Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 147(5): 1118-31, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22118466

RESUMEN

SNAREs provide a large part of the specificity and energy needed for membrane fusion and, to do so, must be localized to their correct membranes. Here, we show that the R-SNAREs VAMP8, VAMP3, and VAMP2, which cycle between the plasma membrane and endosomes, bind directly to the ubiquitously expressed, PtdIns4,5P(2)-binding, endocytic clathrin adaptor CALM/PICALM. X-ray crystallography shows that the N-terminal halves of their SNARE motifs bind the CALM(ANTH) domain as helices in a manner that mimics SNARE complex formation. Mutation of residues in the CALM:SNARE interface inhibits binding in vitro and prevents R-SNARE endocytosis in vivo. Thus, CALM:R-SNARE interactions ensure that R-SNAREs, required for the fusion of endocytic clathrin-coated vesicles with endosomes and also for subsequent postendosomal trafficking, are sorted into endocytic vesicles. CALM's role in directing the endocytosis of small R-SNAREs may provide insight into the association of CALM/PICALM mutations with growth retardation, cognitive defects, and Alzheimer's disease.


Asunto(s)
Endocitosis , Proteínas de Ensamble de Clatrina Monoméricas/metabolismo , Proteínas SNARE/química , Animales , Membrana Celular/metabolismo , Cristalografía por Rayos X , Células HeLa , Humanos , Ratones , Modelos Moleculares , Proteínas R-SNARE/química , Proteínas R-SNARE/metabolismo , Ratas , Proteínas SNARE/metabolismo , Vesículas Transportadoras/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(2): e2213056120, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36595686

RESUMEN

Despite the essential role of plasma cells in health and disease, the cellular mechanisms controlling their survival and secretory capacity are still poorly understood. Here, we identified the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) Sec22b as a unique and critical regulator of plasma cell maintenance and function. In the absence of Sec22b, plasma cells were hardly detectable and serum antibody titers were dramatically reduced. Accordingly, Sec22b-deficient mice fail to mount a protective immune response. At the mechanistic level, we demonstrated that Sec22b contributes to efficient antibody secretion and is a central regulator of plasma cell maintenance through the regulation of their transcriptional identity and of the morphology of the endoplasmic reticulum and mitochondria. Altogether, our results unveil an essential and nonredundant role for Sec22b as a regulator of plasma cell fitness and of the humoral immune response.


Asunto(s)
Células Plasmáticas , Proteínas SNARE , Ratones , Animales , Células Plasmáticas/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Retículo Endoplásmico/metabolismo , Transporte Biológico
3.
EMBO Rep ; 24(12): e57224, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37818801

RESUMEN

The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Liberación del Virus , Humanos , Antígeno 2 del Estroma de la Médula Ósea/antagonistas & inhibidores , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , COVID-19/virología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/genética
4.
Cell ; 141(2): 222-4, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20403318

RESUMEN

To inhibit protein synthesis and induce cell death, plant ricin toxin and bacterial Shiga toxins enter the cell through the endocytic and retrograde secretory pathways. Stechmann et al. (2010) now identify two small-molecule inhibitors that selectively block endosome-to-Golgi retrieval of ricin and Shiga toxins and protect mice from ricin's deadly effects.

5.
Proc Natl Acad Sci U S A ; 119(18): e2201646119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35507892

RESUMEN

Multiple membrane organelles require cholesterol for proper function within cells. The Niemann-Pick type C (NPC) proteins export cholesterol from endosomes to other membrane compartments, including the endoplasmic reticulum (ER), plasma membrane (PM), trans-Golgi network (TGN), and mitochondria, to meet their cholesterol requirements. Defects in NPC cause malfunctions in multiple membrane organelles and lead to an incurable neurological disorder. Acyl-coenzyme A:cholesterol acyltransferase 1 (ACAT1), a resident enzyme in the ER, converts cholesterol to cholesteryl esters for storage. In mutant NPC cells, cholesterol storage still occurs in an NPC-independent manner. Here we report the interesting finding that in a mutant Npc1 mouse (Npc1nmf), Acat1 gene (Soat1) knockout delayed the onset of weight loss, motor impairment, and Purkinje neuron death. It also improved hepatosplenic pathology and prolonged lifespan by 34%. In mutant NPC1 fibroblasts, ACAT1 blockade (A1B) increased cholesterol content associated with TGN-rich membranes and mitochondria, while decreased cholesterol content associated with late endosomes. A1B also restored proper localization of syntaxin 6 and golgin 97 (key proteins in membrane trafficking at TGN) and improved the levels of cathepsin D (a key protease in lysosome and requires Golgi/endosome transport for maturation) and ABCA1 (a key protein controlling cholesterol release at PM). This work supports the hypothesis that diverting cholesterol from storage can benefit multiple diseases that involve cholesterol deficiencies in cell membranes.


Asunto(s)
Longevidad , Enfermedad de Niemann-Pick Tipo C , Acetil-CoA C-Acetiltransferasa , Enfermedad de Alzheimer , Animales , Colesterol , Ésteres del Colesterol , Modelos Animales de Enfermedad , Endosomas/genética , Ratones , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Esterol O-Aciltransferasa
6.
J Cell Sci ; 134(6)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33589494

RESUMEN

In vertebrate photoreceptors, opsins are highly concentrated in a morphologically distinct ciliary compartment known as the outer segment (OS). Opsin is synthesized in the cell body and transported to the OS at a remarkable rate of 100 to 1000 molecules per second. Opsin transport defects contribute to photoreceptor loss and blindness in human ciliopathies. Previous studies revealed that the rhodopsin C-terminal tail, of 44 amino acids, is sufficient to mediate OS targeting in Xenopus photoreceptors. Here, we show that, although the Xenopus C-terminus retains this function in zebrafish, the homologous zebrafish sequence is not sufficient to target opsin to the OS. This functional difference is largely caused by a change of a single amino acid present in Xenopus but not in other vertebrates examined. Furthermore, we find that sequences in the third intracellular cytoplasmic loop (IC3) and adjacent regions of transmembrane helices 6 and 7 are also necessary for opsin transport in zebrafish. Combined with the cytoplasmic tail, these sequences are sufficient to target opsin to the ciliary compartment.


Asunto(s)
Rodopsina , Pez Cebra , Animales , Humanos , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Transporte de Proteínas , Rodopsina/genética , Rodopsina/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
7.
J Cell Sci ; 134(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34042162

RESUMEN

To provide insights into the kiss-and-run and full fusion events resulting in endocytic delivery to lysosomes, we investigated conditions causing increased tethering and pore formation between late endocytic organelles in HeLa cells. Knockout of the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) VAMP7 and VAMP8 showed, by electron microscopy, the accumulation of tethered lysosome-associated membrane protein (LAMP)-carrier vesicles around multivesicular bodies, as well as the appearance of 'hourglass' profiles of late endocytic organelles attached by filamentous tethers, but did not prevent endocytic delivery to lysosomal hydrolases. Subsequent depletion of the SNARE YKT6 reduced this delivery, consistent with it compensating for the absence of VAMP7 and VAMP8. We also investigated filamentous tethering between multivesicular bodies and enlarged endolysosomes following depletion of charged multi-vesicular body protein 6 (CHMP6), and provide the first evidence that pore formation commences at the edge of tether arrays, with pore expansion required for full membrane fusion.


Asunto(s)
Fusión de Membrana , Proteínas SNARE , Endosomas , Células HeLa , Humanos , Lisosomas , Proteínas R-SNARE/genética , Proteínas SNARE/genética
8.
J Biol Chem ; 297(6): 101393, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34762908

RESUMEN

ER-to-Golgi transport is the first step in the constitutive secretory pathway, which, unlike regulated secretion, is believed to proceed nonstop independent of Ca2+ flux. However, here we demonstrate that penta-EF hand (PEF) proteins ALG-2 and peflin constitute a hetero-bifunctional COPII regulator that responds to Ca2+ signaling by adopting one of several distinct activity states. Functionally, these states can adjust the rate of ER export of COPII-sorted cargos up or down by ∼50%. We found that at steady-state Ca2+, ALG-2/peflin hetero-complexes bind to ER exit sites (ERES) through the ALG-2 subunit to confer a low, buffered secretion rate, while peflin-lacking ALG-2 complexes markedly stimulate secretion. Upon Ca2+ signaling, ALG-2 complexes lacking peflin can either increase or decrease the secretion rate depending on signaling intensity and duration-phenomena that could contribute to cellular growth and intercellular communication following secretory increases or protection from excitotoxicity and infection following decreases. In epithelial normal rat kidney (NRK) cells, the Ca2+-mobilizing agonist ATP causes ALG-2 to depress ER export, while in neuroendocrine PC12 cells, Ca2+ mobilization by ATP results in ALG-2-dependent enhancement of secretion. Furthermore, distinct Ca2+ signaling patterns in NRK cells produce opposing ALG-2-dependent effects on secretion. Mechanistically, ALG-2-dependent depression of secretion involves decreased levels of the COPII outer shell and increased peflin targeting to ERES, while ALG-2-dependent enhancement of secretion involves increased COPII outer shell and decreased peflin at ERES. These data provide insights into how PEF protein dynamics affect secretion of important physiological cargoes such as collagen I and significantly impact ER stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Proteínas de Unión al Calcio/genética , Retículo Endoplásmico/genética , Ratones , Células PC12 , Transporte de Proteínas , Ratas
9.
J Biol Chem ; 294(42): 15418-15434, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31467083

RESUMEN

The primary cilium is a cellular sensor that detects light, chemicals, and movement and is important for morphogen and growth factor signaling. The small GTPase Rab11-Rab8 cascade is required for ciliogenesis. Rab11 traffics the guanine nucleotide exchange factor (GEF) Rabin8 to the centrosome to activate Rab8, needed for ciliary growth. Rabin8 also requires the transport particle protein complex (TRAPPC) proteins for centrosome recruitment during ciliogenesis. Here, using an MS-based approach for identifying Rabin8-interacting proteins, we identified C7orf43 (also known as microtubule-associated protein 11 (MAP11)) as being required for ciliation both in human cells and zebrafish embryos. We find that C7orf43 directly binds to Rabin8 and that C7orf43 knockdown diminishes Rabin8 preciliary centrosome accumulation. Interestingly, we found that C7orf43 co-sediments with TRAPPII complex subunits and directly interacts with TRAPPC proteins. Our findings establish that C7orf43 is a TRAPPII-specific complex component, referred to here as TRAPPC14. Additionally, we show that TRAPPC14 is dispensable for TRAPPII complex integrity but mediates Rabin8 association with the TRAPPII complex. Finally, we demonstrate that TRAPPC14 interacts with the distal appendage proteins Fas-binding factor 1 (FBF1) and centrosomal protein 83 (CEP83), which we show here are required for GFP-Rabin8 centrosomal accumulation, supporting a role for the TRAPPII complex in tethering preciliary vesicles to the mother centriole during ciliogenesis. In summary, our findings have revealed an uncharacterized TRAPPII-specific component, C7orf43/TRAPPC14, that regulates preciliary trafficking of Rabin8 and ciliogenesis and support previous findings that the TRAPPII complex functions as a membrane tether.


Asunto(s)
Centriolos/metabolismo , Cilios/metabolismo , Vesículas Citoplasmáticas/metabolismo , Quinasas del Centro Germinal/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Centriolos/genética , Cilios/genética , Vesículas Citoplasmáticas/genética , Quinasas del Centro Germinal/genética , Humanos , Proteínas Asociadas a Microtúbulos/genética , Morfogénesis , Unión Proteica , Pez Cebra
10.
J Cell Sci ; 131(20)2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30254024

RESUMEN

STX19 is an unusual Qa-SNARE as it lacks a C-terminal transmembrane domain. However, it is efficiently targeted to post-Golgi membranes. Here, we set out to determine the intracellular localisation of endogenous STX19 and elucidate the mechanism by which it is targeted to membranes. We have found that a pool of STX19 is localised to tubular recycling endosomes where it colocalises with MICAL-L1 and Rab8 (which has Rab8a and Rab8b forms). Using a combination of genetic, biochemical and cell-based approaches, we have identified that STX19 is S-acylated at its C-terminus and is a substrate for several Golgi-localised S-acyltransferases, suggesting that STX19 is initially S-acylated at the Golgi before trafficking to the plasma membrane and endosomes. Surprisingly, we have found that S-acylation is a key determinant in targeting STX19 to tubular recycling endosomes, suggesting that S-acylation may play a general role in directing proteins to this compartment. In addition, S-acylation also protects STX19 from proteosomal degradation, indicating that S-acylation regulates the function of STX19 at multiple levels.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Acilación/genética , Transporte de Proteínas/genética , Proteínas Q-SNARE/metabolismo , Humanos
11.
PLoS Genet ; 13(4): e1006698, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28403141

RESUMEN

The cellular machinery required for the fusion of constitutive secretory vesicles with the plasma membrane in metazoans remains poorly defined. To address this problem we have developed a powerful, quantitative assay for measuring secretion and used it in combination with combinatorial gene depletion studies in Drosophila cells. This has allowed us to identify at least three SNARE complexes mediating Golgi to PM transport (STX1, SNAP24/29 and Syb; STX1, SNAP24/29 and YKT6; STX4, SNAP24 and Syb). RNAi mediated depletion of YKT6 and VAMP3 in mammalian cells also blocks constitutive secretion suggesting that YKT6 has an evolutionarily conserved role in this process. The unexpected role of YKT6 in plasma membrane fusion may in part explain why RNAi and gene disruption studies have failed to produce the expected phenotypes in higher eukaryotes.


Asunto(s)
Membrana Celular/genética , Proteínas de Drosophila/genética , Proteínas R-SNARE/genética , Proteínas SNARE/genética , Proteína 3 de Membrana Asociada a Vesículas/genética , Animales , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Heterocigoto , Humanos , Fusión de Membrana/genética , Transporte de Proteínas/genética , Proteínas R-SNARE/metabolismo , Interferencia de ARN , Proteínas SNARE/metabolismo , Toxina Shiga I/genética , Toxina Shiga I/metabolismo , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/genética , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo
12.
FASEB J ; 30(12): 4083-4097, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27601439

RESUMEN

α1-Antitrypsin is a serine protease inhibitor produced in the liver that is responsible for the regulation of pulmonary inflammation. The commonest pathogenic gene mutation yields Z-α1-antitrypsin, which has a propensity to self-associate forming polymers that become trapped in inclusions of endoplasmic reticulum (ER). It is unclear whether these inclusions are connected to the main ER network in Z-α1-antitrypsin-expressing cells. Using live cell imaging, we found that despite inclusions containing an immobile matrix of polymeric α1-antitrypsin, small ER resident proteins can diffuse freely within them. Inclusions have many features to suggest they represent fragmented ER, and some are physically separated from the tubular ER network, yet we observed cargo to be transported between them in a cytosol-dependent fashion that is sensitive to N-ethylmaleimide and dependent on Sar1 and sec22B. We conclude that protein recycling occurs between ER inclusions despite their physical separation.-Dickens, J. A., Ordóñez, A., Chambers, J. E., Beckett, A. J., Patel, V., Malzer, E., Dominicus, C. S., Bradley, J., Peden, A. A., Prior, I. A., Lomas, D. A., Marciniak, S. J. The endoplasmic reticulum remains functionally connected by vesicular transport after its fragmentation in cells expressing Z-α1-antitrypsin.


Asunto(s)
Transporte Biológico/fisiología , Retículo Endoplásmico/metabolismo , Hígado/metabolismo , alfa 1-Antitripsina/metabolismo , Animales , Transporte Biológico/genética , Células CHO , Células Cultivadas , Cricetulus , Mutación/genética , alfa 1-Antitripsina/genética
13.
Cell Microbiol ; 18(1): 80-96, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26248337

RESUMEN

Macrophages are critical effectors of the early innate response to bacteria in tissues. Phagocytosis and killing of bacteria are interrelated functions essential for bacterial clearance but the rate-limiting step when macrophages are challenged with large numbers of the major medical pathogen Staphylococcus aureus is unknown. We show that macrophages have a finite capacity for intracellular killing and fail to match sustained phagocytosis with sustained microbial killing when exposed to large inocula of S. aureus (Newman, SH1000 and USA300 strains). S. aureus ingestion by macrophages is associated with a rapid decline in bacterial viability immediately after phagocytosis. However, not all bacteria are killed in the phagolysosome, and we demonstrate reduced acidification of the phagolysosome, associated with failure of phagolysosomal maturation and reduced activation of cathepsin D. This results in accumulation of viable intracellular bacteria in macrophages. We show macrophages fail to engage apoptosis-associated bacterial killing. Ultittop mately macrophages with viable bacteria undergo cell lysis, and viable bacteria are released and can be internalized by other macrophages. We show that cycles of lysis and reuptake maintain a pool of viable intracellular bacteria over time when killing is overwhelmed and demonstrate intracellular persistence in alveolar macrophages in the lungs in a murine model.


Asunto(s)
Macrófagos/inmunología , Macrófagos/microbiología , Viabilidad Microbiana , Fagocitosis , Neumonía Estafilocócica/patología , Staphylococcus aureus/inmunología , Staphylococcus aureus/fisiología , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Ratones
14.
J Neurosci ; 35(19): 7643-53, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25972187

RESUMEN

Dysbindin is a schizophrenia susceptibility factor and subunit of the biogenesis of lysosome-related organelles complex 1 (BLOC-1) required for lysosome-related organelle biogenesis, and in neurons, synaptic vesicle assembly, neurotransmission, and plasticity. Protein networks, or interactomes, downstream of dysbindin/BLOC-1 remain partially explored despite their potential to illuminate neurodevelopmental disorder mechanisms. Here, we conducted a proteome-wide search for polypeptides whose cellular content is sensitive to dysbindin/BLOC-1 loss of function. We identified components of the vesicle fusion machinery as factors downregulated in dysbindin/BLOC-1 deficiency in neuroectodermal cells and iPSC-derived human neurons, among them the N-ethylmaleimide-sensitive factor (NSF). Human dysbindin/BLOC-1 coprecipitates with NSF and vice versa, and both proteins colocalized in a Drosophila model synapse. To test the hypothesis that NSF and dysbindin/BLOC-1 participate in a pathway-regulating synaptic function, we examined the role for NSF in dysbindin/BLOC-1-dependent synaptic homeostatic plasticity in Drosophila. As previously described, we found that mutations in dysbindin precluded homeostatic synaptic plasticity elicited by acute blockage of postsynaptic receptors. This dysbindin mutant phenotype is fully rescued by presynaptic expression of either dysbindin or Drosophila NSF. However, neither reduction of NSF alone or in combination with dysbindin haploinsufficiency impaired homeostatic synaptic plasticity. Our results demonstrate that dysbindin/BLOC-1 expression defects result in altered cellular content of proteins of the vesicle fusion apparatus and therefore influence synaptic plasticity.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Asociadas a la Distrofina/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas Sensibles a N-Etilmaleimida/metabolismo , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Drosophila , Proteínas de Drosophila/genética , Disbindina , Proteínas Asociadas a la Distrofina/genética , Regulación de la Expresión Génica/genética , Humanos , Melanoma/patología , Proteínas Sensibles a N-Etilmaleimida/genética , Red Nerviosa/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/patología , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Proteínas SNARE/metabolismo , Sinapsis/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
15.
J Cell Sci ; 126(Pt 20): 4647-58, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23902685

RESUMEN

Invadopodia-dependent degradation of the basement membrane plays a major role during metastasis of breast cancer cells. Basement membrane degradation is mediated by targeted secretion of various matrix metalloproteinases (MMPs). Specifically, MMP2 and MMP9 (MMP2/9) possess the ability to hydrolyze components of the basement membrane and regulate various aspects of tumor growth and metastasis. However, the membrane transport machinery that mediates targeting of MMP2/9 to the invadopodia during cancer cell invasion remains to be defined. Because Rab GTPases are key regulators of membrane transport, we screened a human Rab siRNA library and identified Rab40b GTPase as a protein required for secretion of MMP2/9. We also have shown that Rab40b functions during at least two distinct steps of MMP2/9 transport. Here, we demonstrate that Rab40b is required for MMP2/9 sorting into VAMP4-containing secretory vesicles. We also show that Rab40b regulates transport of MMP2/9 secretory vesicles during invadopodia formation and is required for invadopodia-dependent extracellular matrix degradation. Finally, we demonstrate that Rab40b is also required for breast cancer cell invasion in vitro. On the basis of these findings, we propose that Rab40b mediates trafficking of MMP2/9 during invadopodia formation and metastasis of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Seudópodos/metabolismo , Seudópodos/patología , Proteínas de Unión al GTP rab/metabolismo , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Femenino , Humanos , Invasividad Neoplásica , Seudópodos/enzimología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transfección
16.
EMBO J ; 29(2): 304-14, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19942856

RESUMEN

Genetic screens in the yeast Saccharomyces cerevisiae have identified many proteins involved in the secretory pathway, most of which have orthologues in higher eukaryotes. To investigate whether there are additional proteins that are required for secretion in metazoans but are absent from yeast, we used genome-wide RNA interference (RNAi) to look for genes required for secretion of recombinant luciferase from Drosophila S2 cells. This identified two novel components of the secretory pathway that are conserved from humans to plants. Gryzun is distantly related to, but distinct from, the Trs130 subunit of the TRAPP complex but is absent from S. cerevisiae. RNAi of human Gryzun (C4orf41) blocks Golgi exit. Kish is a small membrane protein with a previously uncharacterised orthologue in yeast. The screen also identified Drosophila orthologues of almost 60% of the yeast genes essential for secretion. Given this coverage, the small number of novel components suggests that contrary to previous indications the number of essential core components of the secretory pathway is not much greater in metazoans than in yeasts.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Genes de Insecto , Vías Secretoras , Animales , Línea Celular , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Eucariontes/genética , Eucariontes/metabolismo , Aparato de Golgi/metabolismo , Humanos , Interferencia de ARN , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
17.
J Cell Sci ; 124(Pt 3): 469-82, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21242315

RESUMEN

Autophagy is a lysosome-dependent cellular catabolic mechanism that mediates the turnover of intracellular organelles and long-lived proteins. Reduced autophagic activity has been shown to lead to the accumulation of misfolded proteins in neurons and might be involved in chronic neurodegenerative diseases. Here, we uncover an essential role for the syntaxin-5 SNARE complex in autophagy. Using genetic knockdown, we show that the syntaxin-5 SNARE complex regulates the later stages of autophagy after the initial formation of autophagosomes. This SNARE complex acts on autophagy by regulating ER-to-Golgi transport through the secretory pathway, which is essential for the activity of lysosomal proteases such as cathepsins. Depletion of syntaxin-5 complex components results in the accumulation of autophagosomes as a result of lysosomal dysfunction, leading to decreased degradation of autophagic substrates. Our findings provide a novel link between a fundamental process such as intracellular trafficking and human diseases that might be affected by defective biogenesis and/or homeostasis of the autophagosome-lysosome degradation system.


Asunto(s)
Autofagia/fisiología , Lisosomas/enzimología , Proteínas Qa-SNARE/fisiología , Proteínas Adaptadoras del Transporte Vesicular , Transporte Biológico/fisiología , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas R-SNARE/fisiología , Proteínas de Transporte Vesicular/fisiología
18.
iScience ; 26(7): 107056, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37346049

RESUMEN

The prevalence and strength of serological responses mounted toward SARS-CoV-2 proteins other than nucleocapsid (N) and spike (S), which may be of use as additional serological markers, remains underexplored. Using high-content microscopy to assess antibody responses against full-length StrepTagged SARS-CoV-2 proteins, we found that 85% (166/196) of unvaccinated individuals with RT-PCR confirmed SARS-CoV-2 infections and 74% (31/42) of individuals infected after being vaccinated developed detectable IgG against the structural protein M, which is higher than previous estimates. Compared with N antibodies, M IgG displayed a shallower time-dependent decay and greater specificity. Sensitivity for SARS-CoV-2 seroprevalence was enhanced when N and M IgG detection was combined. These findings indicate that screening for M seroconversion may be a good approach for detecting additional vaccine breakthrough infections and highlight the potential to use HCM as a rapidly deployable method to identify the most immunogenic targets of newly emergent pathogens.

19.
Traffic ; 11(9): 1191-204, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20545907

RESUMEN

The role of SNAREs in mammalian constitutive secretion remains poorly defined. To address this, we have developed a novel flow cytometry-based assay for measuring constitutive secretion and have performed a targeted SNARE and Sec1/Munc18 (SM) protein-specific siRNA screen (38 SNAREs, 4 SNARE-like proteins and 7 SM proteins). We have identified the endoplasmic reticulum (ER)/Golgi SNAREs syntaxin 5, syntaxin 17, syntaxin 18, GS27, SLT1, Sec20, Sec22b, Ykt6 and the SM protein Sly1, along with the post-Golgi SNAREs SNAP-29 and syntaxin 19, as being required for constitutive secretion. Depletion of SNAP-29 or syntaxin 19 causes a decrease in the number of fusion events at the cell surface and in SNAP-29-depleted cells causes an increase in the number of docked vesicles at the plasma membrane as determined by total internal reflection fluorescence (TIRF) microscopy. Analysis of syntaxin 19-interacting partners by mass spectrometry indicates that syntaxin 19 can form SNARE complexes with SNAP-23, SNAP-25, SNAP-29, VAMP3 and VAMP8, supporting its role in Golgi to plasma membrane transport or fusion. Surprisingly, we have failed to detect any requirement for a post-Golgi-specific R-SNARE in this process.


Asunto(s)
Citometría de Flujo/métodos , ARN Interferente Pequeño , Proteínas SNARE/metabolismo , Animales , Humanos , Transporte de Proteínas , Proteínas Qa-SNARE/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas SNARE/genética , Transducción de Señal
20.
Hum Mol Genet ; 19(6): 1009-18, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20028792

RESUMEN

Mutations in the functionally uncharacterized protein SH3TC2 are associated with the severe hereditary peripheral neuropathy, Charcot-Marie-Tooth disease type 4C (CMT4C). Similarly, to other proteins mutated in CMT, a role for SH3TC2 in endocytic membrane traffic has been previously proposed. However, recent descriptions of the intracellular localization of SH3TC2 are conflicting. Furthermore, no clear functional pathogenic mechanisms have so far been proposed to explain why both nonsense and missense mutations in SH3TC2 lead to similar clinical phenotypes. Here, we describe our intracellular localization studies, supported by biochemical and functional data, using wild-type and mutant SH3TC2. We show that wild-type SH3TC2 targets to the intracellular recycling endosome by associating with the small GTPase, Rab11, which is known to regulate the recycling of internalized membrane and receptors back to the plasma membrane. Furthermore, we demonstrate that SH3TC2 interacts preferentially with the GTP-bound form of Rab11, identifying SH3TC2 as a novel Rab11 effector. Of clinical pathological relevance, all SH3TC2 constructs harbouring disease-causing mutations are shown to be unable to associate with Rab11 with consequent loss of recycling endosome localization. Moreover, we show that wild-type SH3TC2, but not mutant SH3TC2, influences transferrin receptor dynamics, consistent with a functional role on the endocytic recycling pathway. Our data therefore implicate mistargeting of SH3TC2 away from the recycling endosome as the fundamental molecular defect that leads to CMT4C.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/metabolismo , Endocitosis , Endosomas/metabolismo , Proteínas/metabolismo , Animales , Western Blotting , Extractos Celulares , Membrana Celular/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Espacio Intracelular/metabolismo , Microscopía Confocal , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas/química , Ratas , Receptores de Transferrina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA