Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(21)2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37947636

RESUMO

T cells can express multiple inhibitory receptors. Upon induction of T cell exhaustion in response to a persistent antigen, prominently in the anti-tumor immune response, many are expressed simultaneously. Key inhibitory receptors are CTLA-4, PD-1, LAG3, TIM3, and TIGIT, as investigated here. These receptors are important as central therapeutic targets in cancer immunotherapy. Inhibitory receptors are not constitutively expressed on the cell surface, but substantial fractions reside in intracellular vesicular structures. It remains unresolved to which extent the subcellular localization of different inhibitory receptors is distinct. Using quantitative imaging of subcellular distributions and plasma membrane insertion as complemented by proximity proteomics and biochemical analysis of the association of the inhibitory receptors with trafficking adaptors, the subcellular distributions of the five inhibitory receptors were discrete. The distribution of CTLA-4 was most distinct, with preferential association with lysosomal-derived vesicles and the sorting nexin 1/2/5/6 transport machinery. With a lack of evidence for the existence of specific vesicle subtypes to explain divergent inhibitory receptor distributions, we suggest that such distributions are driven by divergent trafficking through an overlapping joint set of vesicular structures. This extensive characterization of the subcellular localization of five inhibitory receptors in relation to each other lays the foundation for the molecular investigation of their trafficking and its therapeutic exploitation.


Assuntos
Neoplasias , Linfócitos T , Camundongos , Animais , Antígeno CTLA-4/metabolismo , Proteínas de Transporte/metabolismo , Neoplasias/metabolismo , Imunoterapia
2.
bioRxiv ; 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37503045

RESUMO

T cells can express multiple inhibitory receptors. Upon induction of T cell exhaustion in response to persistent antigen, prominently in the anti-tumor immune response, many are expressed simultaneously. Key inhibitory receptors are CTLA-4, PD-1, LAG3, TIM3 and TIGIT, as investigated here. These receptors are important as central therapeutic targets in cancer immunotherapy. Inhibitory receptors are not constitutively expressed on the cell surface, but substantial fractions reside in intracellular vesicular structures. It remains unresolved to which extent the subcellular localization of different inhibitory receptors is distinct. Using quantitative imaging of subcellular distributions and plasma membrane insertion as complemented by proximity proteomics and a biochemical analysis of the association of the inhibitory receptors with trafficking adaptors, the subcellular distributions of the five inhibitory receptors were discrete. The distribution of CTLA-4 was most distinct with preferential association with lysosomal-derived vesicles and the sorting nexin 1/2/5/6 transport machinery. With a lack of evidence for the existence of specific vesicle subtypes to explain divergent inhibitory receptor distributions, we suggest that such distributions are driven by divergent trafficking through an overlapping joint set of vesicular structures. This extensive characterization of the subcellular localization of five inhibitory receptors in relation to each other lays the foundation for the molecular investigation of their trafficking and its therapeutic exploitation.

3.
Science ; 370(6518): 861-865, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33082294

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), uses the viral spike (S) protein for host cell attachment and entry. The host protease furin cleaves the full-length precursor S glycoprotein into two associated polypeptides: S1 and S2. Cleavage of S generates a polybasic Arg-Arg-Ala-Arg carboxyl-terminal sequence on S1, which conforms to a C-end rule (CendR) motif that binds to cell surface neuropilin-1 (NRP1) and NRP2 receptors. We used x-ray crystallography and biochemical approaches to show that the S1 CendR motif directly bound NRP1. Blocking this interaction by RNA interference or selective inhibitors reduced SARS-CoV-2 entry and infectivity in cell culture. NRP1 thus serves as a host factor for SARS-CoV-2 infection and may potentially provide a therapeutic target for COVID-19.


Assuntos
Betacoronavirus/fisiologia , Neuropilina-1/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Motivos de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , COVID-19 , Células CACO-2 , Infecções por Coronavirus/virologia , Cristalografia por Raios X , Furina/metabolismo , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/química , Neuropilina-1/genética , Pandemias , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/virologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Interferência de RNA , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética
4.
J Cell Sci ; 133(16)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843536

RESUMO

The concentration of essential micronutrients, such as copper (used here to describe both Cu+ and Cu2+), within the cell is tightly regulated to avoid their adverse deficiency and toxicity effects. Retromer-mediated sorting and recycling of nutrient transporters within the endo-lysosomal network is an essential process in regulating nutrient balance. Cellular copper homeostasis is regulated primarily by two transporters: the copper influx transporter copper transporter 1 (CTR1; also known as SLC31A1), which controls the uptake of copper, and the copper-extruding ATPase ATP7A, a recognised retromer cargo. Here, we show that in response to fluctuating extracellular copper, retromer controls the delivery of CTR1 to the cell surface. Following copper exposure, CTR1 is endocytosed to prevent excessive copper uptake. We reveal that internalised CTR1 localises on retromer-positive endosomes and, in response to decreased extracellular copper, retromer controls the recycling of CTR1 back to the cell surface to maintain copper homeostasis. In addition to copper, CTR1 plays a central role in the trafficking of platinum. The efficacy of platinum-based cancer drugs has been correlated with CTR1 expression. Consistent with this, we demonstrate that retromer-deficient cells show reduced sensitivity to the platinum-based drug cisplatin.


Assuntos
Proteínas de Transporte de Cátions , Cobre , Animais , Proteínas de Transporte de Cátions/genética , Cisplatino , Cobre/metabolismo , Transportador de Cobre 1 , Homeostase
5.
Nat Commun ; 9(1): 3737, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30213940

RESUMO

Wntless transports Wnt morphogens to the cell surface and is required for Wnt secretion and morphogenic gradients formation. Recycling of endocytosed Wntless requires the sorting nexin-3 (SNX3)-retromer-dependent endosome-to-Golgi transport pathway. Here we demonstrate the essential role of SNX3-retromer assembly for Wntless transport and report that SNX3 associates with an evolutionary conserved endosome-associated membrane re-modelling complex composed of MON2, DOPEY2 and the putative aminophospholipid translocase, ATP9A. In vivo suppression of Ce-mon-2, Ce-pad-1 or Ce-tat-5 (respective MON2, DOPEY2 and ATP9A orthologues) phenocopy a loss of SNX3-retromer function, leading to enhanced lysosomal degradation of Wntless and a Wnt phenotype. Perturbed Wnt signalling is also observed upon overexpression of an ATPase-inhibited TAT-5(E246Q) mutant, suggesting a role for phospholipid flippase activity during SNX3-retromer-mediated Wntless sorting. Together, these findings provide in vitro and in vivo mechanistic details to describe SNX3-retromer-mediated transport during Wnt secretion and the formation of Wnt-morphogenic gradients.


Assuntos
Adenosina Trifosfatases/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Nexinas de Classificação/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas Wnt/metabolismo , Animais , Transporte Biológico , Caenorhabditis elegans , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Mutação , Fenótipo , Ligação Proteica , Domínios Proteicos , Proteômica , Interferência de RNA , Transgenes
6.
Nat Cell Biol ; 19(10): 1214-1225, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28892079

RESUMO

Following endocytosis into the endosomal network, integral membrane proteins undergo sorting for lysosomal degradation or are retrieved and recycled back to the cell surface. Here we describe the discovery of an ancient and conserved multiprotein complex that orchestrates cargo retrieval and recycling and, importantly, is biochemically and functionally distinct from the established retromer pathway. We have called this complex 'retriever'; it is a heterotrimer composed of DSCR3, C16orf62 and VPS29, and bears striking similarity to retromer. We establish that retriever associates with the cargo adaptor sorting nexin 17 (SNX17) and couples to CCC (CCDC93, CCDC22, COMMD) and WASH complexes to prevent lysosomal degradation and promote cell surface recycling of α5ß1 integrin. Through quantitative proteomic analysis, we identify over 120 cell surface proteins, including numerous integrins, signalling receptors and solute transporters, that require SNX17-retriever to maintain their surface levels. Our identification of retriever establishes a major endosomal retrieval and recycling pathway.


Assuntos
Membrana Celular/metabolismo , Endossomos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Modelos Moleculares , Complexos Multiproteicos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Transporte Proteico , Proteínas/química , Proteínas/genética , Proteólise , Proteômica/métodos , Interferência de RNA , Nexinas de Classificação/genética , Nexinas de Classificação/metabolismo , Relação Estrutura-Atividade , Transfecção , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
7.
J Cell Sci ; 130(2): 382-395, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27909246

RESUMO

Retromer and the associated actin-polymerizing WASH complex are essential for the endocytic recycling of a wide range of integral membrane proteins. A hereditary Parkinson's-disease-causing point mutation (D620N) in the retromer subunit VPS35 perturbs retromer's association with the WASH complex and also with the uncharacterized protein ankyrin-repeat-domain-containing protein 50 (ANKRD50). Here, we firmly establish ANKRD50 as a new and essential component of the SNX27-retromer-WASH super complex. Depletion of ANKRD50 in HeLa or U2OS cells phenocopied the loss of endosome-to-cell-surface recycling of multiple transmembrane proteins seen upon suppression of SNX27, retromer or WASH-complex components. Mass-spectrometry-based quantification of the cell surface proteome of ANKRD50-depleted cells identified amino acid transporters of the SLC1A family, among them SLC1A4, as additional cargo molecules that depend on ANKRD50 and retromer for their endocytic recycling. Mechanistically, we show that ANKRD50 simultaneously engages multiple parts of the SNX27-retromer-WASH complex machinery in a direct and co-operative interaction network that is needed to efficiently recycle the nutrient transporters GLUT1 (also known as SLC2A1) and SLC1A4, and potentially many other surface proteins.


Assuntos
Sistema ASC de Transporte de Aminoácidos/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Mapas de Interação de Proteínas , Proteínas de Transporte Vesicular/metabolismo , Motivos de Aminoácidos , Transporte Biológico , Membrana Celular/metabolismo , Endocitose , Endossomos/metabolismo , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Integrinas/metabolismo , Espectrometria de Massas , Fosfoproteínas Fosfatases/química , Ligação Proteica , Proteólise , Proteômica , Nexinas de Classificação/metabolismo , Transferrina/metabolismo , Proteínas de Transporte Vesicular/química
8.
J Biol Chem ; 292(5): 1691-1704, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-27903653

RESUMO

The class I PI3K family of lipid kinases plays an important role in integrin αIIbß3 function, thereby supporting thrombus growth and consolidation. Here, we identify Ras/Rap1GAP Rasa3 (GAP1IP4BP) as a major phosphatidylinositol 3,4,5-trisphosphate-binding protein in human platelets and a key regulator of integrin αIIbß3 outside-in signaling. We demonstrate that cytosolic Rasa3 translocates to the plasma membrane in a PI3K-dependent manner upon activation of human platelets. Expression of wild-type Rasa3 in integrin αIIbß3-expressing CHO cells blocked Rap1 activity and integrin αIIbß3-mediated spreading on fibrinogen. In contrast, Rap1GAP-deficient (P489V) and Ras/Rap1GAP-deficient (R371Q) Rasa3 had no effect. We furthermore show that two Rasa3 mutants (H794L and G125V), which are expressed in different mouse models of thrombocytopenia, lack both Ras and Rap1GAP activity and do not affect integrin αIIbß3-mediated spreading of CHO cells on fibrinogen. Platelets from thrombocytopenic mice expressing GAP-deficient Rasa3 (H794L) show increased spreading on fibrinogen, which in contrast to wild-type platelets is insensitive to PI3K inhibitors. Together, these results support an important role for Rasa3 in PI3K-dependent integrin αIIbß3-mediated outside-in signaling and cell spreading.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Substituição de Aminoácidos/genética , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Células CHO , Cricetinae , Cricetulus , Modelos Animais de Doenças , Proteínas Ativadoras de GTPase/genética , Humanos , Camundongos , Camundongos Mutantes , Mutação de Sentido Incorreto , Fosfatidilinositol 3-Quinases/genética , Fosfatos de Fosfatidilinositol/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Receptores Citoplasmáticos e Nucleares/genética , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patologia
9.
Nat Med ; 22(1): 54-63, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26618722

RESUMO

Mitochondrial dysfunction represents a critical step during the pathogenesis of Parkinson's disease (PD), and increasing evidence suggests abnormal mitochondrial dynamics and quality control as important underlying mechanisms. The VPS35 gene, which encodes a key component of the membrane protein-recycling retromer complex, is the third autosomal-dominant gene associated with PD. However, how VPS35 mutations lead to neurodegeneration remains unclear. Here we demonstrate that PD-associated VPS35 mutations caused mitochondrial fragmentation and cell death in cultured neurons in vitro, in mouse substantia nigra neurons in vivo and in human fibroblasts from an individual with PD who has the VPS35(D620N) mutation. VPS35-induced mitochondrial deficits and neuronal dysfunction could be prevented by inhibition of mitochondrial fission. VPS35 mutants showed increased interaction with dynamin-like protein (DLP) 1, which enhanced turnover of the mitochondrial DLP1 complexes via the mitochondria-derived vesicle-dependent trafficking of the complexes to lysosomes for degradation. Notably, oxidative stress increased the VPS35-DLP1 interaction, which we also found to be increased in the brains of sporadic PD cases. These results revealed a novel cellular mechanism for the involvement of VPS35 in mitochondrial fission, dysregulation of which is probably involved in the pathogenesis of familial, and possibly sporadic, PD.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/metabolismo , Doença de Parkinson/genética , Proteínas de Transporte Vesicular/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Linhagem Celular Tumoral , Dinaminas/metabolismo , Feminino , Recuperação de Fluorescência Após Fotodegradação , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Técnicas In Vitro , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Mitocôndrias/patologia , Neurônios , Estresse Oxidativo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Ratos , Substância Negra/citologia , Imagem com Lapso de Tempo
10.
J Cell Sci ; 127(Pt 22): 4940-53, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25278552

RESUMO

Retromer is a protein assembly that orchestrates the sorting of transmembrane cargo proteins into endosome-to-Golgi and endosome-to-plasma-membrane transport pathways. Here, we have employed quantitative proteomics to define the interactome of human VPS35, the core retromer component. This has identified a number of new interacting proteins, including ankyrin-repeat domain 50 (ANKRD50), seriologically defined colon cancer antigen 3 (SDCCAG3) and VPS9-ankyrin-repeat protein (VARP, also known as ANKRD27). Depletion of these proteins resulted in trafficking defects of retromer-dependent cargo, but differential and cargo-specific effects suggested a surprising degree of functional heterogeneity in retromer-mediated endosome-to-plasma-membrane sorting. Extending this, suppression of the retromer-associated WASH complex did not uniformly affect retromer cargo, thereby confirming cargo-specific functions for retromer-interacting proteins. Further analysis of the retromer-VARP interaction identified a role for retromer in endosome-to-melanosome transport. Suppression of VPS35 led to mistrafficking of the melanogenic enzymes, tyrosinase and tryrosine-related protein 1 (Tyrp1), establishing that retromer acts in concert with VARP in this trafficking pathway. Overall, these data reveal hidden complexities in retromer-mediated sorting and open up new directions in our molecular understanding of this essential sorting complex.


Assuntos
Endossomos/metabolismo , Nexinas de Classificação/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Membrana Celular/metabolismo , Células HeLa , Humanos , Transporte Proteico , Transfecção , Transferrina/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
11.
Curr Biol ; 24(14): 1670-1676, 2014 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-24980502

RESUMO

Retromer is a protein assembly that plays a central role in orchestrating export of transmembrane-spanning cargo proteins from endosomes into retrieval pathways destined for the Golgi apparatus and the plasma membrane [1]. Recently, a specific mutation in the retromer component VPS35, VPS35(D620N), has linked retromer dysfunction to familial autosomal dominant and sporadic Parkinson disease [2, 3]. However, the effect of this mutation on retromer function remains poorly characterized. Here we established that in cells expressing VPS35(D620N) there is a perturbation in endosome-to-TGN transport but not endosome-to-plasma membrane recycling, which we confirm in patient cells harboring the VPS35(D620N) mutation. Through comparative stable isotope labeling by amino acids in cell culture (SILAC)-based analysis of wild-type VPS35 versus the VPS35(D620N) mutant interactomes, we establish that the major defect of the D620N mutation lies in the association to the actin-nucleating Wiskott-Aldrich syndrome and SCAR homolog (WASH) complex. Moreover, using isothermal calorimetry, we establish that the primary defect of the VPS35(D620N) mutant is a 2.2 ± 0.5-fold decrease in affinity for the WASH complex component FAM21. These data define the primary molecular defect in retromer assembly that arises from the VPS35(D620N) mutation and, by revealing functional effects on retromer-mediated endosome-to-TGN transport, provide new insight into retromer deregulation in Parkinson disease.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Doença de Parkinson/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Repetição de Anquirina/genética , Antígenos de Neoplasias/metabolismo , Sítios de Ligação/genética , Linhagem Celular Tumoral , Células Cultivadas , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Chaperonas Moleculares/metabolismo , Mutação , Ligação Proteica/genética , Transporte Proteico
12.
J Cell Sci ; 126(Pt 21): 4885-99, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23986476

RESUMO

Sorting nexins (SNXs) are key regulators of the endosomal network. In designing an RNAi-mediated loss-of-function screen, we establish that of 30 human SNXs only SNX3, SNX5, SNX9, SNX15 and SNX21 appear to regulate EGF receptor degradative sorting. Suppression of SNX15 results in a delay in receptor degradation arising from a defect in movement of newly internalised EGF-receptor-labelled vesicles into early endosomes. Besides a phosphatidylinositol 3-phosphate- and PX-domain-dependent association to early endosomes, SNX15 also associates with clathrin-coated pits and clathrin-coated vesicles by direct binding to clathrin through a non-canonical clathrin-binding box. From live-cell imaging, it was identified that the activated EGF receptor enters distinct sub-populations of SNX15- and APPL1-labelled peripheral endocytic vesicles, which do not undergo heterotypic fusion. The SNX15-decorated receptor-containing sub-population does, however, undergo direct fusion with the Rab5-labelled early endosome. Our data are consistent with a model in which the EGF receptor enters the early endosome following clathrin-mediated endocytosis through at least two parallel pathways: maturation through an APPL1-intermediate compartment and an alternative more direct fusion between SNX15-decorated endocytic vesicles and the Rab5-positive early endosome.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Clatrina/metabolismo , Endocitose , Endossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Nexinas de Classificação/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Clatrina/genética , Endossomos/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HeLa , Humanos , Transporte Proteico , Nexinas de Classificação/genética
13.
J Cell Sci ; 126(Pt 11): 2493-501, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23549789

RESUMO

Many microtubule motors have been shown to couple to endosomal membranes. These motors include dynein in addition to many different kinesin family members. Sorting nexins (SNXs) are central to the organization and function of endosomes. These proteins can actively shape endosomal membranes and couple directly or indirectly to the minus-end microtubule motor dynein. Motor proteins acting on endosomes drive their motility, dictate their morphology and affect cargo segregation. We have used well-characterized members of the SNX family to elucidate motor coupling using high-resolution light microscopy coupled with depletion of specific microtubule motors. Endosomal domains labelled with SNX1, SNX4 and SNX8 couple to discrete combinations of dynein and kinesin motors. These specific combinations govern the structure and motility of each SNX-coated membrane in addition to the segregation of distinct functional endosomal subdomains. Taken together, our data show that these key features of endosome dynamics are governed by the same set of opposing microtubule motors. Thus, microtubule motors help to define the mosaic layout of endosomes that underpins cargo sorting.


Assuntos
Dineínas/metabolismo , Endossomos/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Nexinas de Classificação/metabolismo , Transporte Biológico Ativo/fisiologia , Linhagem Celular , Dineínas/genética , Endossomos/genética , Humanos , Membranas Intracelulares , Cinesinas/genética , Microtúbulos/genética , Nexinas de Classificação/genética
14.
Nat Cell Biol ; 15(5): 461-71, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23563491

RESUMO

The PDZ-domain-containing sorting nexin 27 (SNX27) promotes recycling of internalized transmembrane proteins from endosomes to the plasma membrane by linking PDZ-dependent cargo recognition to retromer-mediated transport. Here, we employed quantitative proteomics of the SNX27 interactome and quantification of the surface proteome of SNX27- and retromer-suppressed cells to dissect the assembly of the SNX27 complex and provide an unbiased global view of SNX27-mediated sorting. Over 100 cell surface proteins, many of which interact with SNX27, including the glucose transporter GLUT1, the Menkes disease copper transporter ATP7A, various zinc and amino acid transporters, and numerous signalling receptors, require SNX27-retromer to prevent lysosomal degradation and maintain surface levels. Furthermore, we establish that direct interaction of the SNX27 PDZ domain with the retromer subunit VPS26 is necessary and sufficient to prevent lysosomal entry of SNX27 cargo. Our data identify the SNX27-retromer as a major endosomal recycling hub required to maintain cellular nutrient homeostasis.


Assuntos
Glucose/metabolismo , Proteômica/métodos , Nexinas de Classificação/análise , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Western Blotting , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Biologia Computacional/métodos , ATPases Transportadoras de Cobre , Meios de Cultura/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Células HeLa , Humanos , Transporte de Íons , Marcação por Isótopo/métodos , Lisossomos/metabolismo , Complexos Multiproteicos/metabolismo , Domínios PDZ , Dobramento de Proteína , Mapeamento de Interação de Proteínas , Transporte Proteico , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Nexinas de Classificação/genética , Nexinas de Classificação/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
15.
J Cell Biol ; 197(2): 219-30, 2012 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-22492727

RESUMO

The FERM-like domain-containing sorting nexins of the SNX17/SNX27/SNX31 family have been proposed to mediate retrieval of transmembrane proteins from the lysosomal pathway. In this paper, we describe a stable isotope labeling with amino acids in culture-based quantitative proteomic approach that allows an unbiased, global identification of transmembrane cargoes that are rescued from lysosomal degradation by SNX17. This screen revealed that several integrins required SNX17 for their stability, as depletion of SNX17 led to a loss of ß1 and ß5 integrins and associated a subunits from HeLa cells as a result of increased lysosomal degradation. SNX17 bound to the membrane distal NPXY motif in ß integrin cytoplasmic tails, thereby preventing lysosomal degradation of ß integrins and their associated a subunits. Furthermore, SNX17-dependent retrieval of integrins did not depend on the retromer complex. Consistent with an effect on integrin recycling, depletion of SNX17 also caused alterations in cell migration. Our data provide mechanistic insight into the retrieval of internalized integrins from the lysosomal degradation pathway, a prerequisite for subsequent recycling of these matrix receptors.


Assuntos
Lisossomos/metabolismo , Transporte Proteico , Nexinas de Classificação/metabolismo , Transporte Biológico Ativo , Linhagem Celular Tumoral , Movimento Celular/genética , Células HEK293 , Células HeLa , Humanos , Cadeias beta de Integrinas/metabolismo , Integrina beta1/metabolismo , Transporte Proteico/genética , Proteômica , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
16.
PLoS One ; 7(2): e32386, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384237

RESUMO

The inositol (1,4,5) trisphosphate 3-kinases comprise a family of enzymes (A, B, and C) that phosphorylate the calcium mobilising molecule inositol (1,4,5) trisphosphate (IP(3)) to generate inositol (1,3,4,5) tetrakisphosphate. This molecule can function as a second messenger, but its roles are not completely understood. The A isoform of inositol (1,4,5) trisphosphate 3-kinase localises to filamentous actin within dendritic spines in the hippocampus and is implicated in the regulation of spine morphology and long term potentiation, however the mechanisms through which it signals in neuronal cells are not completely understood. We have used NGF driven neurite outgrowth from PC12 cells as a platform to examine the impact of signaling via inositol (1,4,5) trisphosphate 3-kinase activity in a neuronal cell. We have found that the catalytic activity of the enzyme opposes neurite outgrowth, whilst pharmacological inhibition of inositol (1,4,5) trisphosphate 3-kinase leads to a significant increase in neurite outgrowth, and we show that the reduction in neurite outgrowth in response to inositol (1,4,5) trisphosphate 3-kinase activity correlates with reduced ERK activity as determined by western blotting using phosphorylation-specific antibodies. Our findings suggest a novel neuronal signaling pathway linking metabolism of IP(3) to signaling via ERK.


Assuntos
Fator de Crescimento Neural/metabolismo , Neuritos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Actinas/metabolismo , Animais , Catálise , Diferenciação Celular , Separação Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/metabolismo , Modelos Biológicos , Neurônios/metabolismo , Células PC12 , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ratos , Transdução de Sinais
17.
Dev Cell ; 17(1): 110-22, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19619496

RESUMO

Early endosome-to-trans-Golgi network (TGN) transport is organized by the retromer complex. Consisting of cargo-selective and membrane-bound subcomplexes, retromer coordinates sorting with membrane deformation and carrier formation. Here, we describe four mammalian retromers whose membrane-bound subcomplexes contain specific combinations of the sorting nexins (SNX), SNX1, SNX2, SNX5, and SNX6. We establish that retromer requires a dynamic spatial organization of the endosomal network, which is regulated through association of SNX5/SNX6 with the p150(glued) component of dynactin, an activator of the minus-end directed microtubule motor dynein; an association further defined through genetic studies in C. elegans. Finally, we also establish that the spatial organization of the retromer pathway is mediated through the association of SNX1 with the proposed TGN-localized tether Rab6-interacting protein-1. These interactions describe fundamental steps in retromer-mediated transport and establish that the spatial organization of the retromer network is a critical element required for efficient retromer-mediated sorting.


Assuntos
Proteínas de Transporte/metabolismo , Dineínas/metabolismo , Endossomos/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Rede trans-Golgi/metabolismo , Animais , Transporte Biológico/fisiologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Transporte/classificação , Proteínas de Transporte/genética , Linhagem Celular , Complexo Dinactina , Dineínas/genética , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Complexos Multiproteicos/metabolismo , Filogenia , Isoformas de Proteínas/genética , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Nexinas de Classificação , Técnicas do Sistema de Duplo-Híbrido , Proteínas de Transporte Vesicular/classificação , Proteínas de Transporte Vesicular/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
18.
Nat Cell Biol ; 9(12): 1370-80, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17994011

RESUMO

SNX-BAR proteins are a sub-family of sorting nexins implicated in endosomal sorting. Here, we establish that through its phox homology (PX) and Bin-Amphiphysin-Rvs (BAR) domains, sorting nexin-4 (SNX4) is associated with tubular and vesicular elements of a compartment that overlaps with peripheral early endosomes and the juxtanuclear endocytic recycling compartment (ERC). Suppression of SNX4 perturbs transport between these compartments and causes lysosomal degradation of the transferrin receptor (TfnR). Through an interaction with KIBRA, a protein previously shown to bind dynein light chain 1, we establish that SNX4 associates with the minus end-directed microtubule motor dynein. Although suppression of KIBRA and dynein perturbs early endosome-to-ERC transport, TfnR sorting is maintained. We propose that by driving membrane tubulation, SNX4 coordinates iterative, geometric-based sorting of the TfnR with the long-range transport of carriers from early endosomes to the ERC. Finally, these data suggest that by associating with molecular motors, SNX-BAR proteins may coordinate sorting with carrier transport between donor and recipient membranes.


Assuntos
Dineínas/fisiologia , Endocitose , Receptores da Transferrina/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Compartimento Celular , Membrana Celular/metabolismo , Endossomos/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Microtúbulos/metabolismo , Fosfoproteínas , Ligação Proteica , Transporte Proteico , Proteínas/metabolismo , Nexinas de Classificação
19.
Proc Natl Acad Sci U S A ; 104(35): 13978-83, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17709751

RESUMO

The contribution of the B isoform of inositol 1,4,5-trisphosphate [Ins(1,4,5)P(3)] 3-kinase (or Itpkb) and inositol 1,3,4,5-tetrakisphosphate [Ins(1,3,4,5)P(4)], its reaction product, to B cell function and development remains unknown. Here, we show that mice deficient in Itpkb have defects in B cell survival leading to specific and intrinsic developmental alterations in the B cell lineage and antigen unresponsiveness in vivo. The decreased B cell survival is associated with a decreased phosphorylation of Erk1/2 and increased Bim gene expression. B cell survival, development, and antigen responsiveness are normalized in parallel to reduced expression of Bim in Itpkb(-/-) Bim(+/-) mice. Analysis of the signaling pathway downstream of Itpkb revealed that Ins(1,3,4,5)P(4) regulates subcellular distribution of Rasa3, a Ras GTPase-activating protein acting as an Ins(1,3,4,5)P(4) receptor. Together, our results indicate that Itpkb and Ins(1,3,4,5)P(4) mediate a survival signal in B cells via a Rasa3-Erk signaling pathway controlling proapoptotic Bim gene expression.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Linfócitos B/citologia , Linfócitos B/fisiologia , Fosfatos de Inositol/farmacologia , Proteínas de Membrana/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Proto-Oncogênicas/genética , Animais , Apoptose , Proteínas Reguladoras de Apoptose/deficiência , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/fisiologia , Linfócitos B/imunologia , Proteína 11 Semelhante a Bcl-2 , Medula Óssea/imunologia , Sobrevivência Celular , Regulação da Expressão Gênica , Imunoglobulina D/análise , Imunoglobulina M/análise , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Proteínas Proto-Oncogênicas/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/imunologia
20.
Proc Natl Acad Sci U S A ; 104(30): 12353-8, 2007 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-17640920

RESUMO

Ras has achieved notoriety as an oncogene aberrantly activated in multiple human tumors. Approximately 30% of all human tumors express an oncogenic form of this GTPase that is locked in an active conformation as a result of being insensitive to Ras GTPase-activating proteins (GAPs), proteins that normally regulate the inactivation of Ras by enhancing its intrinsic GTPase activity. Besides oncogenic mutations in Ras, signaling by wild-type Ras is also frequently deregulated in tumors through aberrant coupling to activated cell surface receptors. This indicates that alternative mechanisms of aberrant wild-type Ras activation may be involved in tumorigenesis. Here, we describe another mechanism through which aberrant Ras activation is achieved in human cancers. We have established that Ras GTPase-activating-like protein (RASAL), a Ca(2+)-regulated Ras GAP that decodes the frequency of Ca(2+) oscillations, is silenced through CpG methylation in multiple tumors. With the finding that ectopic expression of catalytically active RASAL leads to growth inhibition of these tumor cells by Ras inactivation, we have provided evidence that epigenetically silencing of this Ras GAP represents a mechanism of aberrant Ras activation in certain cancers. Our demonstration that RASAL constitutes a tumor suppressor gene has therefore further emphasized the importance of Ca(2+) in the regulation of Ras signaling and has established that deregulation of this pathway is an important step in Ras-mediated tumorigenesis.


Assuntos
Cálcio/metabolismo , Inativação Gênica , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas ras/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos Par 12/genética , Ilhas de CpG , Regulação para Baixo , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Metilação , Neoplasias/patologia , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA