Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 166(1): 193-208, 2016 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-27293189

RESUMEN

γ-Secretases are a family of intramembrane-cleaving proteases involved in various signaling pathways and diseases, including Alzheimer's disease (AD). Cells co-express differing γ-secretase complexes, including two homologous presenilins (PSENs). We examined the significance of this heterogeneity and identified a unique motif in PSEN2 that directs this γ-secretase to late endosomes/lysosomes via a phosphorylation-dependent interaction with the AP-1 adaptor complex. Accordingly, PSEN2 selectively cleaves late endosomal/lysosomal localized substrates and generates the prominent pool of intracellular Aß that contains longer Aß; familial AD (FAD)-associated mutations in PSEN2 increased the levels of longer Aß further. Moreover, a subset of FAD mutants in PSEN1, normally more broadly distributed in the cell, phenocopies PSEN2 and shifts its localization to late endosomes/lysosomes. Thus, localization of γ-secretases determines substrate specificity, while FAD-causing mutations strongly enhance accumulation of aggregation-prone Aß42 in intracellular acidic compartments. The findings reveal potentially important roles for specific intracellular, localized reactions contributing to AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/análisis , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Presenilina-2/análisis , Complejo 1 de Proteína Adaptadora/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secuencias de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular Tumoral , Endosomas/química , Humanos , Lisosomas/química , Ratones , Presenilina-1/análisis , Presenilina-1/química , Presenilina-1/genética , Presenilina-1/metabolismo , Presenilina-2/química , Presenilina-2/genética , Presenilina-2/metabolismo , Ratas , Especificidad por Sustrato
2.
PLoS Genet ; 14(4): e1007363, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29698489

RESUMEN

The hereditary spastic paraplegias (HSP) are a clinically and genetically heterogeneous group of disorders characterized by progressive lower limb spasticity. Mutations in subunits of the heterotetrameric (ε-ß4-µ4-σ4) adaptor protein 4 (AP-4) complex cause an autosomal recessive form of complicated HSP referred to as "AP-4 deficiency syndrome". In addition to lower limb spasticity, this syndrome features intellectual disability, microcephaly, seizures, thin corpus callosum and upper limb spasticity. The pathogenetic mechanism, however, remains poorly understood. Here we report the characterization of a knockout (KO) mouse for the AP4E1 gene encoding the ε subunit of AP-4. We find that AP-4 ε KO mice exhibit a range of neurological phenotypes, including hindlimb clasping, decreased motor coordination and weak grip strength. In addition, AP-4 ε KO mice display a thin corpus callosum and axonal swellings in various areas of the brain and spinal cord. Immunohistochemical analyses show that the transmembrane autophagy-related protein 9A (ATG9A) is more concentrated in the trans-Golgi network (TGN) and depleted from the peripheral cytoplasm both in skin fibroblasts from patients with mutations in the µ4 subunit of AP-4 and in various neuronal types in AP-4 ε KO mice. ATG9A mislocalization is associated with increased tendency to accumulate mutant huntingtin (HTT) aggregates in the axons of AP-4 ε KO neurons. These findings indicate that the AP-4 ε KO mouse is a suitable animal model for AP-4 deficiency syndrome, and that defective mobilization of ATG9A from the TGN and impaired autophagic degradation of protein aggregates might contribute to neuroaxonal dystrophy in this disorder.


Asunto(s)
Complejo 4 de Proteína Adaptadora/deficiencia , Complejo 4 de Proteína Adaptadora/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas de la Membrana/metabolismo , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Complejo 4 de Proteína Adaptadora/química , Subunidades del Complejo de Proteínas Adaptadoras/química , Subunidades del Complejo de Proteínas Adaptadoras/deficiencia , Subunidades del Complejo de Proteínas Adaptadoras/genética , Animales , Axones/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neuronas/metabolismo , Agregado de Proteínas/genética , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Receptores de Glutamato/metabolismo , Paraplejía Espástica Hereditaria/patología , Médula Espinal/metabolismo , Médula Espinal/patología , Red trans-Golgi/metabolismo
3.
Biochem Soc Trans ; 48(5): 1877-1888, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33084855

RESUMEN

Heterotetrameric adaptor protein (AP) complexes play key roles in protein sorting and transport vesicle formation in the endomembrane system of eukaryotic cells. One of these complexes, AP-4, was identified over 20 years ago but, up until recently, its function remained unclear. AP-4 associates with the trans-Golgi network (TGN) through interaction with small GTPases of the ARF family and recognizes transmembrane proteins (i.e. cargos) having specific sorting signals in their cytosolic domains. Recent studies identified accessory proteins (tepsin, RUSC2 and the FHF complex) that co-operate with AP-4, and cargos (amyloid precursor protein, ATG9A and SERINC3/5) that are exported from the TGN in an AP-4-dependent manner. Defective export of ATG9A from the TGN in AP-4-deficient cells was shown to reduce ATG9A delivery to pre-autophagosomal structures, impairing autophagosome formation and/or maturation. In addition, mutations in AP-4-subunit genes were found to cause neurological dysfunction in mice and a form of complicated hereditary spastic paraplegia referred to as 'AP-4-deficiency syndrome' in humans. These findings demonstrated that mammalian AP-4 is required for the development and function of the central nervous system, possibly through its role in the sorting of ATG9A for the maintenance of autophagic homeostasis. In this article, we review the properties and functions of AP-4, and discuss how they might explain the clinical features of AP-4 deficiency.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Mutación , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo , Red trans-Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Autofagosomas/metabolismo , Autofagia , Sitios de Unión , Caenorhabditis elegans , Cryptococcus neoformans , Drosophila melanogaster , Hongos , Humanos , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Vesículas Transportadoras/metabolismo , Tirosina/química , Proteínas de Transporte Vesicular/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(50): E10697-E10706, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180427

RESUMEN

AP-4 is a member of the heterotetrameric adaptor protein (AP) complex family involved in protein sorting in the endomembrane system of eukaryotic cells. Interest in AP-4 has recently risen with the discovery that mutations in any of its four subunits cause a form of hereditary spastic paraplegia (HSP) with intellectual disability. The critical sorting events mediated by AP-4 and the pathogenesis of AP-4 deficiency, however, remain poorly understood. Here we report the identification of ATG9A, the only multispanning membrane component of the core autophagy machinery, as a specific AP-4 cargo. AP-4 promotes signal-mediated export of ATG9A from the trans-Golgi network to the peripheral cytoplasm, contributing to lipidation of the autophagy protein LC3B and maturation of preautophagosomal structures. These findings implicate AP-4 as a regulator of autophagy and altered autophagy as a possible defect in AP-4-deficient HSP.


Asunto(s)
Complejo 4 de Proteína Adaptadora/metabolismo , Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Complejo 4 de Proteína Adaptadora/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencias de Aminoácidos , Animales , Autofagia , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(36): E5318-27, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27559088

RESUMEN

An open question in cell biology is how the general intracellular transport machinery is adapted to perform specialized functions in polarized cells such as neurons. Here we illustrate this adaptation by elucidating a role for the ubiquitous small GTPase Ras-related protein in brain 5 (Rab5) in neuronal polarity. We show that inactivation or depletion of Rab5 in rat hippocampal neurons abrogates the somatodendritic polarity of the transferrin receptor and several glutamate receptor types, resulting in their appearance in the axon. This loss of polarity is not caused primarily by increased transport from the soma to the axon but rather by decreased retrieval from the axon to the soma. Retrieval is also dependent on the Rab5 effector Fused Toes (FTS)-Hook-FTS and Hook-interacting protein (FHIP) (FHF) complex, which interacts with the minus-end-directed microtubule motor dynein and its activator dynactin to drive a population of axonal retrograde carriers containing somatodendritic proteins toward the soma. These findings emphasize the importance of both biosynthetic sorting and axonal retrieval for the polarized distribution of somatodendritic receptors at steady state.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Axones/metabolismo , Neuronas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Axones/patología , Polaridad Celular/genética , Complejo Dinactina/genética , Complejo Dinactina/metabolismo , Dineínas/química , Dineínas/metabolismo , Endosomas/genética , Endosomas/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Neuronas/fisiología , Transporte de Proteínas , Ratas
6.
Am J Hum Genet ; 97(5): 715-25, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26544806

RESUMEN

Stuttering is a common, highly heritable neurodevelopmental disorder characterized by deficits in the volitional control of speech. Whole-exome sequencing identified two heterozygous AP4E1 coding variants, c.1549G>A (p.Val517Ile) and c.2401G>A (p.Glu801Lys), that co-segregate with persistent developmental stuttering in a large Cameroonian family, and we observed the same two variants in unrelated Cameroonians with persistent stuttering. We found 23 other rare variants, including predicted loss-of-function variants, in AP4E1 in unrelated stuttering individuals in Cameroon, Pakistan, and North America. The rate of rare variants in AP4E1 was significantly higher in unrelated Pakistani and Cameroonian stuttering individuals than in population-matched control individuals, and coding variants in this gene are exceptionally rare in the general sub-Saharan West African, South Asian, and North American populations. Clinical examination of the Cameroonian family members failed to identify any symptoms previously reported in rare individuals carrying homozygous loss-of-function mutations in this gene. AP4E1 encodes the ε subunit of the heterotetrameric (ε-ß4-µ4-σ4) AP-4 complex, involved in protein sorting at the trans-Golgi network. We found that the µ4 subunit of AP-4 interacts with NAGPA, an enzyme involved in the synthesis of the mannose 6-phosphate signal that targets acid hydrolases to the lysosome and the product of a gene previously associated with stuttering. These findings implicate deficits in intracellular trafficking in persistent stuttering.


Asunto(s)
Complejo 4 de Proteína Adaptadora/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Hidrolasas Diéster Fosfóricas/genética , Transporte de Proteínas/genética , Tartamudeo/genética , Tartamudeo/patología , Pueblo Asiatico , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Sitios Genéticos , Heterocigoto , Humanos , Masculino , Linaje , Pronóstico , Red trans-Golgi
7.
J Biol Chem ; 290(52): 30736-49, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26542808

RESUMEN

The heterotetrameric (ϵ-ß4-µ4-σ4) complex adaptor protein 4 (AP-4) is a component of a non-clathrin coat involved in protein sorting at the trans-Golgi network (TGN). Considerable interest in this complex has arisen from the recent discovery that mutations in each of its four subunits are the cause of a congenital intellectual disability and movement disorder in humans. Despite its physiological importance, the structure and function of this coat remain poorly understood. To investigate the assembly of the AP-4 coat, we dissected the determinants of interaction of AP-4 with its only known accessory protein, the ENTH/VHS-domain-containing protein tepsin. Using a variety of protein interaction assays, we found that tepsin comprises two phylogenetically conserved peptide motifs, [GS]LFXG[ML]X[LV] and S[AV]F[SA]FLN, within its C-terminal unstructured region, which interact with the C-terminal ear (or appendage) domains of the ß4 and ϵ subunits of AP-4, respectively. Structure-based mutational analyses mapped the binding site for the [GS]LFXG[ML]X[LV] motif to a conserved, hydrophobic surface on the ß4-ear platform fold. Both peptide-ear interactions are required for efficient association of tepsin with AP-4, and for recruitment of tepsin to the TGN. The bivalency of the interactions increases the avidity of tepsin for AP-4 and may enable cross-linking of multiple AP-4 heterotetramers, thus contributing to the assembly of the AP-4 coat. In addition to revealing critical aspects of this coat, our findings extend the paradigm of peptide-ear interactions, previously established for clathrin-AP-1/AP-2 coats, to a non-clathrin coat.


Asunto(s)
Complejo 4 de Proteína Adaptadora/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Complejo 4 de Proteína Adaptadora/química , Complejo 4 de Proteína Adaptadora/genética , Proteínas Adaptadoras del Transporte Vesicular/química , Proteínas Adaptadoras del Transporte Vesicular/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Dimerización , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Red trans-Golgi/metabolismo
8.
PLoS Pathog ; 10(5): e1004107, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24831812

RESUMEN

Newly synthesized envelope glycoproteins of neuroinvasive viruses can be sorted in a polarized manner to the somatodendritic and/or axonal domains of neurons. Although critical for transneuronal spread of viruses, the molecular determinants and interregulation of this process are largely unknown. We studied the polarized sorting of the attachment (NiV-G) and fusion (NiV-F) glycoproteins of Nipah virus (NiV), a paramyxovirus that causes fatal human encephalitis, in rat hippocampal neurons. When expressed individually, NiV-G exhibited a non-polarized distribution, whereas NiV-F was specifically sorted to the somatodendritic domain. Polarized sorting of NiV-F was dependent on interaction of tyrosine-based signals in its cytosolic tail with the clathrin adaptor complex AP-1. Co-expression of NiV-G with NiV-F abolished somatodendritic sorting of NiV-F due to incorporation of NiV-G•NiV-F complexes into axonal transport carriers. We propose that faster biosynthetic transport of unassembled NiV-F allows for its proteolytic activation in the somatodendritic domain prior to association with NiV-G and axonal delivery of NiV-G•NiV-F complexes. Our study reveals how interactions of viral glycoproteins with the host's transport machinery and between themselves regulate their polarized sorting in neurons.


Asunto(s)
Polaridad Celular/fisiología , Neuronas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Embrión de Mamíferos , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Neuronas/fisiología , Virus Nipah , Señales de Clasificación de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Proteínas del Envoltorio Viral/química , Ensamble de Virus
9.
Proc Natl Acad Sci U S A ; 109(10): 3820-5, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22343291

RESUMEN

The coxsackie and adenovirus receptor (CAR) plays key roles in epithelial barrier function at the tight junction, a localization guided in part by a tyrosine-based basolateral sorting signal, (318)YNQV(321). Sorting motifs of this type are known to route surface receptors into clathrin-mediated endocytosis through interaction with the medium subunit (µ2) of the clathrin adaptor AP-2, but how they guide new and recycling membrane proteins basolaterally is unknown. Here, we show that YNQV functions as a canonical YxxΦ motif, with both Y318 and V321 required for the correct basolateral localization and biosynthetic sorting of CAR, and for interaction with a highly conserved pocket in the medium subunits (µ1A and µ1B) of the clathrin adaptors AP-1A and AP-1B. Knock-down experiments demonstrate that AP-1A plays a role in the biosynthetic sorting of CAR, complementary to the role of AP-1B in basolateral recycling of this receptor. Our study illustrates how two clathrin adaptors direct basolateral trafficking of a plasma membrane protein through interaction with a canonical YxxΦ motif.


Asunto(s)
Complejo 1 de Proteína Adaptadora/química , Receptores Virales/química , Complejo 2 de Proteína Adaptadora/química , Secuencias de Aminoácidos , Animales , Línea Celular , Membrana Celular/metabolismo , Clatrina/química , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Perros , Endocitosis , Endosomas/metabolismo , Células Epiteliales/citología , Exocitosis , Peces , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mutación , Conformación Proteica , Transporte de Proteínas , Ranidae
10.
J Clin Invest ; 133(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36951961

RESUMEN

Spastic paraplegia 50 (SPG50) is an ultrarare childhood-onset neurological disorder caused by biallelic loss-of-function variants in the AP4M1 gene. SPG50 is characterized by progressive spastic paraplegia, global developmental delay, and subsequent intellectual disability, secondary microcephaly, and epilepsy. We preformed preclinical studies evaluating an adeno-associated virus (AAV)/AP4M1 gene therapy for SPG50 and describe in vitro studies that demonstrate transduction of patient-derived fibroblasts with AAV2/AP4M1, resulting in phenotypic rescue. To evaluate efficacy in vivo, Ap4m1-KO mice were intrathecally (i.t.) injected with 5 × 1011, 2.5 × 1011, or 1.25 × 1011 vector genome (vg) doses of AAV9/AP4M1 at P7-P10 or P90. Age- and dose-dependent effects were observed, with early intervention and higher doses achieving the best therapeutic benefits. In parallel, three toxicology studies in WT mice, rats, and nonhuman primates (NHPs) demonstrated that AAV9/AP4M1 had an acceptable safety profile up to a target human dose of 1 × 1015 vg. Of note, similar degrees of minimal-to-mild dorsal root ganglia (DRG) toxicity were observed in both rats and NHPs, supporting the use of rats to monitor DRG toxicity in future i.t. AAV studies. These preclinical results identify an acceptably safe and efficacious dose of i.t.-administered AAV9/AP4M1, supporting an investigational gene transfer clinical trial to treat SPG50.


Asunto(s)
Paraplejía Espástica Hereditaria , Humanos , Ratas , Ratones , Animales , Niño , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/terapia , Terapia Genética , Dependovirus/genética , Vectores Genéticos , Paraplejía/genética , Paraplejía/terapia
11.
J Biol Chem ; 286(3): 2022-30, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21097499

RESUMEN

The clathrin-associated, heterotetrameric adaptor protein (AP) complexes, AP-1, AP-2, and AP-3, recognize signals in the cytosolic domains of transmembrane proteins, leading to their sorting to endosomes, lysosomes, lysosome-related organelles, and/or the basolateral membrane of polarized epithelial cells. One type of signal, referred to as "dileucine-based," fits the consensus motif (D/E)XXXL(L/I). Previous biochemical analyses showed that (D/E)XXXL(L/I) signals bind to a combination of two subunits of each AP complex, namely the AP-1 γ-σ1, AP-2 α-σ2, and AP-3 δ-σ3 hemicomplexes, and structural studies revealed that an imperfect variant of this motif lacking the (D/E) residue binds to a site straddling the interface of α and σ2. Herein, we report mutational and binding analyses showing that canonical (D/E)XXXL(L/I) signals bind to this same site on AP-2, and to similar sites on AP-1 and AP-3. The strength and amino acid requirements of different interactions depend on the specific signals and AP complexes involved. We also demonstrate the occurrence of diverse AP-1 heterotetramers by combinatorial assembly of various γ and σ1 subunit isoforms encoded by different genes. These AP-1 variants bind (D/E)XXXL(L/I) signals with marked preferences for certain sequences, implying that they are not functionally equivalent. Our results thus demonstrate that different AP complexes share a conserved binding site for (D/E)XXXL(L/I) signals. However, the characteristics of the binding site on each complex vary, providing for the specific recognition of a diverse repertoire of (D/E)XXXL(L/I) signals.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Polaridad Celular/fisiología , Células Epiteliales/metabolismo , Complejos Multiproteicos/metabolismo , Señales de Clasificación de Proteína/fisiología , Proteínas Adaptadoras del Transporte Vesicular/genética , Secuencias de Aminoácidos , Sitios de Unión , Línea Celular , Células Epiteliales/citología , Humanos , Complejos Multiproteicos/genética , Mutación
12.
EMBO J ; 27(19): 2484-94, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18772883

RESUMEN

Rab GTPases and ubiquitination are critical regulators of transmembrane cargo sorting in endocytic and lysosomal targeting pathways. The endosomal protein Rabex-5 intersects these two layers of regulation by being both a guanine nucleotide exchange factor (GEF) for Rab5 and a substrate for ubiquitin (Ub) binding and conjugation. The ability of trafficking machinery components to bind ubiquitinated proteins is known to have a function in cargo sorting. Here, we demonstrate that Ub binding is essential for the recruitment of Rabex-5 from the cytosol to endosomes, independently of its GEF activity and of Rab5. We also show that monoubiquitinated Rabex-5 is enriched in the cytosol. These observations are consistent with a model whereby a cycle of Ub binding and monoubiquitination regulates the association of Rabex-5 with endosomes.


Asunto(s)
Endosomas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ubiquitina/metabolismo , Animales , Bovinos , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Células HeLa , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ubiquitina/química , Ubiquitina/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
13.
Mol Biol Cell ; 33(12): ar109, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35976721

RESUMEN

Adaptor protein 4 (AP-4) is a heterotetrameric complex composed of ε, ß4, µ4, and σ4 subunits that mediates export of a subset of transmembrane cargos, including autophagy protein 9A (ATG9A), from the trans-Golgi network (TGN). AP-4 has received particular attention in recent years because mutations in any of its subunits cause a complicated form of hereditary spastic paraplegia referred to as "AP-4-deficiency syndrome." The identification of proteins that interact with AP-4 has shed light on the mechanisms of AP-4-dependent cargo sorting and distribution within the cell. However, the mechanisms by which the AP-4 complex itself is assembled have remained unknown. Here, we report that the alpha- and gamma-adaptin-binding protein (AAGAB, also known as p34) binds to and stabilizes the AP-4 ε and σ4 subunits, thus promoting complex assembly. The physiological importance of these interactions is underscored by the observation that AAGAB-knockout cells exhibit reduced levels of AP-4 subunits and accumulation of ATG9A at the TGN like those in cells with mutations in AP-4-subunit genes. These findings demonstrate that AP-4 assembly is not spontaneous but AAGAB-assisted, further contributing to the understanding of an adaptor protein complex that is critically involved in development of the central nervous system.


Asunto(s)
Subunidades del Complejo de Proteínas Adaptadoras , Proteínas de la Membrana , Complejo 1 de Proteína Adaptadora/metabolismo , Complejo 2 de Proteína Adaptadora/metabolismo , Subunidades del Complejo de Proteínas Adaptadoras/metabolismo , Subunidades gamma de Complejo de Proteína Adaptadora/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas , Red trans-Golgi/metabolismo
14.
Nat Struct Mol Biol ; 13(3): 264-71, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16462746

RESUMEN

Rabex-5 is an exchange factor for Rab5, a master regulator of endosomal trafficking. Rabex-5 binds monoubiquitin, undergoes covalent ubiquitination and contains an intrinsic ubiquitin ligase activity, all of which require an N-terminal A20 zinc finger followed immediately by a helix. The structure of the N-terminal portion of Rabex-5 bound to ubiquitin at 2.5-A resolution shows that Rabex-5-ubiquitin interactions occur at two sites. The first site is a new type of ubiquitin-binding domain, an inverted ubiquitin-interacting motif, which binds with approximately 29-microM affinity to the canonical Ile44 hydrophobic patch on ubiquitin. The second is a diaromatic patch on the A20 zinc finger, which binds with approximately 22-microM affinity to a polar region centered on Asp58 of ubiquitin. The A20 zinc-finger diaromatic patch mediates ubiquitin-ligase activity by directly recruiting a ubiquitin-loaded ubiquitin-conjugating enzyme.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Bovinos , Secuencia Conservada , Cristalografía por Rayos X , Análisis Mutacional de ADN , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Especificidad por Sustrato , Termodinámica , Ubiquitina/química , Ubiquitina-Proteína Ligasas/metabolismo , Dedos de Zinc
15.
Mol Biol Cell ; 32(21): ar25, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34432492

RESUMEN

Autophagy-related protein 9 (ATG9) is a transmembrane protein component of the autophagy machinery that cycles between the trans-Golgi network (TGN) in the perinuclear area and other compartments in the peripheral area of the cell. In mammalian cells, export of the ATG9A isoform from the TGN into ATG9A-containing vesicles is mediated by the adaptor protein 4 (AP-4) complex. However, the mechanisms responsible for the subsequent distribution of these vesicles to the cell periphery are unclear. Herein we show that the AP-4-accessory protein RUSC2 couples ATG9A-containing vesicles to the plus-end-directed microtubule motor kinesin-1 via an interaction between a disordered region of RUSC2 and the kinesin-1 light chain. This interaction is counteracted by the microtubule-associated protein WDR47. These findings uncover a mechanism for the peripheral distribution of ATG9A-containing vesicles involving the function of RUSC2 as a kinesin-1 adaptor and WDR47 as a negative regulator of this function.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Autofagosomas/metabolismo , Autofagia , Proteínas Portadoras/fisiología , Células HEK293 , Células HeLa , Humanos , Cinesinas/metabolismo , Microtúbulos/metabolismo , Transporte de Proteínas/fisiología , Red trans-Golgi/metabolismo
16.
J Virol ; 83(6): 2518-30, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19129443

RESUMEN

A critical function of the human immunodeficiency virus type 1 Nef protein is the downregulation of CD4 from the surfaces of infected cells. Nef is believed to act by linking the cytosolic tail of CD4 to the endocytic machinery, thereby increasing the rate of CD4 internalization. In support of this model, weak binary interactions between CD4, Nef, and the endocytic adaptor complex, AP-2, have been reported. In particular, dileucine and diacidic motifs in the C-terminal flexible loop of Nef have been shown to mediate binding to a combination of the alpha and sigma2 subunits of AP-2. Here, we report the identification of a potential binding site for the Nef diacidic motif on alpha-adaptin. This site comprises two basic residues, lysine-297 and arginine-340, on the alpha-adaptin trunk domain. The mutation of these residues specifically inhibits the ability of Nef to bind AP-2 and downregulate CD4. We also present evidence that the diacidic motif on Nef and the basic patch on alpha-adaptin are both required for the cooperative assembly of a CD4-Nef-AP-2 complex. This cooperativity explains how Nef is able to efficiently downregulate CD4 despite weak binary interactions between components of the tripartite complex.


Asunto(s)
Complejo 2 de Proteína Adaptadora/metabolismo , Subunidades alfa de Complejo de Proteína Adaptadora/metabolismo , Antígenos CD4/metabolismo , VIH-1/fisiología , Dominios y Motivos de Interacción de Proteínas , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Subunidades alfa de Complejo de Proteína Adaptadora/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Células HeLa , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación Missense , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Alineación de Secuencia
17.
J Virol ; 83(13): 6578-90, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19403684

RESUMEN

The Nef protein of human immunodeficiency virus type 1 downregulates the CD4 coreceptor from the surface of host cells by accelerating the rate of CD4 endocytosis through a clathrin/AP-2 pathway. Herein, we report that Nef has the additional function of targeting CD4 to the multivesicular body (MVB) pathway for eventual delivery to lysosomes. This targeting involves the endosomal sorting complex required for transport (ESCRT) machinery. Perturbation of this machinery does not prevent removal of CD4 from the cell surface but precludes its lysosomal degradation, indicating that accelerated endocytosis and targeting to the MVB pathway are separate functions of Nef. We also show that both CD4 and Nef are ubiquitinated on lysine residues, but this modification is dispensable for Nef-induced targeting of CD4 to the MVB pathway.


Asunto(s)
Antígenos CD4/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Regulación hacia Abajo , VIH-1/metabolismo , Células HeLa , Humanos , Interferencia de ARN , Ubiquitinación
18.
Mol Biol Cell ; 18(9): 3486-501, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17596511

RESUMEN

The sorting of acid hydrolase precursors at the trans-Golgi network (TGN) is mediated by binding to mannose 6-phosphate receptors (MPRs) and subsequent capture of the hydrolase-MPR complexes into clathrin-coated vesicles or transport carriers (TCs) destined for delivery to endosomes. This capture depends on the function of three monomeric clathrin adaptors named GGAs. The GGAs comprise a C-terminal "ear" domain that binds a specific set of accessory proteins. Herein we show that one of these accessory proteins, p56, colocalizes and physically interacts with the three GGAs at the TGN. Moreover, overexpression of the GGAs enhances the association of p56 with the TGN, and RNA interference (RNAi)-mediated depletion of the GGAs decreases the TGN association and total levels of p56. RNAi-mediated depletion of p56 or the GGAs causes various degrees of missorting of the precursor of the acid hydrolase, cathepsin D. In the case of p56 depletion, this missorting correlates with decreased mobility of GGA-containing TCs. Transfection with an RNAi-resistant p56 construct, but not with a p56 construct lacking the GGA-ear-interacting motif, restores the mobility of the TCs. We conclude that p56 tightly cooperates with the GGAs in the sorting of cathepsin D to lysosomes, probably by enabling the movement of GGA-containing TCs.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Clatrina/metabolismo , Lisosomas/enzimología , Vesículas Transportadoras/metabolismo , Red trans-Golgi/metabolismo , Factores de Ribosilacion-ADP/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Animales , Células COS , Catepsina D/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Fibroblastos/citología , Fibroblastos/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Humanos , Unión Proteica , Transporte de Proteínas , Interferencia de ARN , Ratas , Proteínas Recombinantes de Fusión/metabolismo
19.
Mol Biol Cell ; 18(8): 2991-3001, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17538018

RESUMEN

The adaptor protein 1 (AP1) complex is a heterotetramer that participates in cargo sorting into clathrin-coated vesicles at the trans-Golgi network (TGN) and endosomes. The gamma subunit of AP1 possesses a C-terminal "ear" domain that recruits a cohort of accessory proteins through recognition of a shared canonical motif, PsiG[PDE][PsiLM] (where Psi is an aromatic residue). The physiological relevance of these ear-motif interactions, however, remains to be demonstrated. Here we report that the cyclin G-associated kinase (GAK) has two sequences fitting this motif, FGPL and FGEF, which mediate binding to the AP1-gamma-ear domain in vitro. Mutation of both gamma-ear-binding sequences or depletion of AP1-gamma by RNA interference (RNAi) decreases the association of GAK with the TGN in vivo. Depletion of GAK by RNAi impairs the sorting of the acid hydrolase, cathepsin D, to lysosomes. Importantly, expression of RNAi-resistant GAK restores the lysosomal sorting of cathepsin D in cells depleted of endogenous GAK, whereas expression of a similar construct bearing mutations in both gamma-ear-binding sequences fails to correct the sorting defect. Thus, interactions between the PsiG[PDE][PsiLM]-motif sequences in GAK and the AP1-gamma-ear domain are critical for the recruitment of GAK to the TGN and the function of GAK in lysosomal enzyme sorting.


Asunto(s)
Complejo 1 de Proteína Adaptadora/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Supervivencia Celular , Secuencia de Consenso , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Cinética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Estructura Terciaria de Proteína , Transporte de Proteínas , Red trans-Golgi/metabolismo
20.
Mol Biol Cell ; 31(9): 963-979, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32073997

RESUMEN

The heterotetrameric adaptor protein complex 4 (AP-4) is a component of a protein coat associated with the trans-Golgi network (TGN). Mutations in AP-4 subunits cause a complicated form of autosomal-recessive hereditary spastic paraplegia termed AP-4-deficiency syndrome. Recent studies showed that AP-4 mediates export of the transmembrane autophagy protein ATG9A from the TGN to preautophagosomal structures. To identify additional proteins that cooperate with AP-4 in ATG9A trafficking, we performed affinity purification-mass spectrometry followed by validation of the hits by biochemical and functional analyses. This approach resulted in the identification of the fused toes homolog-Hook-FHIP (FHF) complex as a novel AP-4 accessory factor. We found that the AP-4-FHF interaction is mediated by direct binding of the AP-4 µ4 subunit to coiled-coil domains in the Hook1 and Hook2 subunits of FHF. Knockdown of FHF subunits resulted in dispersal of AP-4 and ATG9A from the perinuclear region of the cell, consistent with the previously demonstrated role of the FHF complex in coupling organelles to the microtubule (MT) retrograde motor dynein-dynactin. These findings thus uncover an additional mechanism for the distribution of ATG9A within cells and provide further evidence for a role of protein coats in coupling transport vesicles to MT motors.


Asunto(s)
Complejo 4 de Proteína Adaptadora/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Red trans-Golgi/metabolismo , Línea Celular Tumoral , Dineínas/metabolismo , Células HEK293 , Células HeLa , Humanos , Microtúbulos/metabolismo , Transporte de Proteínas , Paraplejía Espástica Hereditaria/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda