgi/metabolismo
2.
Cancer Sci ; 112(10): 3979-3994, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34252266

RESUMO

Pyroptosis refers to the process of gasdermin (GSDM)-mediated programmed cell death (PCD). Our understanding of pyroptosis has expanded beyond cells and is known to involve extracellular responses. Recently, there has been an increasing interest in pyroptosis due to its emerging role in activating the immune system. In the meantime, pyroptosis-mediated therapies, which use the immune response to kill cancer cells, have also achieved notable success in a clinical setting. In this review, we discuss that the immune response induced by pyroptosis activation is a double-edged sword that affects all stages of tumorigenesis. On the one hand, the activation of inflammasome-mediated pyroptosis and the release of pyroptosis-produced cytokines alter the immune microenvironment and promote the development of tumors by evading immune surveillance. On the other hand, pyroptosis-produced cytokines can also collect immune cells and ignite the immune system to improve the efficiency of tumor immunotherapies. Pyroptosis is also related to some immune checkpoints, especially programmed death-1 (PD-1) or programmed death- ligand 1 (PD-L1). In this review, we mainly focus on our current understanding of the interplay between the immune system and tumors that process through pyroptosis, and debate their use as potential therapeutic targets.


Assuntos
Sistema Imunitário/imunologia , Neoplasias/imunologia , Piroptose/fisiologia , Microambiente Tumoral/imunologia , Antígeno B7-H1/imunologia , Citocinas/metabolismo , Progressão da Doença , Humanos , Evasão da Resposta Imune , Imunoterapia , Inflamassomos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Neoplasias/etiologia , Neoplasias/prevenção & controle , Neoplasias/terapia , Receptor de Morte Celular Programada 1/imunologia , Piroptose/imunologia , Vesículas Secretórias/fisiologia , Evasão Tumoral/imunologia
3.
Mol Biol Cell ; 32(10): 1064-1080, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33788598

RESUMO

Proper Golgi complex function depends on the activity of Arf1, a GTPase whose effectors assemble and transport outgoing vesicles. Phosphatidylinositol 4-phosphate (PI4P) generated at the Golgi by the conserved PI 4-kinase Pik1 (PI4KIIIß) is also essential for Golgi function, although its precise roles in vesicle formation are less clear. Arf1 has been reported to regulate PI4P production, but whether Pik1 is a direct Arf1 effector is not established. Using a combination of live-cell time-lapse imaging analyses, acute PI4P depletion experiments, and in vitro protein-protein interaction assays on Golgi-mimetic membranes, we present evidence for a model in which Arf1 initiates the final stages of Golgi maturation by tightly controlling PI4P production through direct recruitment of the Pik1-Frq1 PI4-kinase complex. This PI4P serves as a critical signal for AP-1 and secretory vesicle formation, the final events at maturing Golgi compartments. This work therefore establishes the regulatory and temporal context surrounding Golgi PI4P production and its precise roles in Golgi maturation.


Assuntos
Complexo de Golgi/fisiologia , Fosfatos de Fosfatidilinositol/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , 1-Fosfatidilinositol 4-Quinase/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Ligação ao Cálcio/fisiologia , Diglicerídeos/metabolismo , Ligação Proteica , Saccharomycetales , Vesículas Secretórias/fisiologia , Imagem com Lapso de Tempo
4.
Exp Cell Res ; 399(1): 112449, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33347856

RESUMO

Delayed endothelial healing after drug eluting stent (DES) implantation is a critical clinical problem in treatment of coronary artery diseases. Exosomes exhibit proangiogenic potential in a variety of ischemic diseases. However, the association of exosomes with endothelial regeneration after DES implantation has been rarely reported. In this study, we aimed to investigate the therapeutic effects of mesenchymal stem cell (MSC)-derived exosomes on endothelial cells treated with rapamycin and explore the potential mechanisms of MSC-derived exosomes in promoting endothelial regeneration. Exosomes were isolated from MSCs by ultracentrifugation and identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blot assay. The in vitro effects of MSC-derived exosomes on the proliferation and migration of endothelial cells treated with rapamycin were evaluated by integrated experiment, cell counting kit-8, scratch, tube formation, and transwell assays. And the apoptosis of rapamycin-induced endothelial cells loaded with MSC-derived exosomes was detected using TUNEL and Annexin-V FITC and PI double-staining assays. The microRNA (miRNA) cargo of MSC-derived exosomes was identified by high-throughput RNA sequencing. Pro-angiogenic miRNAs and key pathways were further characterized. Our results indicated that MSC-derived exosomes could be ingested into umbilical vein endothelial cells (HUVECs) and significantly enhanced cell proliferation rate, migratory and tube-forming capabilities in vitro. MSC-derived exosomes also inhibited the apoptosis of HUVECs induced by rapamycin. A distinct class of exosomal miRNAs was further identified, including six miRNAs tightly related to neovasculogenesis. Silencing the expression of exosomal miRNA-21-5p and let-7c-5p attenuated the pro-proliferative and pro-migratory capacity of MSC-derived exosomes. Moreover, functional enrichment analysis indicated that metabolic pathways might contribute to reendothelialization. This study highlights a proregenerative effect of MSC-derived exosomes in vitro, which may be partly explained by the delivery of pro-angiogenic miRNAs to endothelial cells.


Assuntos
Exossomos/fisiologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/metabolismo , Sirolimo/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/genética , Regeneração/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Vesículas Secretórias/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Cicatrização/efeitos dos fármacos
5.
Plant J ; 104(6): 1685-1697, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33067901

RESUMO

F-actin and myosin XI play important roles in plant organelle movement. A few myosin XI genes in the genome of Arabidopsis are mainly expressed in mature pollen, which suggests that they may play a crucial role in pollen germination and pollen tube tip growth. In this study, a genetic complementation assay was conducted in a myosin xi-c (myo11c1) myosin xi-e (myo11c2) double mutant, and fluorescence labeling combined with microscopic observation was applied. We found that myosin XI-E (Myo11C2)-green fluorescent protein (GFP) restored the slow pollen tube growth and seed deficiency phenotypes of the myo11c1 myo11c2 double mutant and Myo11C2-GFP partially colocalized with mitochondria, peroxisomes and Golgi stacks. Furthermore, decreased mitochondrial movement and subapical accumulation were detected in myo11c1 myo11c2 double mutant pollen tubes. Fluorescence recovery after photobleaching experiments showed that the fluorescence recoveries of GFP-RabA4d and AtPRK1-GFP at the pollen tube tip of the myo11c1 myo11c2 double mutant were lower than those of the wild type were after photobleaching. These results suggest that Myo11C2 may be associated with mitochondria, peroxisomes and Golgi stacks, and play a crucial role in organelle movement and apical accumulation of secretory vesicles in pollen tubes of Arabidopsis thaliana.


Assuntos
Proteínas de Arabidopsis/fisiologia , Arabidopsis/fisiologia , Miosinas/fisiologia , Organelas/fisiologia , Tubo Polínico/fisiologia , Vesículas Secretórias/fisiologia , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Complexo de Golgi/metabolismo , Mitocôndrias/metabolismo , Miosinas/metabolismo , Organelas/metabolismo , Peroxissomos/metabolismo , Tubo Polínico/crescimento & desenvolvimento , Tubo Polínico/metabolismo , Vesículas Secretórias/metabolismo
6.
Cell Rep ; 30(11): 3632-3643.e8, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32187536

RESUMO

Although similar in molecular composition, synapses can exhibit strikingly distinct functional transmitter release and plasticity characteristics. To determine whether ultrastructural differences co-define this functional heterogeneity, we combine hippocampal organotypic slice cultures, high-pressure freezing, freeze substitution, and 3D-electron tomography to compare two functionally distinct synapses: hippocampal Schaffer collateral and mossy fiber synapses. We find that mossy fiber synapses, which exhibit a lower release probability and stronger short-term facilitation than Schaffer collateral synapses, harbor lower numbers of docked synaptic vesicles at active zones and a second pool of possibly tethered vesicles in their vicinity. Our data indicate that differences in the ratio of docked versus tethered vesicles at active zones contribute to distinct functional characteristics of synapses.


Assuntos
Hipocampo/fisiologia , Hipocampo/ultraestrutura , Terminações Pré-Sinápticas/fisiologia , Terminações Pré-Sinápticas/ultraestrutura , Sinapses/fisiologia , Sinapses/ultraestrutura , Animais , AMP Cíclico/metabolismo , Potenciais Pós-Sinápticos Excitadores , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musgosas Hipocampais/fisiologia , Fibras Musgosas Hipocampais/ultraestrutura , Neurotransmissores/metabolismo , Técnicas de Cultura de Órgãos , Vesículas Secretórias/fisiologia , Vesículas Secretórias/ultraestrutura , Vesículas Sinápticas/ultraestrutura , Fixação de Tecidos
7.
Fungal Genet Biol ; 135: 103286, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31672687

RESUMO

The fungal cell wall consists of proteins and polysaccharides, formed by the co-ordinated activity of enzymes, such as chitin or glucan synthases. These enzymes are delivered via secretory vesicles to the hyphal tip. In the ascomycete Neurospora crassa, chitin synthases and ß(1,3)-glucan synthase are transported in different vesicles, whereas they co-travel along microtubules in the basidiomycete Ustilago maydis. This suggests fundamental differences in wall synthesis between taxa. Here, we visualize the class V chitin synthase ZtChs5 and the ß(1,3)-glucan synthase ZtGcs1 in the ascomycete Zymoseptoria tritici. Live cell imaging demonstrate that both enzymes co-locate to the apical plasma membrane, but are not concentrated in the Spitzenkörper. Delivery involves co-transport along microtubules of the chitin and glucan synthase. Live cell imaging and electron microscopy suggest that both cell wall synthases locate in the same vesicle. Thus, microtubule-dependent co-delivery of cell wall synthases in the same vesicle is found in asco- and basidiomycetes.


Assuntos
Ascomicetos/enzimologia , Quitina Sintase/metabolismo , Glucosiltransferases/metabolismo , Vesículas Secretórias/fisiologia , Ascomicetos/genética , Basidiomycota/metabolismo , Quitina Sintase/genética , Citoplasma/metabolismo , Citoplasma/ultraestrutura , Glucosiltransferases/genética , Proteínas de Fluorescência Verde/metabolismo , Microscopia Eletrônica , Neurospora crassa/metabolismo , Vesículas Secretórias/ultraestrutura
8.
Ann Am Thorac Soc ; 15(Suppl 3): S159-S163, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30431338

RESUMO

The respiratory system is protected from inhaled particles and microbes by the mucociliary system. This system differs between animal species, where pigs and humans have numerous submucosal glands. The polymer-forming mucin, MUC5B, is packed in a highly organized way in granules of the mucus-secreting cells in the glands. Upon secretion, the packed MUC5B is flushed out by a chloride- and bicarbonate-rich fluid from the cystic fibrosis transmembrane conductance regulator-expressing serosal cells located at the most distal part of the gland. The bicarbonate raises the pH and removes calcium from the N terminus of MUC5B, allowing the mucin to be pulled out into a linear polymer. Thousands of such polymers gather in bundles in the submucosal gland duct, and these bundles appear at the opening of the glands. They are moved by the beating cilia, and sweep over the airway surface and are patchily coated with the MUC5AC mucin from the surface goblet cells. The movement of these bundles is controlled by the MUC5AC mucin attachment/detachment to the goblet cells. Thus, higher animals with submucosal glands and large diameters of the proximal airways are efficiently cleaned by the thick mucus bundles sweeping the airway surface and moving particles and bacteria toward the larynx.


Assuntos
Pneumopatias/etiologia , Mucinas/fisiologia , Animais , Modelos Animais de Doenças , Humanos , Pneumopatias/diagnóstico , Pneumopatias/terapia , Depuração Mucociliar/fisiologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Mucosa Respiratória/fisiopatologia , Vesículas Secretórias/fisiologia , Suínos
9.
Ann Am Thorac Soc ; 15(Suppl 3): S154-S158, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30431345

RESUMO

Mucociliary clearance is a crucial component of innate defense of the lung. In respiratory diseases, such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis, mucus with abnormal properties contributes to obstruction of the airways. The failure in function of mucus in airway clearance and pathogen protection leads to chronic infection and risk of death. Polymeric mucins (MUC5AC and MUC5B) provide the structural framework of the airway mucus gel. The intracellular synthesis and assembly of these enormous, polymeric O-linked glycoproteins is a complex, multistage process involving intra- and intermolecular disulfide bond formation and extensive addition of O-glycan chains. The fully formed polymers are packaged in a highly organized and condensed form within secretory granules inside specialized secretory cells, and after the appropriate stimulus, mucins are released and expand to form mucus. This short article brings together the current knowledge on the different steps in the production of mucin polymers and the molecular mechanisms that condense them into a packaged form in secretory granules. It is by unraveling the molecular mechanisms that control intracellular mucin supramolecular structure that we might gain new insight into what determines mucus gel properties in health and disease.


Assuntos
Pneumopatias/etiologia , Mucinas/metabolismo , Humanos , Pneumopatias/diagnóstico , Pneumopatias/terapia , Depuração Mucociliar , Vesículas Secretórias/fisiologia
10.
Biochem Soc Trans ; 46(5): 1021-1027, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30154095

RESUMO

As part of their life cycle, Gram-negative bacteria produce and release microvesicles (outer membrane vesicles, OMVs) consisting of spherical protrusions of the outer membrane that encapsulate periplasmic contents. OMVs produced by commensal bacteria in the gastrointestinal (GI) tract of animals are dispersed within the gut lumen with their cargo and enzymes being distributed across and throughout the GI tract. Their ultimate destination and fate is unclear although they can interact with and cross the intestinal epithelium using different entry pathways and access underlying immune cells in the lamina propria. OMVs have also been found in the bloodstream from which they can access various tissues and possibly the brain. The nanosize and non-replicative status of OMVs together with their resistance to enzyme degradation and low pH, alongside their ability to interact with the host, make them ideal candidates for delivering biologics to mucosal sites, such as the GI and the respiratory tract. In this mini-review, we discuss the fate of OMVs produced in the GI tract of animals with a focus on vesicles released by Bacteroides species and the use of OMVs as vaccine delivery vehicles and other potential applications.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Microbioma Gastrointestinal , Bactérias Gram-Negativas/fisiologia , Vesículas Secretórias/fisiologia , Animais , Bacteroides , Encéfalo/microbiologia , Células Epiteliais/microbiologia , Trato Gastrointestinal/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Sistema Imunitário , Mucosa Intestinal/microbiologia
11.
Artigo em Inglês | MEDLINE | ID: mdl-29496823

RESUMO

The delivery of intracellular material within cells is crucial for maintaining normal function. Myosins transport a wide variety of cargo, ranging from vesicles to ribonuclear protein particles (RNPs), in plants, fungi, and metazoa. The properties of a given myosin transporter are adapted to move on different actin filament tracks, either on the disordered actin networks at the cell cortex or along highly organized actin bundles to distribute their cargo in a localized manner or move it across long distances in the cell. Transport is controlled by selective recruitment of the myosin to its cargo that also plays a role in activation of the motor.


Assuntos
Miosinas/metabolismo , Animais , Transporte Biológico , Corrente Citoplasmática , Humanos , Organelas/fisiologia , Plantas/metabolismo , RNA/metabolismo , Ribonucleoproteínas/metabolismo , Vesículas Secretórias/fisiologia , Vesículas Transportadoras/fisiologia
12.
Gastroenterology ; 154(6): 1805-1821.e5, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29360461

RESUMO

BACKGROUND & AIMS: Pancreatic acinar cells are polarized epithelial cells that store enzymes required for digestion as inactive zymogens, tightly packed at the cell apex. Stimulation of acinar cells causes the zymogen granules to fuse with the apical membrane, and the cells undergo exocytosis to release proteases into the intestinal lumen. Autophagy maintains homeostasis of pancreatic acini. Syntaxin 2 (STX2), an abundant soluble N-ethyl maleimide sensitive factor attachment protein receptor in pancreatic acini, has been reported to mediate apical exocytosis. Using human pancreatic tissues and STX2-knockout (KO) mice, we investigated the functions of STX2 in zymogen granule-mediated exocytosis and autophagy. METHODS: We obtained pancreatic tissues from 5 patients undergoing surgery for pancreatic cancer and prepared 80-µm slices; tissues were exposed to supramaximal cholecystokinin octapeptide (CCK-8) or ethanol and a low concentration of CCK-8 and analyzed by immunoblot and immunofluorescence analyses. STX2-KO mice and syntaxin 2+/+ C57BL6 mice (controls) were given intraperitoneal injections of supramaximal caerulein (a CCK-8 analogue) or fed ethanol and then given a low dose of caerulein to induce acute pancreatitis, or saline (controls); pancreata were isolated and analyzed by histology and immunohistochemistry. Acini were isolated from mice, incubated with CCK-8, and analyzed by immunofluorescence microscopy or used in immunoprecipitation experiments. Exocytosis was quantified using live-cell exocytosis and Ca2+ imaging analyses and based on formation of exocytotic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes. Dysregulations in autophagy were identified using markers, electron and immunofluorescence microscopy, and protease activation assays. RESULTS: Human pancreatic tissues and dispersed pancreatic acini from control mice exposed to CCK-8 or ethanol plus CCK-8 were depleted of STX2. STX2-KO developed more severe pancreatitis after administration of supramaximal caerulein or a 6-week ethanol diet compared with control. Acini from STX2-KO mice had increased apical exocytosis after exposure to CCK-8, as well as increased basolateral exocytosis, which led to ectopic release of proteases. These increases in apical and basolateral exocytosis required increased formation of fusogenic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes, mediated by STX3 and STX4. STX2 bound ATG16L1 and prevented it from binding clathrin. Deletion of STX2 from acini increased binding of AT16L1 to clathrin, increasing formation of pre-autophagosomes and inducing autophagy. Induction of autophagy promoted the CCK-8-induced increase in autolysosome formation and the activation of trypsinogen. CONCLUSIONS: In studies of human pancreatic tissues and pancreata from STX2-KO and control mice, we found STX2 to block STX3- and STX4-mediated fusion of zymogen granules with the plasma membrane and exocytosis and prevent binding of ATG16L1 to clathrin, which contributes to induction of autophagy. Exposure of pancreatic tissues to CCK-8 or ethanol depletes acinar cells of STX2, increasing basolateral exocytosis and promoting autophagy induction, leading to activation of trypsinogen.


Assuntos
Autofagia/genética , Exocitose/genética , Pâncreas/citologia , Pancreatite/genética , Sintaxina 1/metabolismo , Células Acinares/metabolismo , Animais , Membrana Celular/metabolismo , Ceruletídeo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/cirurgia , Pancreatite/induzido quimicamente , Vesículas Secretórias/fisiologia , Tripsinogênio/metabolismo
13.
J Biol Chem ; 292(13): 5144-5165, 2017 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-28174296

RESUMO

The role of Golgi apparatus during phagocytic uptake by macrophages has been ruled out in the past. Notably, all such reports were limited to Fcγ receptor-mediated phagocytosis. Here, we unravel a highly devolved mechanism for recruitment of Golgi-derived secretory vesicles during phagosome biogenesis, which was important for uptake of most cargos, except the IgG-coated ones. We report recruitment of mannosidase-II-positive Golgi-derived vesicles during uptake of diverse targets, including latex beads, Escherichia coli, Salmonella typhimurium, and Mycobacterium tuberculosis in human and mouse macrophages. The recruitment of mannosidase-II vesicles was an early event mediated by focal exocytosis and coincided with the recruitment of transferrin receptor, VAMP3, and dynamin-2. Brefeldin A treatment inhibited mannosidase-II recruitment and phagocytic uptake of serum-coated or -uncoated latex beads and E. coli However, consistent with previous studies, brefeldin A treatment did not affect uptake of IgG-coated latex beads. Mechanistically, recruitment of mannosidase-II vesicles during phagocytic uptake required Ca2+ from both extra- and intracellular sources apart from PI3K, microtubules, and dynamin-2. Extracellular Ca2+ via voltage-gated Ca2+ channels established a Ca2+-dependent local phosphatidylinositol 1,4,5-trisphosphate gradient, which guides the focal movement of Golgi-derived vesicles to the site of uptake. We confirmed Golgi-derived vesicles recruited during phagocytosis were secretory vesicles as their recruitment was sensitive to depletion of VAMP2 or NCS1, whereas recruitment of the recycling endosome marker VAMP3 was unaffected. Depletion of both VAMP2 and NCS1 individually resulted in the reduced uptake by macrophages. Together, the study provides a previously unprecedented role of Golgi-derived secretory vesicles in phagocytic uptake, the key innate defense function.


Assuntos
Cálcio/farmacologia , Exocitose/fisiologia , Complexo de Golgi/fisiologia , Macrófagos/metabolismo , Fagocitose/fisiologia , Vesículas Secretórias/fisiologia , Animais , Linhagem Celular , Humanos , Imunidade Inata , Manosidases/metabolismo , Camundongos , Polifosfatos/metabolismo , Células RAW 264.7 , Vesículas Secretórias/metabolismo
14.
J Immunol ; 197(6): 2090-101, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27534551

RESUMO

Helper and cytotoxic T cells accomplish focused secretion through the movement of vesicles toward the microtubule organizing center (MTOC) and translocation of the MTOC to the target contact site. In this study, using Jurkat cells and OT-I TCR transgenic primary murine CTLs, we show that the dynein-binding proteins nuclear distribution E homolog 1 (NDE1) and dynactin (as represented by p150(Glued)) form mutually exclusive complexes with dynein, exhibit nonoverlapping distributions in target-stimulated cells, and mediate different transport events. When Jurkat cells expressing a dominant negative form of NDE1 (NDE1-enhanced GFP fusion) were activated by Staphylococcus enterotoxin E-coated Raji cells, NDE1 and dynein failed to accumulate at the immunological synapse (IS) and MTOC translocation was inhibited. Knockdown of NDE1 in Jurkat cells or primary mouse CTLs also inhibited MTOC translocation and CTL-mediated killing. In contrast to NDE1, knockdown of p150(Glued), which depleted the alternative dynein/dynactin complex, resulted in impaired accumulation of CTLA4 and granzyme B-containing intracellular vesicles at the IS, whereas MTOC translocation was not affected. Depletion of p150(Glued) in CTLs also inhibited CTL-mediated lysis. We conclude that the NDE1/Lissencephaly 1 and dynactin complexes separately mediate two key components of T cell-focused secretion, namely translocation of the MTOC and lytic granules to the IS, respectively.


Assuntos
Complexo Dinactina/fisiologia , Dineínas/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Linfócitos T/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/fisiologia , Sinalização do Cálcio , Citotoxicidade Imunológica , Humanos , Células Jurkat , Centro Organizador dos Microtúbulos/metabolismo , Vesículas Secretórias/fisiologia , Sinapses/metabolismo , Linfócitos T Citotóxicos/imunologia
15.
Oncotarget ; 7(37): 60687-60697, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27463001

RESUMO

Despite improvements in the diagnosis and treatment of hepatocellular carcinoma (HCC), the prognosis is still poor. Pioneering work has demonstrated a potential role for tumour cell-derived exosomes (TEXs) in HCC. TEXs can mediate immune responses, antigen presentation and intracellular communication by serving as vehicles for the transfer of proteins, viruses, lipids and RNA between cells. An improved understanding of the roles played by exosomes could lead to a powerful new strategy for preventing and treating HCC. In this review, we summarise current understanding on the topic. The literature points to two faces of TEXs in HCC: 1) They can promote invasion, metastasis, immune evasion and modulation and 2) they can act as diagnostic and prognostic biomarkers, and can be used in anti-cancer drug resistance and immunotherapy in the future.


Assuntos
Biomarcadores Tumorais/fisiologia , Carcinoma Hepatocelular/metabolismo , Exossomos/fisiologia , Imunoterapia/métodos , Neoplasias Hepáticas/metabolismo , Vesículas Secretórias/fisiologia , Animais , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/patologia , Metástase Neoplásica , Evasão Tumoral , Microambiente Tumoral
16.
Mol Biol Cell ; 27(13): 2080-9, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27146112

RESUMO

The survival of Toxoplasma gondii within its host cell requires protein release from secretory vesicles, called dense granules, to maintain the parasite's intracellular replicative niche. Despite the importance of DGs, nothing is known about the mechanisms underlying their transport. In higher eukaryotes, secretory vesicles are transported to the plasma membrane by molecular motors moving on their respective cytoskeletal tracks (i.e., microtubules and actin). Because the organization of these cytoskeletal structures differs substantially in T. gondii, the molecular motor dependence of DG trafficking is far from certain. By imaging the motions of green fluorescent protein-tagged DGs in intracellular parasites with high temporal and spatial resolution, we show through a combination of molecular genetics and chemical perturbations that directed DG transport is independent of microtubules and presumably their kinesin/dynein motors. However, directed DG transport is dependent on filamentous actin and a unique class 27 myosin, TgMyoF, which has structural similarity to myosin V, the prototypical cargo transporter. Actomyosin DG transport was unexpected, since filamentous parasite actin has yet to be visualized in vivo due in part to the prevailing model that parasite actin forms short, unstable filaments. Thus our data uncover new critical roles for these essential proteins in the lytic cycle of this devastating pathogen.


Assuntos
Miosina Tipo V/metabolismo , Toxoplasma/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Membrana Celular/metabolismo , Movimento Celular , Vesículas Citoplasmáticas/metabolismo , Citoesqueleto/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Miosinas , Transporte Proteico , Vesículas Secretórias/metabolismo , Vesículas Secretórias/fisiologia , Análise Espaço-Temporal , Toxoplasma/genética
17.
J Neurochem ; 137(6): 897-903, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26990968

RESUMO

The accumulation of neurotransmitters within secretory vesicles (SVs) far exceeds the theoretical tonic concentrations in the cytosol, a phenomenon that has captivated the attention of scientists for decades. For instance, chromaffin granules can accumulate close to molar concentrations of catecholamines, along with many other products like ATP, calcium, peptides, chromogranins, ascorbate, and other nucleotides. In this short review, we will summarize the interactions that are currently believed to occur between the elements that make up the vesicular cocktail in the acidic environment of SVs, and how they permit the accumulation of such high concentrations of certain components. In addition, we will examine how the vesicular cocktail regulates the exocytosis of neurotransmitters. In this review, we have highlighted the mechanisms that permit the storage of neurotransmitters and hormones inside secretory vesicles. We also have proposed a novel model based in the intravesicular interactions of the main components of this inner cocktail - catecholamines, ATP, and chromogranins - to allow the accumulation of near molar concentrations of transmitters in secretory vesicles. This article is part of a mini review series on Chromaffin cells (ISCCB Meeting, 2015).


Assuntos
Exocitose/fisiologia , Neurotransmissores/metabolismo , Vesículas Secretórias/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Grânulos Cromafim/fisiologia , Cromograninas/metabolismo , Humanos , Modelos Biológicos
18.
Biochem Biophys Res Commun ; 465(4): 825-31, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26319431

RESUMO

The aim of the present study was to evaluate the effects of hydrogen sulfide (H2S) on the membrane potential, action potential discharge and exocytosis of secretory granules in neurosecretory pituitary tumor cells (GH3). The H2S donor - sodium hydrosulfide (NaHS) induced membrane hyperpolarization, followed by truncation of spontaneous electrical activity and decrease of the membrane resistance. The NaHS effect was dose-dependent with an EC50 of 152 µM (equals effective H2S of 16-19 µM). NaHS effects were not altered after inhibition of maxi conductance calcium-activated potassium (BK) channels by tetraethylammonium or paxilline, but were significantly reduced after inhibition or activation of ATP-dependent potassium channels (KATP) by glibenclamide or by diazoxide, respectively. In whole-cell recordings NaHS increased the amplitude of KATP currents, induced by hyperpolarizing pulses and subsequent application of glibenclamide decreased currents to control levels. Using the fluorescent dye FM 1-43 exocytosis of secretory granules was analyzed in basal and stimulated conditions (high K(+) external solution). Prior application of NaHS decreased the fluorescence of the cell membrane in both conditions which links with activation of KATP currents (basal secretion) and activation of KATP currents and BK-currents (stimulated exocytosis). We suggest that H2S induces hyperpolarization of GH3 cells by activation of KATP channels which results in a truncation of spontaneous action potentials and a decrease of hormone release.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Neoplasias Hipofisárias/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Linhagem Celular Tumoral , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Sulfeto de Hidrogênio/farmacologia , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/deficiência , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Ratos , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/fisiologia , Sulfetos/metabolismo , Sulfetos/farmacologia
19.
Leuk Res ; 39(8): 921-4, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26105049

RESUMO

The expression of BCR-ABL in hematopoietic stem cells is a well-defined primary event in chronic myeloid leukemia (CML). Some reports have described the presence of BCR-ABL on endothelial cells from CML patients, suggesting the origin of the disease in a primitive hemangioblastic cell. On the other hand, extracellular vesicles (EVs) released by CML leukemic cells are involved in the angiogenesis modulation process. In the current work we hypothesized that EVs released from BCR-ABL(+) cells may carry inside the oncogene that can be transferred to endothelial cells leading to the expression of both BCR-ABL transcript and the oncoprotein. EVs from K562 cells and plasma of newly diagnosed CML patients were isolated by ultracentrifugation. RT-PCR analysis detected the presence of BCR-ABL RNA in the EVs isolated from both K562 cells and plasma of CML patients. The incorporation of these EVs into endothelial cells was demonstrated by flow cytometry and fluorescence microscopy showed that after 24h of incubation most EVs were incorporated. BCR-ABL transcripts were detected in all experiments on endothelial cells incubated with EVs from both sources. The presence of BCR-ABL on endothelial cells incubated with Philadelphia(+) EVs was also confirmed by Western blot assays. In summary, endothelial cells acquire BCR-ABL RNA and the oncoprotein after incubation with EVs released from Ph(+) positive cells (either from K562 cells or from plasma of newly diagnosed CML patients). This results challenge the hypothesis that endothelial cells may be part of the Philadelphia(+) clone in CML.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Neoplásicas/patologia , Vesículas Secretórias/fisiologia , Células Cultivadas , Células Clonais/metabolismo , Células Clonais/patologia , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Células-Tronco Hematopoéticas/patologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo
20.
Nat Commun ; 6: 6297, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25708831

RESUMO

In neurosecretory cells, secretory vesicles (SVs) undergo Ca(2+)-dependent fusion with the plasma membrane to release neurotransmitters. How SVs cross the dense mesh of the cortical actin network to reach the plasma membrane remains unclear. Here we reveal that, in bovine chromaffin cells, SVs embedded in the cortical actin network undergo a highly synchronized transition towards the plasma membrane and Munc18-1-dependent docking in response to secretagogues. This movement coincides with a translocation of the cortical actin network in the same direction. Both effects are abolished by the knockdown or the pharmacological inhibition of myosin II, suggesting changes in actomyosin-generated forces across the cell cortex. Indeed, we report a reduction in cortical actin network tension elicited on secretagogue stimulation that is sensitive to myosin II inhibition. We reveal that the cortical actin network acts as a 'casting net' that undergoes activity-dependent relaxation, thereby driving tethered SVs towards the plasma membrane where they undergo Munc18-1-dependent docking.


Assuntos
Actinas/metabolismo , Células Cromafins/fisiologia , Proteínas Munc18/metabolismo , Miosina Tipo II/metabolismo , Neurossecreção , Vesículas Secretórias/fisiologia , Animais , Bovinos , Compostos Heterocíclicos de 4 ou mais Anéis , Células PC12 , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA