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










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38252473

RESUMO

Fibroblast growth factor 2 (FGF2) exits cells by direct translocation across the plasma membrane, a type I pathway of unconventional protein secretion. This process is initiated by phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2)-dependent formation of highly dynamic FGF2 oligomers at the inner plasma membrane leaflet, inducing the formation of lipidic membrane pores. Cell surface heparan sulfate chains linked to glypican-1 (GPC1) capture FGF2 at the outer plasma membrane leaflet, completing FGF2 membrane translocation into the extracellular space. While the basic steps of this pathway are well understood, the molecular mechanism by which FGF2 oligomerizes on membrane surfaces remains unclear. In the current study, we demonstrate the initial step of this process to depend on C95-C95 disulfide-bridge-mediated FGF2 dimerization on membrane surfaces, producing the building blocks for higher FGF2 oligomers that drive the formation of membrane pores. We find FGF2 with a C95A substitution to be defective in oligomerization, pore formation, and membrane translocation. Consistently, we demonstrate a C95A variant of FGF2 to be characterized by a severe secretion phenotype. By contrast, while also important for efficient FGF2 secretion from cells, a second cysteine residue on the molecular surface of FGF2 (C77) is not involved in FGF2 oligomerization. Rather, we find C77 to be part of the interaction interface through which FGF2 binds to the α1 subunit of the Na,K-ATPase, the landing platform for FGF2 at the inner plasma membrane leaflet. Using cross-linking mass spectrometry, atomistic molecular dynamics simulations combined with a machine learning analysis and cryo-electron tomography, we propose a mechanism by which disulfide-bridged FGF2 dimers bind with high avidity to PI(4,5)P2 on membrane surfaces. We further propose a tight coupling between FGF2 secretion and the formation of ternary signaling complexes on cell surfaces, hypothesizing that C95-C95-bridged FGF2 dimers are functioning as the molecular units triggering autocrine and paracrine FGF2 signaling.


Assuntos
Espaço Extracelular , Fator 2 de Crescimento de Fibroblastos , Dimerização , ATPase Trocadora de Sódio-Potássio , Dissulfetos
2.
Elife ; 112022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35348113

RESUMO

Fibroblast growth factor 2 (FGF2) is a tumor cell survival factor that is transported into the extracellular space by an unconventional secretory mechanism. Cell surface heparan sulfate proteoglycans are known to play an essential role in this process. Unexpectedly, we found that among the diverse subclasses consisting of syndecans, perlecans, glypicans, and others, Glypican-1 (GPC1) is the principle and rate-limiting factor that drives unconventional secretion of FGF2. By contrast, we demonstrate GPC1 to be dispensable for FGF2 signaling into cells. We provide first insights into the structural basis for GPC1-dependent FGF2 secretion, identifying disaccharides with N-linked sulfate groups to be enriched in the heparan sulfate chains of GPC1 to which FGF2 binds with high affinity. Our findings have broad implications for the role of GPC1 as a key molecule in tumor progression.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Glipicanas , Membrana Celular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glipicanas/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Humanos
3.
Commun Biol ; 3(1): 141, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32214225

RESUMO

FGF2 is a tumor cell survival factor that is exported from cells by an ER/Golgi-independent secretory pathway. This unconventional mechanism of protein secretion is based on direct translocation of FGF2 across the plasma membrane. The Na,K-ATPase has previously been shown to play a role in this process, however, the underlying mechanism has remained elusive. Here, we define structural elements that are critical for a direct physical interaction between FGF2 and the α1 subunit of the Na,K-ATPase. In intact cells, corresponding FGF2 mutant forms were impaired regarding both recruitment at the inner plasma membrane leaflet and secretion. Ouabain, a drug that inhibits both the Na,K-ATPase and FGF2 secretion, was found to impair the interaction of FGF2 with the Na,K-ATPase in cells. Our findings reveal the Na,K-ATPase as the initial recruitment factor for FGF2 at the inner plasma membrane leaflet being required for efficient membrane translocation of FGF2 to cell surfaces.


Assuntos
Membrana Celular/enzimologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Células CHO , Cricetulus , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Sistemas do Segundo Mensageiro , Via Secretória , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/genética
4.
FEBS Lett ; 593(16): 2162-2176, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31135968

RESUMO

Fibroblast growth factor 2 (FGF2) is a cell survival factor with crucial functions in tumor-induced angiogenesis. Here, we describe a novel time-resolved FGF2 signaling assay based upon live cell imaging of neuroblastoma cells. To validate this system, we tested 8960 small molecules for inhibition of FGF2 signaling with kinetic resolution. Hit compounds were validated in dose-response experiments for FGF2 signaling, FGF receptor antagonism, downstream ERK phosphorylation and FGF2-dependent chemoresistance in a cellular leukemia model system. The new screening system for FGF2 signaling inhibitors has unique features, deselecting compounds with pleiotropic effects on cell proliferation and, along with the experimental pipeline reported, great potential for the discovery of new classes of FGF2 signaling inhibitors that block FGF2 dependent tumor cell survival.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Neuroblastoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neuroblastoma/tratamento farmacológico , Fosforilação , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores
5.
J Cell Biol ; 218(2): 683-699, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30470711

RESUMO

FGF2 is exported from cells by an unconventional secretory mechanism. Here, we directly visualized individual FGF2 membrane translocation events at the plasma membrane using live cell TIRF microscopy. This process was dependent on both PI(4,5)P2-mediated recruitment of FGF2 at the inner leaflet and heparan sulfates capturing FGF2 at the outer plasma membrane leaflet. By simultaneous imaging of both FGF2 membrane recruitment and the appearance of FGF2 at the cell surface, we revealed the kinetics of FGF2 membrane translocation in living cells with an average duration of ∼200 ms. Furthermore, we directly demonstrated FGF2 oligomers at the inner leaflet of living cells with a FGF2 dimer being the most prominent species. We propose this dimer to represent a key intermediate in the formation of higher FGF2 oligomers that form membrane pores and put forward a kinetic model explaining the mechanism by which membrane-inserted FGF2 oligomers serve as dynamic translocation intermediates during unconventional secretion of FGF2.


Assuntos
Membrana Celular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Animais , Células CHO , Cricetulus , Fator 2 de Crescimento de Fibroblastos/genética , Células HEK293 , Heparitina Sulfato/metabolismo , Humanos , Cinética , Microscopia de Fluorescência , Modelos Biológicos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Multimerização Proteica , Transporte Proteico , Via Secretória
6.
J Biol Chem ; 291(34): 17787-803, 2016 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-27382052

RESUMO

Fibroblast growth factor 2 (FGF2) is a potent mitogen promoting both tumor cell survival and tumor-induced angiogenesis. It is secreted by an unconventional secretory mechanism that is based upon direct translocation across the plasma membrane. Key steps of this process are (i) phosphoinositide-dependent membrane recruitment, (ii) FGF2 oligomerization and membrane pore formation, and (iii) extracellular trapping mediated by membrane-proximal heparan sulfate proteoglycans. Efficient secretion of FGF2 is supported by Tec kinase that stimulates membrane pore formation based upon tyrosine phosphorylation of FGF2. Here, we report the biochemical characterization of the direct interaction between FGF2 and Tec kinase as well as the identification of small molecules that inhibit (i) the interaction of FGF2 with Tec, (ii) tyrosine phosphorylation of FGF2 mediated by Tec in vitro and in a cellular context, and (iii) unconventional secretion of FGF2 from cells. We further demonstrate the specificity of these inhibitors for FGF2 because tyrosine phosphorylation of a different substrate of Tec is unaffected in their presence. Building on previous evidence using RNA interference, the identified compounds corroborate the role of Tec kinase in unconventional secretion of FGF2. In addition, they are valuable lead compounds with great potential for drug development aiming at the inhibition of FGF2-dependent tumor growth and metastasis.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Multimerização Proteica/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , Proteínas Tirosina Quinases/genética , Interferência de RNA
7.
PLoS One ; 11(4): e0153009, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27100999

RESUMO

Cell membranes contain hundreds to thousands of individual lipid species that are of structural importance but also specifically interact with proteins. Due to their highly controlled synthesis and role in signaling events sphingolipids are an intensely studied class of lipids. In order to investigate their metabolism and to study proteins interacting with sphingolipids, metabolic labeling based on photoactivatable sphingoid bases is the most straightforward approach. In order to monitor protein-lipid-crosslink products, sphingosine derivatives containing a reporter moiety, such as a radiolabel or a clickable group, are used. In normal cells, degradation of sphingoid bases via action of the checkpoint enzyme sphingosine-1-phosphate lyase occurs at position C2-C3 of the sphingoid base and channels the resulting hexadecenal into the glycerolipid biosynthesis pathway. In case the functionalized sphingosine looses the reporter moiety during its degradation, specificity towards sphingolipid labeling is maintained. In case degradation of a sphingosine derivative does not remove either the photoactivatable or reporter group from the resulting hexadecenal, specificity towards sphingolipid labeling can be achieved by blocking sphingosine-1-phosphate lyase activity and thus preventing sphingosine derivatives to be channeled into the sphingolipid-to-glycerolipid metabolic pathway. Here we report an approach using clustered, regularly interspaced, short palindromic repeats (CRISPR)-associated nuclease Cas9 to create a sphingosine-1-phosphate lyase (SGPL1) HeLa knockout cell line to disrupt the sphingolipid-to-glycerolipid metabolic pathway. We found that the lipid and protein compositions as well as sphingolipid metabolism of SGPL1 knock-out HeLa cells only show little adaptations, which validates these cells as model systems to study transient protein-sphingolipid interactions.


Assuntos
Aldeído Liases/genética , Metabolismo dos Lipídeos , Proteínas/metabolismo , Aldeído Liases/metabolismo , Animais , Células Cultivadas , Cromatografia em Camada Delgada , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Camundongos , Ligação Proteica
8.
J Proteome Res ; 14(8): 3229-38, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26189946

RESUMO

Recent evidence suggests that the extracellular protein milieu is much more complex than previously assumed as various secretome analyses from different cell types described the release of hundreds to thousands of proteins. The extracellular function of many of these proteins has yet to be determined particularly in the context of three-dimensional tissues with abundant cell-cell contacts. Toward this goal, we developed a strategy of dual SILAC labeling astrocytic cultures for in silico exclusion of unlabeled proteins from serum or neurons used for stimulation. For constitutive secretion, this strategy allowed the precise quantification of the extra-to-intracellular protein ratio of more than 2000 identified proteins. Ratios covered 4 orders of magnitude indicating that the intracellular vs extracellular contributions of different proteins can be variable. Functionally, the secretome of labeled forebrain astrocytic cultures specifically changed within hours after adding unlabeled, "physiological" forebrain neurons. "Nonphysiological" cerebellar hindbrain neurons, however, elicited a different, highly repulsive secretory response. Our data also suggest a significant association of constitutive secretion with the classical secretion pathway and regulated secretion with unconventional pathways. We conclude that quantitative proteomics can help to elucidate general principles of cellular secretion and provide functional insight into the abundant extracellular presence of proteins.


Assuntos
Aminoácidos/metabolismo , Comunicação Celular , Proteoma/metabolismo , Proteômica/métodos , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Western Blotting , Células Cultivadas , Marcação por Isótopo/métodos , Espectrometria de Massas , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Prosencéfalo/citologia , Ratos , Transdução de Sinais
9.
J Biol Chem ; 290(14): 8925-37, 2015 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-25694424

RESUMO

Fibroblast growth factor 2 (FGF2) is a key signaling molecule in tumor-induced angiogenesis. FGF2 is secreted by an unconventional secretory mechanism that involves phosphatidylinositol 4,5-bisphosphate-dependent insertion of FGF2 oligomers into the plasma membrane. This process is regulated by Tec kinase-mediated tyrosine phosphorylation of FGF2. Molecular interactions driving FGF2 monomers into membrane-inserted FGF2 oligomers are unknown. Here we identify two surface cysteines that are critical for efficient unconventional secretion of FGF2. They represent unique features of FGF2 as they are absent from all signal-peptide-containing members of the FGF protein family. We show that phosphatidylinositol 4,5-bisphosphate-dependent FGF2 oligomerization concomitant with the generation of membrane pores depends on FGF2 surface cysteines as either chemical alkylation or substitution with alanines impairs these processes. We further demonstrate that the FGF2 variant forms lacking the two surface cysteines are not secreted from cells. These findings were corroborated by experiments redirecting a signal-peptide-containing FGF family member from the endoplasmic reticulum/Golgi-dependent secretory pathway into the unconventional secretory pathway of FGF2. Cis elements known to be required for unconventional secretion of FGF2, including the two surface cysteines, were transplanted into a variant form of FGF4 without signal peptide. The resulting FGF4/2 hybrid protein was secreted by unconventional means. We propose that the formation of disulfide bridges drives membrane insertion of FGF2 oligomers as intermediates in unconventional secretion of FGF2.


Assuntos
Dissulfetos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Eletroforese em Gel de Poliacrilamida , Fator 2 de Crescimento de Fibroblastos/química , Dados de Sequência Molecular , Polimerização , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
10.
J Biol Chem ; 290(6): 3654-65, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25533462

RESUMO

Previous studies proposed a role for the Na/K-ATPase in unconventional secretion of fibroblast growth factor 2 (FGF2). This conclusion was based upon pharmacological inhibition of FGF2 secretion in the presence of ouabain. However, neither independent experimental evidence nor a potential mechanism was provided. Based upon an unbiased RNAi screen, we now report the identification of ATP1A1, the α1-chain of the Na/K-ATPase, as a factor required for efficient secretion of FGF2. As opposed to ATP1A1, down-regulation of the ß1- and ß3-chains (ATP1B1 and ATP1B3) of the Na/K-ATPase did not affect FGF2 secretion, suggesting that they are dispensable for this process. These findings indicate that it is not the membrane potential-generating function of the Na/K-ATPase complex but rather a so far unidentified role of potentially unassembled α1-chains that is critical for unconventional secretion of FGF2. Consistently, in the absence of ß-chains, we found a direct interaction between the cytoplasmic domain of ATP1A1 and FGF2 with submicromolar affinity. Based upon these observations, we propose that ATP1A1 is a recruitment factor for FGF2 at the inner leaflet of plasma membranes that may control phosphatidylinositol 4,5-bisphosphate-dependent membrane translocation as part of the unconventional secretory pathway of FGF2.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Via Secretória , ATPase Trocadora de Sódio-Potássio/metabolismo , Células HeLa , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/genética
11.
Genome Res ; 21(11): 1955-68, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21795383

RESUMO

SRC proteins are non-receptor tyrosine kinases that play key roles in regulating signal transduction by a diverse set of cell surface receptors. They contain N-terminal SH4 domains that are modified by fatty acylation and are functioning as membrane anchors. Acylated SH4 domains are both necessary and sufficient to mediate specific targeting of SRC kinases to the inner leaflet of plasma membranes. Intracellular transport of SRC kinases to the plasma membrane depends on microdomains into which SRC kinases partition upon palmitoylation. In the present study, we established a live-cell imaging screening system to identify gene products involved in plasma membrane targeting of SRC kinases. Based on siRNA arrays and a human model cell line expressing two kinds of SH4 reporter molecules, we conducted a genome-wide analysis of SH4-dependent protein targeting using an automated microscopy platform. We identified and validated 54 gene products whose down-regulation causes intracellular retention of SH4 reporter molecules. To detect and quantify this phenotype, we developed a software-based image analysis tool. Among the identified gene products, we found factors involved in lipid metabolism, intracellular transport, and cellular signaling processes. Furthermore, we identified proteins that are either associated with SRC kinases or are related to various known functions of SRC kinases such as other kinases and phosphatases potentially involved in SRC-mediated signal transduction. Finally, we identified gene products whose function is less defined or entirely unknown. Our findings provide a major resource for future studies unraveling the molecular mechanisms that underlie proper targeting of SRC kinases to the inner leaflet of plasma membranes.


Assuntos
Membrana Celular/enzimologia , Genoma Humano , Fenótipo , Quinases da Família src/metabolismo , Linhagem Celular Tumoral , Proteína Coatomer/genética , Proteína Coatomer/metabolismo , Células HeLa , Ensaios de Triagem em Larga Escala , Homeostase , Humanos , Espaço Intracelular/metabolismo , Metabolismo dos Lipídeos , Lipoilação , Domínios e Motivos de Interação entre Proteínas , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-yes/metabolismo , RNA Interferente Pequeno , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Quinases da Família src/genética
12.
Traffic ; 11(6): 813-26, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20230531

RESUMO

Fibroblast growth factor 2 (FGF2) is a potent mitogen that is exported from cells by an endoplasmic reticulum (ER)/Golgi-independent mechanism. Unconventional secretion of FGF2 occurs by direct translocation across plasma membranes, a process that depends on the phosphoinositide phosphatidylinositol 4,5-biphosphate (PI(4,5)P(2)) at the inner leaflet as well as heparan sulfate proteoglycans at the outer leaflet of plasma membranes; however, additional core and regulatory components of the FGF2 export machinery have remained elusive. Here, using a highly effective RNAi screening approach, we discovered Tec kinase as a novel factor involved in unconventional secretion of FGF2. Tec kinase does not affect FGF2 secretion by an indirect mechanism, but rather forms a heterodimeric complex with FGF2 resulting in phosphorylation of FGF2 at tyrosine 82, a post-translational modification shown to be essential for FGF2 membrane translocation to cell surfaces. Our findings suggest a crucial role for Tec kinase in regulating FGF2 secretion under various physiological conditions and, therefore, provide a new perspective for the development of a novel class of antiangiogenic drugs targeting the formation of the FGF2/Tec complex.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Células 3T3 , Animais , Membrana Celular/metabolismo , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Fosfatidilinositol 4,5-Difosfato/química , Fosforilação , Transporte Proteico , Proteínas/química , Interferência de RNA , Tirosina/química
13.
Traffic ; 10(8): 1047-60, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19453972

RESUMO

Acylated SH4 domains represent N-terminal targeting signals that anchor peripheral membrane proteins such as Src kinases in the inner leaflet of plasma membranes. Here we provide evidence for a novel regulatory mechanism that may control the levels of SH4 proteins being associated with plasma membranes. Using a fusion protein of the SH4 domain of Leishmania HASPB and GFP as a model system, we demonstrate that threonine 6 is a substrate for phosphorylation. Substitution of threonine 6 by glutamate (to mimic a phosphothreonine residue) resulted in a dramatic redistribution from plasma membranes to intracellular sites with a particular accumulation in a perinuclear region. As shown by both pharmacological inhibition and RNAi-mediated down-regulation of the threonine/ serine-specific phosphatases PP1 and PP2A, recycling back to the plasma membrane required dephosphorylation of threonine 6. We provide evidence that a cycle of phosphorylation and dephosphorylation may also be involved in intracellular targeting of other SH4 proteins such as the Src kinase Yes.


Assuntos
Antígenos de Protozoários/metabolismo , Membrana Celular/metabolismo , Transporte Proteico/fisiologia , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Antígenos de Protozoários/genética , Células CHO , Cricetinae , Cricetulus , Endossomos/metabolismo , Endossomos/ultraestrutura , Inibidores Enzimáticos/metabolismo , Ácido Glutâmico/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Leishmania/metabolismo , Toxinas Marinhas , Mutação , Oxazóis/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas c-yes/metabolismo , Proteínas de Protozoários/genética , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Treonina/metabolismo
14.
FEBS Lett ; 582(16): 2387-92, 2008 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-18538671

RESUMO

FGF-2 is a proangiogenic growth factor secreted by unconventional means. It is unknown why FGF-2 takes an ER/Golgi-independent secretory route. We find that secretion of FGF-2 via the ER/Golgi system causes post-translational modifications that prevent binding to heparan sulfate proteoglycans (HSPGs), an interaction that is critically important for both FGF-2 storage and signal transduction. This loss of function is due to artificial O-glycosylation mainly resulting in the addition of glycosaminoglycan chains of the chrondroitin sulfate type. Our findings suggest that the unconventional mechanism of FGF-2 export is an ancient pathway of protein secretion that, in the course of evolution, has been kept due to the inability of the classical secretory pathway to export FGF-2 in a functional form.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Células CHO , Sulfatos de Condroitina/metabolismo , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Glicosilação , Complexo de Golgi/metabolismo , Heparina/metabolismo , Ligação Proteica , Sinais Direcionadores de Proteínas
15.
Proc Natl Acad Sci U S A ; 103(42): 15479-84, 2006 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-17030799

RESUMO

FGF-2 is an unconventionally secreted lectin that transmits proangiogenic signals through a ternary complex with high-affinity FGF receptors and heparan sulfate proteoglycans (HSPGs). Although FGF-2 signal transduction is understood in great detail, its mechanism of release from cells, which is independent of the classical secretory pathway, remains elusive. To test the hypothesis that FGF-2 secretion is linked to its cell-surface ligands, we studied FGF-2 release using mutants defective for HSPG binding and cells with impaired HSPG biosynthesis. Here, we report that a functional interaction between FGF-2 and HSPGs is required for net export of FGF-2 from mammalian cells. FGF-2 release requires extracellular, membrane-proximal HSPGs. We propose that extracellular HSPGs form a molecular trap that drives FGF-2 translocation across the plasma membrane.


Assuntos
Membrana Celular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Células CHO , Membrana Celular/química , Técnicas de Cocultura , Cricetinae , Fator 2 de Crescimento de Fibroblastos/genética , Proteoglicanas de Heparan Sulfato/química , Ligação Proteica , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/fisiologia
16.
J Cell Biol ; 171(2): 373-81, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16247033

RESUMO

Galectin-1 is a component of the extracellular matrix as well as a ligand of cell surface counter receptors such as beta-galactoside-containing glycolipids, however, the molecular mechanism of galectin-1 secretion has remained elusive. Based on a nonbiased screen for galectin-1 export mutants we have identified 26 single amino acid changes that cause a defect of both export and binding to counter receptors. When wild-type galectin-1 was analyzed in CHO clone 13 cells, a mutant cell line incapable of expressing functional galectin-1 counter receptors, secretion was blocked. Intriguingly, we also find that a distant relative of galectin-1, the fungal lectin CGL-2, is a substrate for nonclassical export from Chinese hamster ovary (CHO) cells. Alike mammalian galectin-1, a CGL-2 mutant defective in beta-galactoside binding, does not get exported from CHO cells. We conclude that the beta-galactoside binding site represents the primary targeting motif of galectins defining a galectin export machinery that makes use of beta-galactoside-containing surface molecules as export receptors for intracellular galectin-1.


Assuntos
Galectina 1/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Células CHO , Cricetinae , Proteínas Fúngicas/metabolismo , Galactosídeos/química , Galactosídeos/metabolismo , Galectina 1/genética , Galectina 1/farmacologia , Galectina 2 , Galectinas/metabolismo , Regulação da Expressão Gênica , Glicolipídeos/antagonistas & inibidores , Glicolipídeos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Ligantes , Mutação , Transporte Proteico/fisiologia , Receptores de Superfície Celular/antagonistas & inibidores
17.
J Cell Sci ; 118(Pt 3): 517-27, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15657075

RESUMO

Leishmania HASPB is a lipoprotein that is exported to the extracellular space from both Leishmania parasites and mammalian cells via an unconventional secretory pathway. Exported HASPB remains anchored in the outer leaflet of the plasma membrane mediated by myristate and palmitate residues covalently attached to the N-terminal SH4 domain of HASPB. HASPB targeting to the plasma membrane depends on SH4 acylation that occurs at intracellular membranes. How acylated HASPB is targeted to the plasma membrane and, in particular, the subcellular site of HASPB membrane translocation is unknown. In order to address this issue, we screened for clonal CHO mutants that are incapable of exporting HASPB. A detailed characterization of such a CHO mutant cell line revealed that the expression level of the HASPB reporter molecule is unchanged compared to CHO wild-type cells; that it is both myristoylated and palmitoylated; and that it is mainly localized to the plasma membrane as judged by confocal microscopy and subcellular fractionation. However, based on a quantitative flow cytometry assay and a biochemical biotinylation assay of surface proteins, HASPB transport to the outer leaflet of the plasma membrane is largely reduced in this mutant. From these data, we conclude that the subcellular site of HASPB membrane translocation is the plasma membrane as the reporter molecule accumulates in this location when export is blocked. Thus, these results allow us to define a two-step process of HASPB cell surface biogenesis in which SH4 acylation of HASPB firstly mediates intracellular targeting to the plasma membrane. In a second step, the plasma membrane-resident machinery, which is apparently disrupted in the CHO mutant cell line, mediates membrane translocation of HASPB. Intriguingly, the angiogenic growth factor FGF-2, another protein secreted by unconventional means, is shown to be secreted normally from the HASPB export mutant cell line. These observations demonstrate that the export machinery component defective in the export mutant cell line functions specifically in the HASPB export pathway.


Assuntos
Antígenos de Protozoários/metabolismo , Membrana Celular/metabolismo , Mutação , Proteínas de Protozoários/metabolismo , Acilação , Animais , Antígenos de Protozoários/genética , Biotinilação , Células CHO , Membrana Celular/química , Cricetinae , Cricetulus , Citosol/química , Doxiciclina/farmacologia , Ácidos Graxos/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Membranas Intracelulares/química , Leishmania/fisiologia , Proteínas de Membrana/análise , Mutagênese Insercional , Parasitos/fisiologia , Fragmentos de Peptídeos/genética , Transporte Proteico , Proteínas de Protozoários/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética
18.
J Cell Sci ; 117(Pt 9): 1727-36, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15075234

RESUMO

Endoplasmic reticulum/Golgi-dependent protein secretion depends on signal peptides that mediate membrane translocation of nascent secretory proteins into the lumen of the endoplasmic reticulum. Classical secretory proteins are transported across the membrane of the endoplasmic reticulum in an unfolded conformation, which is similar to protein import into mitochondria. This process is mediated by Sec61, the protein-conducting channel of the endoplasmic reticulum. Employing both FACS-based in vivo transport assays and confocal microscopy, we now show that fibroblast growth factor 2 (FGF-2), a pro-angiogenic mediator exported from mammalian cells by an unconventional secretory pathway, does not need to be unfolded in order to be released into the extracellular space. These findings suggest that the molecular apparatus mediating export of FGF-2 is not only distinct from classical translocation machineries in terms of molecular identity but also operates in a mechanistically distinct manner that allows membrane translocation of FGF-2 in a folded conformation.


Assuntos
Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/metabolismo , Membranas Intracelulares/metabolismo , Dobramento de Proteína , Aminopterina/farmacologia , Animais , Células CHO , Cricetinae , Fator 2 de Crescimento de Fibroblastos/genética , Citometria de Fluxo , Camundongos , Microscopia Confocal , Mitocôndrias/metabolismo , Modelos Biológicos , Conformação Proteica , Desnaturação Proteica , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Transfecção
19.
J Cell Sci ; 116(Pt 7): 1305-18, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12615972

RESUMO

CA125 is an ovarian cancer antigen whose recently elucidated primary structure suggests that CA125 is a giant mucin-like glycoprotein present on the cell surface of tumor cells. Here, we establish a functional link between CA125 and beta-galactoside-binding, cell-surface lectins, which are components of the extracellular matrix implicated in the regulation of cell adhesion, apoptosis, cell proliferation and tumor progression. On the basis of mass spectrometry and immunological analyses, we find that CA125 is a counter receptor for galectin-1, as both soluble and membrane-associated fragments of CA125 derived from HeLa cell lysates are shown to bind specifically to human galectin-1 with high efficiency. This interaction is demonstrated (1) to depend on beta-galactose-terminated, O-linked oligosaccharide chains of CA125, (2) to be preferential for galectin-1 versus galectin-3 and (3) to be regulated by the cellular background in which CA125 is expressed. Despite lacking a conventional signal peptide, a CA125 C-terminal fragment of 1148 amino acids, representing less than 10% of the full-length protein, retains the ability to integrate into secretory membranes such as the endoplasmic reticulum (ER) and the Golgi, and is targeted to the plasma membrane by conventional secretory transport. As demonstrated by a novel assay that reconstitutes non-conventional secretion of galectin-1 based on fluorescence-activated cell sorting (FACS), we find that tumor-derived HeLa cells expressing endogenous CA125 present more than ten times as much galectin-1 on their surface compared with non-tumor-derived, CA125-deficient CHO cells. Intriguingly, both the galectin-1 expression level and the cell-surface binding capacity for galectin-1 are shown to be similar in CHO and HeLa cells, suggesting that CA125 might be a factor involved in the regulation of galectin-1 export to the cell surface.


Assuntos
Antígenos de Superfície/metabolismo , Antígeno Ca-125/metabolismo , Galectina 1/metabolismo , Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos/fisiologia , Animais , Sítios de Ligação/fisiologia , Células CHO , Membrana Celular/metabolismo , Cricetinae , Galectina 3/metabolismo , Células HeLa , Humanos , Neoplasias/diagnóstico , Oligossacarídeos de Cadeias Ramificadas/metabolismo , Fragmentos de Peptídeos/fisiologia , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia
20.
J Cell Sci ; 115(Pt 18): 3619-31, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12186948

RESUMO

Basic fibroblast growth factor (FGF-2) is a secretory protein that lacks a signal peptide. Consistently, FGF-2 has been shown to be secreted by an ER-Golgi-independent mechanism; however, the machinery mediating this process remains to be established at the molecular level. Here we introduce a novel experimental system based on flow cytometry that allows the quantitative assessment of non-classical FGF-2 secretion in living cells. Stable cell lines have been created by retroviral transduction that express various kinds of FGF-2-GFP fusion proteins in a doxicyclin-dependent manner. Following induction of protein expression, biosynthetic FGF-2-GFP is shown to translocate to the outer surface of the plasma membrane as determined by both fluorescence activated cell sorting (FACS) and confocal microscopy. Both N- and C-terminal GFP tagging of FGF-2 is compatible with FGF-2 export, which is shown to occur in a controlled fashion rather than through unspecific release. The experimental system described has strong implications for the identification of both FGF-2 secretion inhibitors and molecular components involved in FGF-2 secretion. In the second part of this study we made use of the FGF-2 export system described to analyze the fate of biosynthetic FGF-2-GFP following export to the extracellular space. We find that secreted FGF-2 fusion proteins accumulate in large heparan sulfate proteoglycan (HSPG)-containing protein clusters on the extracellular surface of the plasma membrane. These microdomains are shown to be distinct from caveolae-like lipid rafts known to play a role in FGF-2-mediated signal transduction. Since CHO cells lack FGF high-affinity receptors (FGFRs), it can be concluded that FGFRs mediate the targeting of FGF-2 to lipid rafts. Consistently, FGF-2-GFP-secreting CHO cells do not exhibit increased proliferation activity. Externalization and deposition of biosynthetic FGF-2 in HSPG-containing protein clusters are independent processes, as a soluble secreted intermediate was demonstrated. The balance between intracellular FGF-2 and HSPG-bound secreted FGF-2 is shown not to be controlled by the availability of cell surface HSPGs, indicating that the FGF-2 secretion machinery itself is rate-limiting.


Assuntos
Células CHO/metabolismo , Membrana Celular/metabolismo , Células Eucarióticas/metabolismo , Espaço Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glicoproteínas de Membrana/metabolismo , Animais , Células CHO/citologia , Comunicação Celular/genética , Membrana Celular/genética , Cricetinae , Células Eucarióticas/citologia , Espaço Extracelular/genética , Fator 2 de Crescimento de Fibroblastos/genética , Vetores Genéticos , Proteínas de Fluorescência Verde , Proteoglicanas de Heparan Sulfato/metabolismo , Proteínas Luminescentes , Glicoproteínas de Membrana/genética , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Microscopia Confocal , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...