Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
Nat Commun ; 12(1): 6439, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750373

RESUMO

The S. cerevisiae plasma membrane H+-ATPase, Pma1, is a P3A-type ATPase and the primary protein component of the membrane compartment of Pma1 (MCP). Like other plasma membrane H+-ATPases, Pma1 assembles and functions as a hexamer, a property unique to this subfamily among the larger family of P-type ATPases. It has been unclear how Pma1 organizes the yeast membrane into MCP microdomains, or why it is that Pma1 needs to assemble into a hexamer to establish the membrane electrochemical proton gradient. Here we report a high-resolution cryo-EM study of native Pma1 hexamers embedded in endogenous lipids. Remarkably, we found that the Pma1 hexamer encircles a liquid-crystalline membrane domain composed of 57 ordered lipid molecules. The Pma1-encircled lipid patch structure likely serves as the building block of the MCP. At pH 7.4, the carboxyl-terminal regulatory α-helix binds to the phosphorylation domains of two neighboring Pma1 subunits, locking the hexamer in the autoinhibited state. The regulatory helix becomes disordered at lower pH, leading to activation of the Pma1 hexamer. The activation process is accompanied by a 6.7 Å downward shift and a 40° rotation of transmembrane helices 1 and 2 that line the proton translocation path. The conformational changes have enabled us to propose a detailed mechanism for ATP-hydrolysis-driven proton pumping across the plasma membrane. Our structures will facilitate the development of antifungal drugs that target this essential protein.


Assuntos
Membrana Celular/enzimologia , Microdomínios da Membrana/enzimologia , ATPases Translocadoras de Prótons/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Transporte Biológico/genética , Domínio Catalítico , Membrana Celular/ultraestrutura , Microscopia Crioeletrônica , Ativação Enzimática , Hidrólise , Microdomínios da Membrana/ultraestrutura , Modelos Moleculares , Mutação , Conformação Proteica , Multimerização Proteica , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/metabolismo , Prótons , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Front Immunol ; 10: 1729, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31404305

RESUMO

Extracellular adenine nucleotides participate in cell-to-cell communication and modulate the immune response. The concerted action of ectonucleotidases CD39 and CD73 plays a major role in the local production of anti-inflammatory adenosine, but both ectonucleotidases are rarely co-expressed by human T cells. The expression of CD39 on T cells increases upon T cell activation and is high at sites of inflammation. CD73, in contrast, disappears from the cellular membrane after activation. The possibility that CD73 could act in trans would resolve the conundrum of both enzymes being co-expressed for the degradation of ATP and the generation of adenosine. An enzymatically active soluble form of CD73 has been reported, and AMPase activity has been detected in body fluids of patients with inflammation and cancer. It is not yet clear how CD73, a glycosylphosphatidylinositol (GPI)-anchored protein, is released from the cell membrane, but plausible mechanisms include cleavage by metalloproteinases and shedding mediated by cell-associated phospholipases. Importantly, like many other GPI-anchored proteins, CD73 at the cell membrane is preferentially localized in detergent-resistant domains or lipid rafts, which often contribute to extracellular vesicles (EVs). Indeed, CD73-containing vesicles of different size and origin and with immunomodulatory function have been found in the tumor microenvironment. The occurrence of CD73 as non-cell-bound molecule widens the range of action of this enzyme at sites of inflammation. In this review, we will discuss the generation of non-cell-bound CD73 and its physiological role in inflammation.


Assuntos
5'-Nucleotidase/fisiologia , Inflamação/imunologia , 5'-Nucleotidase/biossíntese , 5'-Nucleotidase/genética , Adenosina/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Comunicação Celular , Membrana Celular/enzimologia , Líquido Extracelular/metabolismo , Vesículas Extracelulares/enzimologia , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/fisiologia , Glicosilfosfatidilinositóis/metabolismo , Humanos , Inflamação/metabolismo , Ativação Linfocitária , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Microdomínios da Membrana/enzimologia , Camundongos , Proteínas de Neoplasias/fisiologia , Neoplasias/imunologia , Neoplasias/patologia , Receptores Purinérgicos P1/fisiologia , Solubilidade , Especificidade da Espécie , Microambiente Tumoral
4.
Haematologica ; 104(10): 1984-1994, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30819915

RESUMO

Transmembrane protein 30A (Tmem30a) is the ß-subunit of P4-ATPases which function as flippase that transports aminophospholipids such as phosphatidylserine from the outer to the inner leaflets of the plasma membrane to maintain asymmetric distribution of phospholipids. It has been documented that deficiency of Tmem30a led to exposure of phosphatidylserine. However, the role of Tmem30a in vivo remains largely unknown. Here we found that Vav-Cre-driven conditional deletion of Tmem30a in hematopoietic cells led to embryonic lethality due to severe anemia by embryonic day 16.5. The numbers of erythroid colonies and erythroid cells were decreased in the Tmem30a deficient fetal liver. This was accompanied by increased apoptosis of erythroid cells. Confocal microscopy analysis revealed an increase of localization of erythropoietin receptor to areas of membrane raft microdomains in response to erythropoietin stimulation in Ter119-erythroid progenitors, which was impaired in Tmem30a deficient cells. Moreover, erythropoietin receptor (EPOR)-mediated activation of the STAT5 pathway was significantly reduced in Tmem30a deficient fetal liver cells. Consistently, knockdown of TMEM30A in human CD34+ cells also impaired erythropoiesis. Our findings demonstrate that Tmem30a plays a critical role in erythropoiesis by regulating the EPOR signaling pathway through the formation of membrane rafts in erythroid cells.


Assuntos
Eritropoese , Feto/embriologia , Hematopoese Extramedular , Células-Tronco Hematopoéticas/enzimologia , Fígado/embriologia , Proteínas de Membrana/deficiência , Animais , Feto/citologia , Células-Tronco Hematopoéticas/citologia , Fígado/citologia , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais
5.
Artigo em Inglês | MEDLINE | ID: mdl-30196154

RESUMO

The mitochondrial F1FO-ATPase, the key enzyme in cell bioenergetics, apparently works in the same way in mollusks and in mammals. We previously pointed out a raft-like arrangement in mussel gill mitochondrial membranes, which apparently distinguishes bivalve mollusks from mammals. To explore the relationship between the microenvironmental features and the enzyme activity, the physico-chemical features of mitochondrial membranes and the F1FO-ATPase activity temperature-dependence are here explored in the Manila clam (Ruditapes philippinarum). Similarly to the mussel, clam gill mitochondrial membrane lipids exhibit a high sterol content (42 mg/g protein), mainly due to phytosterols (cholesterol only attains 42% of total sterols), and abundant polyunsaturated fatty acids (PUFA) (70% of total fatty acids), especially of the n-3 family. However, the F1FO-ATPase activation energies above and below the break in the Arrhenius plot (22.1 °C) are lower than in mussel and mammalian mitochondria. Laurdan fluorescence spectroscopy analyses carried out at 10 °C, 20 °C and 30 °C on mitochondrial membranes and on lipid vesicles obtained from total lipid extracts of mitochondria, indicate a physical state without coexisting domains. This mitochondrial membrane constitution, allowed by lipid-lipid and lipidprotein interactions and involving PUFA-rich phospholipids, phytosterols (much more diversified in clams than in mussels) and proteins, enables the maintenance of a homogeneous physical state in the range 10-30 °C. Consistently, this molecular interaction network would somehow extend the temperature range of the F1FO-ATPase activity and may contribute to clam resilience to temperature changes.


Assuntos
Bivalves/fisiologia , Mudança Climática , Metabolismo dos Lipídeos , Membranas Mitocondriais/metabolismo , Modelos Biológicos , ATPases Translocadoras de Prótons/metabolismo , Animais , Bivalves/enzimologia , Bivalves/crescimento & desenvolvimento , Ativação Enzimática , Estabilidade Enzimática , Ácidos Graxos Ômega-3/análise , Ácidos Graxos Ômega-3/química , Ácidos Graxos Insaturados/análise , Ácidos Graxos Insaturados/química , Feminino , Temperatura Alta/efeitos adversos , Itália , Bicamadas Lipídicas , Lipossomos , Masculino , Mar Mediterrâneo , Microdomínios da Membrana/química , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/metabolismo , Membranas Mitocondriais/química , Fitosteróis/análise , Fitosteróis/metabolismo , ATPases Translocadoras de Prótons/química , Especificidade da Espécie , Esteróis/análise , Esteróis/metabolismo
6.
Methods Mol Biol ; 1821: 393-399, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30062426

RESUMO

Rho-type small GTPases (Rho GTPases) play central roles in various cellular events. Rho GTPases are often activated locally on the plasma membrane, forming plasma membrane domains, which induce downstream signaling. We describe an experimental procedure designed for inducing the production of de novo plasma membrane domains using Arabidopsis ROP11 GTPase. Introduction of ROP11 and its activator and inactivator into the tobacco leaf epidermis leads to formation of ROP11-activated plasma membrane domains on the plasma membrane. Effectors and marker genes can also be introduced alongside ROP11. This reconstruction system allows identifying molecules regulating Rho GTPase polarization.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimologia , Microdomínios da Membrana/enzimologia , Proteínas rho de Ligação ao GTP/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Microdomínios da Membrana/genética , Proteínas rho de Ligação ao GTP/genética
7.
Proc Natl Acad Sci U S A ; 115(28): E6497-E6506, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29941564

RESUMO

Evidence supporting the heterogeneity in cAMP and PKA signaling is rapidly accumulating and has been largely attributed to the localization or activity of adenylate cyclases, phosphodiesterases, and A-kinase-anchoring proteins in different cellular subcompartments. However, little attention has been paid to the possibility that, despite homogeneous cAMP levels, a major heterogeneity in cAMP/PKA signaling could be generated by the spatial distribution of the final terminators of this cascade, i.e., the phosphatases. Using FRET-based sensors to monitor cAMP and PKA-dependent phosphorylation in the cytosol and outer mitochondrial membrane (OMM) of primary rat cardiomyocytes, we demonstrate that comparable cAMP increases in these two compartments evoke higher levels of PKA-dependent phosphorylation in the OMM. This difference is most evident for small, physiological increases of cAMP levels and with both OMM-located probes and endogenous OMM proteins. We demonstrate that this disparity depends on differences in the rates of phosphatase-dependent dephosphorylation of PKA targets in the two compartments. Furthermore, we show that the activity of soluble phosphatases attenuates PKA-driven activation of the cAMP response element-binding protein while concurrently enhancing PKA-dependent mitochondrial elongation. We conclude that phosphatases can sculpt functionally distinct cAMP/PKA domains even in the absence of gradients or microdomains of this messenger. We present a model that accounts for these unexpected results in which the degree of PKA-dependent phosphorylation is dictated by both the subcellular distribution of the phosphatases and the different accessibility of membrane-bound and soluble phosphorylated substrates to the cytosolic enzymes.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/metabolismo , Membranas Mitocondriais/enzimologia , Proteínas Mitocondriais/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/genética , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Humanos , Microdomínios da Membrana/genética , Proteínas de Membrana/genética , Proteínas Mitocondriais/genética , Ratos , Ratos Sprague-Dawley
8.
Am J Respir Cell Mol Biol ; 58(4): 530-541, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29262264

RESUMO

Two cAMP signaling compartments centered on adenylyl cyclase (AC) exist in human airway smooth muscle (HASM) cells, one containing ß2-adrenergic receptor AC6 and another containing E prostanoid receptor AC2. We hypothesized that different PDE isozymes selectively regulate cAMP signaling in each compartment. According to RNA-sequencing data, 18 of 24 PDE genes were expressed in primary HASM cells derived from age- and sex-matched donors with and without asthma. PDE8A was the third most abundant of the cAMP-degrading PDE genes, after PDE4A and PDE1A. Knockdown of PDE8A using shRNA evoked twofold greater cAMP responses to 1 µM forskolin in the presence of 3-isobutyl-1-methylxanthine. Overexpression of AC2 did not alter this response, but overexpression of AC6 increased cAMP responses an additional 80%. We examined cAMP dynamics in live HASM cells using a fluorescence sensor. PF-04957325, a PDE8-selective inhibitor, increased basal cAMP concentrations by itself, indicating a significant basal level of cAMP synthesis. In the presence of an AC inhibitor to reduce basal signaling, PF-04957325 accelerated cAMP production and increased the inhibition of cell proliferation induced by isoproterenol, but it had no effect on cAMP concentrations or cell proliferation regulated by prostaglandin E2. Lipid raft fractionation of HASM cells revealed PDE8A immunoreactivity in buoyant fractions containing caveolin-1 and AC5/6 immunoreactivity. Thus, PDE8 is expressed in lipid rafts of HASM cells, where it specifically regulates ß2-adrenergic receptor AC6 signaling without effects on signaling by the E prostanoid receptors 2/4-AC2 complex. In airway diseases such as asthma and chronic obstructive pulmonary disease, PDE8 may represent a novel therapeutic target to modulate HASM responsiveness and airway remodeling.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Adenilil Ciclases/metabolismo , Asma/enzimologia , AMP Cíclico/metabolismo , Músculo Liso/enzimologia , Miócitos de Músculo Liso/enzimologia , Receptores Adrenérgicos beta 2/metabolismo , Sistema Respiratório/enzimologia , 3',5'-AMP Cíclico Fosfodiesterases/genética , Adenilil Ciclases/genética , Remodelação das Vias Aéreas , Asma/genética , Asma/patologia , Asma/fisiopatologia , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Humanos , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/patologia , Músculo Liso/patologia , Músculo Liso/fisiopatologia , Miócitos de Músculo Liso/patologia , Receptores Adrenérgicos beta 2/genética , Sistema Respiratório/patologia , Sistema Respiratório/fisiopatologia , Sistemas do Segundo Mensageiro , Fatores de Tempo
9.
PLoS Negl Trop Dis ; 11(8): e0005805, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28829771

RESUMO

BACKGROUND: Leishmaniasis is the world's second deadliest parasitic disease after malaria, and current treatment of the different forms of this disease is far from satisfactory. Alkylphospholipid analogs (APLs) are a family of anticancer drugs that show antileishmanial activity, including the first oral drug (miltefosine) for leishmaniasis and drugs in preclinical/clinical oncology trials, but their precise mechanism of action remains to be elucidated. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that the tumor cell apoptosis-inducer edelfosine was the most effective APL, as compared to miltefosine, perifosine and erucylphosphocholine, in killing Leishmania spp. promastigotes and amastigotes as well as tumor cells, as assessed by DNA breakdown determined by flow cytometry. In studies using animal models, we found that orally-administered edelfosine showed a potent in vivo antileishmanial activity and diminished macrophage pro-inflammatory responses. Edelfosine was also able to kill Leishmania axenic amastigotes. Edelfosine was taken up by host macrophages and killed intracellular Leishmania amastigotes in infected macrophages. Edelfosine accumulated in tumor cell mitochondria and Leishmania kinetoplast-mitochondrion, and led to mitochondrial transmembrane potential disruption, and to the successive breakdown of parasite mitochondrial and nuclear DNA. Ectopic expression of Bcl-XL inhibited edelfosine-induced cell death in both Leishmania parasites and tumor cells. We found that the cytotoxic activity of edelfosine against Leishmania parasites and tumor cells was associated with a dramatic recruitment of FOF1-ATP synthase into lipid rafts following edelfosine treatment in both parasites and cancer cells. Raft disruption and specific FOF1-ATP synthase inhibition hindered edelfosine-induced cell death in both Leishmania parasites and tumor cells. Genetic deletion of FOF1-ATP synthase led to edelfosine drug resistance in Saccharomyces cerevisiae yeast. CONCLUSIONS/SIGNIFICANCE: The present study shows that the antileishmanial and anticancer actions of edelfosine share some common signaling processes, with mitochondria and raft-located FOF1-ATP synthase being critical in the killing process, thus identifying novel druggable targets for the treatment of leishmaniasis.


Assuntos
Antineoplásicos/farmacologia , Antiprotozoários/farmacologia , Leishmania/efeitos dos fármacos , Microdomínios da Membrana/enzimologia , Mitocôndrias/enzimologia , Éteres Fosfolipídicos/farmacologia , ATPases Translocadoras de Prótons/antagonistas & inibidores , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Deleção de Genes , Humanos , Leishmaniose/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Resultado do Tratamento
10.
Mol Biol Cell ; 28(8): 1147-1159, 2017 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-28228554

RESUMO

Lipopolysaccharide (LPS) is the component of Gram-negative bacteria that activates Toll-like receptor 4 (TLR4) to trigger proinflammatory responses. We examined the involvement of Lyn tyrosine kinase in TLR4 signaling of macrophages, distinguishing its catalytic activity and intermolecular interactions. For this, a series of Lyn-GFP constructs bearing point mutations in particular domains of Lyn were overexpressed in RAW264 macrophage-like cells or murine peritoneal macrophages, and their influence on LPS-induced responses was analyzed. Overproduction of wild-type or constitutively active Lyn inhibited production of TNF-α and CCL5/RANTES cytokines and down-regulated the activity of NFκB and IRF3 transcription factors in RAW264 cells. The negative influence of Lyn was nullified by point mutations of Lyn catalytic domain or Src homology 2 (SH2) or SH3 domains or of the cysteine residue that undergoes LPS-induced palmitoylation. Depending on the cell type, overproduction of those mutant forms of Lyn could even up-regulate LPS-induced responses, and this effect was reproduced by silencing of endogenous Lyn expression. Simultaneously, the Lyn mutations blocked its LPS-induced accumulation in the raft fraction of RAW264 cells. These data indicate that palmitoylation, SH2- and SH3-mediated intermolecular interactions, and the catalytic activity of Lyn are required for its accumulation in rafts, thereby determining the negative regulation of TLR4 signaling.


Assuntos
Microdomínios da Membrana/enzimologia , Quinases da Família src/genética , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Quimiocina CCL5/metabolismo , Proteínas de Fluorescência Verde , Fator Regulador 3 de Interferon/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
FEBS J ; 284(9): 1267-1278, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27973739

RESUMO

Phosphatidylinositol-3-phosphate (PI3P) is a key player in membrane dynamics and trafficking regulation. Most PI3P is associated with endosomal membranes and with the autophagosome preassembly machinery, presumably at the endoplasmic reticulum. The enzyme responsible for most PI3P synthesis, VPS34 and proteins such as Beclin1 and ATG14L that regulate PI3P levels are positive modulators of autophagy initiation. It had been assumed that a local PI3P pool was present at autophagosomes and preautophagosomal structures, such as the omegasome and the phagophore. This was recently confirmed by the demonstration that PI3P-binding proteins participate in the complex sequence of signalling that results in autophagosome assembly and activity. Here we summarize the historical discoveries of PI3P lipid kinase involvement in autophagy, and we discuss the proposed role of PI3P during autophagy, notably during the autophagosome biogenesis sequence.


Assuntos
Autofagossomos/fisiologia , Autofagia , Microdomínios da Membrana/fisiologia , Modelos Biológicos , Biogênese de Organelas , Fosfatos de Fosfatidilinositol/metabolismo , Sistemas do Segundo Mensageiro , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Autofagossomos/enzimologia , Proteínas Relacionadas à Autofagia/metabolismo , Proteína Beclina-1/metabolismo , Classe II de Fosfatidilinositol 3-Quinases/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/fisiologia , Endossomos/enzimologia , Endossomos/fisiologia , Humanos , Lisossomos/enzimologia , Lisossomos/fisiologia , Microdomínios da Membrana/enzimologia
12.
Adv Exp Med Biol ; 981: 3-21, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29594855

RESUMO

In this chapter the four different genes of the mammalian plasma membrane calcium ATPase (PMCA) and their spliced isoforms are discussed with respect to the structural and functional properties of PMCA, the tissue distribution of the different isoforms, including their differences during development. The importance of PMCA for regulating Ca2+ signaling in microdomains under different conditions is also discussed.


Assuntos
Sinalização do Cálcio/fisiologia , Microdomínios da Membrana/enzimologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Animais , Humanos , Microdomínios da Membrana/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética
13.
Elife ; 52016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27700986

RESUMO

We aimed to understand how spatial compartmentalization in the plasma membrane might contribute to the functions of the ubiquitous class IA phosphoinositide 3-kinase (PI3K) isoforms, p110α and p110ß. We found that p110ß localizes to membrane rafts in a Rac1-dependent manner. This localization potentiates Akt activation by G-protein-coupled receptors (GPCRs). Thus genetic targeting of a Rac1 binding-deficient allele of p110ß to rafts alleviated the requirement for p110ß-Rac1 association for GPCR signaling, cell growth and migration. In contrast, p110α, which does not play a physiological role in GPCR signaling, is found to reside in nonraft regions of the plasma membrane. Raft targeting of p110α allowed its EGFR-mediated activation by GPCRs. Notably, p110ß dependent, PTEN null tumor cells critically rely upon raft-associated PI3K activity. Collectively, our findings provide a mechanistic account of how membrane raft localization regulates differential activation of distinct PI3K isoforms and offer insight into why PTEN-deficient cancers depend on p110ß.


Assuntos
Membrana Celular/enzimologia , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Microdomínios da Membrana/enzimologia , Neuropeptídeos/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Fibroblastos/fisiologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
14.
Biochim Biophys Acta ; 1863(11): 2795-2808, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27599715

RESUMO

By mediating proteolytic shedding on the cell surface the disintegrin and metalloproteinases ADAM10 and ADAM17 function as critical regulators of growth factors, cytokines and adhesion molecules. We here report that stimulation of lung epithelial A549 tumor cells with phorbol-12-myristate-13-acetate (PMA) leads to the downregulation of the surface expressed mature form of ADAM17 without affecting ADAM10 expression. This reduction could not be sufficiently explained by metalloproteinase-mediated degradation, dynamin-mediated internalization or microdomain redistribution of ADAM17. Instead, surface downregulation of ADAM17 was correlated with the presence of its mature form in exosomes. Exosomal ADAM17 release was also observed in monocytic and primary endothelial cells where it could be induced by stimulation with lipopolysaccharide. Antibody-mediated surface labelling of ADAM17 revealed that at least part of exosomal ADAM17 was oriented with the metalloproteinase domain outside and had been expressed on the cell surface. Suppression of iRHOM2-mediated ADAM17 maturation prevented surface expression and exosomal release of ADAM17. Further, deletion of the protease's C-terminus or cell treatment with a calcium chelator diminished exosomal release as well as surface downregulation of ADAM17, underlining that both processes are closely associated. Co-incubation of ADAM17 containing exosomes with cells expressing the ADAM17 substrates TGFα or amphiregulin lead to increased shedding of both substrates. This was prevented when exosomes were prepared from cells with shRNA-mediated ADAM17 knockdown. These data indicate that cell stimulation can downregulate expression of mature ADAM17 from the cell surface and induce release of exosomal ADAM17, which can then distribute and contribute to substrate shedding on more distant cells.


Assuntos
Proteína ADAM17/metabolismo , Exossomos/enzimologia , Células A549 , Proteína ADAM10/metabolismo , Proteína ADAM17/genética , Anfirregulina/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Sinalização do Cálcio , Proteínas de Transporte/metabolismo , Células Endoteliais/enzimologia , Ativação Enzimática , Exossomos/efeitos dos fármacos , Exossomos/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos/farmacologia , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/metabolismo , Monócitos/enzimologia , Transporte Proteico , Interferência de RNA , Especificidade por Substrato , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Fator de Crescimento Transformador alfa/metabolismo
15.
Cell Oncol (Dordr) ; 39(4): 353-63, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27042827

RESUMO

PURPOSE: Tumor progression is associated with cell migration, invasion and metastasis. These processes are accompanied by the activation of specific proteases that are either linked to cellular membranes or are secreted into extracellular spaces. TNF-α is known to play an important role in various aspects of tumor progression. The aim of this work was to assess the effect of TNF-α on the migration of breast cancer cells and, in addition, to assess its association with the location of membrane-associated proteases in lipid rafts. METHODS: Wound scratch healing and Transwell migration assays were used to study the effect of TNF-α on the migration of both hormone-dependent and hormone-independent breast cancer-derived cells, i.e., MCF7 and MDA-MB-231, respectively. The expression and secretion of three matrix metalloproteases, MMP9, MMP2 and MT1-MMP, and two dipeptidyl peptidases, CD26 and FAP-α, was investigated using RT-PCR, Western blotting and gelatin zymography. In addition, activation of the MAPK/ERK signaling pathway was investigated by Western blotting. RESULTS: We found that a TNF-α-induced enhancement of breast cancer cell migration was accompanied by an increased secretion of MMP9, but not MMP2, into the culture media. We also found that TNF-α upregulated the expression of the dipeptidyl peptidases CD26 and FAP-α in a dose-dependent manner and, in addition, enhanced the concentration of all five proteases in lipid rafts in the breast cancer-derived cells tested, regardless of cell type. Furthermore, we found that TNF-α activated the MAPK/ERK signaling pathway by increasing the ERK1/2 phosphorylation level. Application of the MEK/ERK1/2 inhibitor U-0126 resulted in down-regulation of TNF-α-induced MMP9 secretion and abrogation of the enhanced concentration of proteases in the lipid rafts. CONCLUSIONS: From our results we conclude that TNF-α-induced activation of the MAPK/ERK signaling pathway may promote breast cancer cell migration via both upregulation of MMP9, CD26 and FAP-α and concentration of these proteases, as also MT1-MMP and MMP2, in the lipid rafts. TNF-α may serve as a potential therapeutic target in breast cancers susceptible to TNF-α stimulation.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular , Microdomínios da Membrana/enzimologia , Peptídeo Hidrolases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Western Blotting , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Feminino , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
16.
Oncotarget ; 7(25): 37536-37555, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-26918609

RESUMO

The desmosomal cadherin, desmoglein 2 (Dsg2), is deregulated in a variety of human cancers including those of the skin. When ectopically expressed in the epidermis of transgenic mice, Dsg2 activates multiple mitogenic signaling pathways and increases susceptibility to tumorigenesis. However, the molecular mechanism responsible for Dsg2-mediated cellular signaling is poorly understood. Here we show overexpression as well as co-localization of Dsg2 and EGFR in cutaneous SCCs in vivo. Using HaCaT keratinocytes, knockdown of Dsg2 decreases EGFR expression and abrogates the activation of EGFR, c-Src and Stat3, but not Erk1/2 or Akt, in response to EGF ligand stimulation. To determine whether Dsg2 mediates signaling through lipid microdomains, sucrose density fractionation illustrated that Dsg2 is recruited to and displaces Cav1, EGFR and c-Src from light density lipid raft fractions. STED imaging confirmed that the presence of Dsg2 disperses Cav1 from the cell-cell borders. Perturbation of lipid rafts with the cholesterol-chelating agent MßCD also shifts Cav1, c-Src and EGFR out of the rafts and activates signaling pathways. Functionally, overexpression of Dsg2 in human SCC A431 cells enhances EGFR activation and increases cell proliferation and migration through a c-Src and EGFR dependent manner. In summary, our data suggest that Dsg2 stimulates cell growth and migration by positively regulating EGFR level and signaling through a c-Src and Cav1-dependent mechanism using lipid rafts as signal modulatory platforms.


Assuntos
Caveolina 1/metabolismo , Desmogleína 2/biossíntese , Receptores ErbB/biossíntese , Quinases da Família src/metabolismo , Proteína Tirosina Quinase CSK , Caveolina 1/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Desmogleína 2/genética , Desmogleína 2/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Humanos , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Regulação para Cima , Quinases da Família src/genética
17.
Am J Physiol Gastrointest Liver Physiol ; 310(7): G510-25, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26718771

RESUMO

Although significant research data exist on the pathophysiology of nonalcoholic steatohepatitis (NASH), finding an efficient treatment regimen for it remains elusive. The present study used sparstolonin B (SsnB), a novel TLR4 antagonist derived from the Chinese herb Sparganium stoloniferum, as a possible drug to mitigate early inflammation in NASH. This study used an early steatohepatitic injury model in high-fat-fed mice with CYP2E1-mediated oxidative stress as a second hit. SsnB was administered for 1 wk along with bromodichloromethane (BDCM), an inducer of CYP2E1-mediated oxidative stress. Results showed that SsnB administration attenuated inflammatory morphology and decreased elevation of the liver enzyme alanine aminotransferase (ALT). Mice administered SsnB also showed decreased mRNA expression of proinflammatory cytokines TNF-α, IFN-γ, IL-1ß, and IL-23, while protein levels of both TNF-α and IL-1ß were significantly decreased. SsnB significantly decreased Kupffer cell activation as evidenced by reduction in CD68 and monocyte chemoattractant protein-1 (MCP1) mRNA and protein levels with concomitant inhibition of macrophage infiltration in the injured liver. Mechanistically, SsnB decreased TLR4 trafficking to the lipid rafts, a phenomenon described by the colocalization of TLR4 and lipid raft marker flotillin in tissues and immortalized Kupffer cells. Since we have shown previously that NADPH oxidase drives TLR4 trafficking in NASH, we studied the role of SsnB in modulating this pathway. SsnB prevented NADPH oxidase activation in vivo and in vitro as indicated by decreased peroxynitrite formation. In summary, the present study reports a novel use of the TLR4 antagonist SsnB in mitigating inflammation in NASH and in parallel shows a unique molecular mechanism of decreasing nitrative stress.


Assuntos
Anti-Inflamatórios/farmacologia , Hepatite/prevenção & controle , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Fígado/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , NADPH Oxidases/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Linhagem Celular , Citocromo P-450 CYP2E1/biossíntese , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Indução Enzimática , Hepatite/enzimologia , Hepatite/genética , Hepatite/patologia , Mediadores da Inflamação/metabolismo , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Fígado/enzimologia , Fígado/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Masculino , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Estresse Oxidativo/efeitos dos fármacos , PTEN Fosfo-Hidrolase/metabolismo , Ácido Peroxinitroso/metabolismo , Transporte Proteico , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
18.
J Biol Chem ; 291(12): 6331-46, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26769967

RESUMO

Sustained activation of PKCα is required for long term physiological responses, such as growth arrest and differentiation. However, studies with pharmacological agonists (e.g. phorbol 12-myristate 13-acetate (PMA)) indicate that prolonged stimulation leads to PKCα desensitization via dephosphorylation and/or degradation. The current study analyzed effects of chronic stimulation with the physiological agonist diacylglycerol. Repeated addition of 1,2-dioctanoyl-sn-glycerol (DiC8) resulted in sustained plasma membrane association of PKCα in a pattern comparable with that induced by PMA. However, although PMA potently down-regulated PKCα, prolonged activation by DiC8 failed to engage known desensitization mechanisms, with the enzyme remaining membrane-associated and able to support sustained downstream signaling. DiC8-activated PKCα did not undergo dephosphorylation, ubiquitination, or internalization, early events in PKCα desensitization. Although DiC8 efficiently down-regulated novel PKCs PKCδ and PKCϵ, differences in Ca(2+) sensitivity and diacylglycerol affinity were excluded as mediators of the selective resistance of PKCα. Roles for Hsp/Hsc70 and Hsp90 were also excluded. PMA, but not DiC8, targeted PKCα to detergent-resistant membranes, and disruption of these domains with cholesterol-binding agents demonstrated a role for differential membrane compartmentalization in selective agonist-induced degradation. Chronic DiC8 treatment failed to desensitize PKCα in several cell types and did not affect PKCßI; thus, conventional PKCs appear generally insensitive to desensitization by sustained diacylglycerol stimulation. Consistent with this conclusion, prolonged (several-day) membrane association/activation of PKCα is seen in self-renewing epithelium of the intestine, cervix, and skin. PKCα deficiency affects gene expression, differentiation, and tumorigenesis in these tissues, highlighting the importance of mechanisms that protect PKCα from desensitization in vivo.


Assuntos
Diglicerídeos/farmacologia , Proteína Quinase C-alfa/metabolismo , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Ativação Enzimática , Humanos , Mucosa Intestinal/enzimologia , Microdomínios da Membrana/enzimologia , Transporte Proteico , Proteólise , Ratos , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia
19.
Endocrinology ; 157(1): 54-60, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26523491

RESUMO

Supramolecular cup-shaped lipoprotein structures called porosomes embedded in the cell plasma membrane mediate fractional release of intravesicular contents from cells during secretion. The presence of porosomes, have been documented in many cell types including neurons, acinar cells of the exocrine pancreas, GH-secreting cells of the pituitary, and insulin-secreting pancreatic ß-cells. Functional reconstitution of porosomes into artificial lipid membranes, have also been accomplished. Earlier studies on mouse insulin-secreting Min6 cells report 100-nm porosome complexes composed of nearly 30 proteins. In the current study, porosomes have been functionally reconstituted for the first time in live cells. Isolated Min6 porosomes reconstituted into live Min6 cells demonstrate augmented levels of porosome proteins and a consequent increase in the potency and efficacy of glucose-stimulated insulin release. Elevated glucose-stimulated insulin secretion 48 hours after reconstitution, reflects on the remarkable stability and viability of reconstituted porosomes, documenting the functional reconstitution of native porosomes in live cells. These results, establish a new paradigm in porosome-mediated insulin secretion in ß-cells.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Microdomínios da Membrana/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Sintaxina 1/metabolismo , Animais , Linhagem Celular Tumoral , Hiperglicemia/sangue , Hiperglicemia/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/ultraestrutura , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/ultraestrutura , Camundongos , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Estabilidade Proteica , Transporte Proteico , Espalhamento a Baixo Ângulo , Taxa Secretória , Proteína 25 Associada a Sinaptossoma/isolamento & purificação , Difração de Raios X
20.
Cell Signal ; 28(2): 81-93, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26631574

RESUMO

Platelet derived growth factor receptors (PDGFR) play an important role in tumor pathogenesis and are frequently overexpressed in glioblastoma. Earlier we have shown that only confluent glioblastoma cell cultures exhibit a biphasic calcium transient upon PDGF stimulation. Here, we examined how the change in cell density leads to differential cellular responses to the same PDGF stimulus. PDGF beta receptors and their specific phosphotyrosine residues were fluorescently co-labeled on A172 and T98G glioblastoma cells. The distribution in cell membrane microdomains (lipid rafts) and the phosphorylation state of PDGFR was measured by confocal microscopy and quantitated by digital image processing. Corresponding bulk data were obtained by Western blotting. Activation of relevant downstream signaling pathways was assessed by immunofluorescence in confocal microscopy and by Western blot analysis. Functional outcomes were confirmed with bulk and single cell proliferation assays and motility measurements. In non-confluent (sparse) cultures PDGF-BB stimulation significantly increased phosphorylation of Tyr716 specific for the Ras/MAPK pathway and Tyr751 specific for the phosphoinositide 3-kinase/Akt pathway. As cell monolayers reached confluence, Tyr771 and Tyr1021 were the prominently phosphorylated residues. Tyr771 serves as adaptor for Ras-GAP, which inactivates the MAPK pathway, and Tyr1021 feeds into the phospholipase C-gamma/PKC pathway. Coherent with this, MAPK phosphorylation, Ki-67 positivity and proliferation dominated in dispersed cells, and could be abolished with inhibitors of the MAPK pathway. At the same time, RhoA activation, redistribution of cortactin to leading edges, and increased motility were the prominent output features in confluent cultures. Importantly, the stimulus-evoked confluence-specific changes in the phosphorylation of tyrosine residues occurred mainly in GM1-rich lipid microdomains (rafts). These observations suggest that the same stimulus is able to promote distinctly relevant signaling outputs through a confluence dependent, lipid raft-based regulatory mechanism. In particular, cell division and survival in sparse cultures and inhibition of proliferation and promotion of migration in confluent monolayers. In our model, the ability to switch the final output of the same stimulus as a function of cell density could be a key to the balance of proliferation and invasion in malignant glioblastoma.


Assuntos
Movimento Celular , Proliferação de Células , Glioblastoma/enzimologia , Sistema de Sinalização das MAP Quinases , Microdomínios da Membrana/enzimologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Linhagem Celular Tumoral , Glioblastoma/fisiopatologia , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/química , Fosfolipases Tipo C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA