Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-36951961

ABSTRACT

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.


Subject(s)
Spastic Paraplegia, Hereditary , Humans , Rats , Mice , Animals , Child , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/therapy , Genetic Therapy , Dependovirus/genetics , Genetic Vectors , Paraplegia/genetics , Paraplegia/therapy
2.
Mol Biol Cell ; 33(12): ar109, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35976721

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex Subunits , Membrane Proteins , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex Subunits/metabolism , Adaptor Protein Complex gamma Subunits/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Membrane Proteins/metabolism , Protein Transport , trans-Golgi Network/metabolism
3.
Mol Biol Cell ; 32(21): ar25, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34432492

ABSTRACT

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.


Subject(s)
Autophagy-Related Proteins/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Vesicular Transport Proteins/metabolism , Autophagosomes/metabolism , Autophagy , Carrier Proteins/physiology , HEK293 Cells , HeLa Cells , Humans , Kinesins/metabolism , Microtubules/metabolism , Protein Transport/physiology , trans-Golgi Network/metabolism
4.
Biochem Soc Trans ; 48(5): 1877-1888, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33084855

ABSTRACT

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.


Subject(s)
Autophagy-Related Proteins/metabolism , Mutation , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , trans-Golgi Network/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Animals , Autophagosomes/metabolism , Autophagy , Binding Sites , Caenorhabditis elegans , Cryptococcus neoformans , Drosophila melanogaster , Fungi , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Protein Binding , Protein Conformation , Protein Transport , Transport Vesicles/metabolism , Tyrosine/chemistry , Vesicular Transport Proteins/metabolism
5.
Mol Biol Cell ; 31(9): 963-979, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32073997

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 4/metabolism , Autophagy-Related Proteins/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins/metabolism , trans-Golgi Network/metabolism , Cell Line, Tumor , Dyneins/metabolism , HEK293 Cells , HeLa Cells , Humans , Microtubules/metabolism , Protein Transport , Spastic Paraplegia, Hereditary/metabolism
6.
PLoS Genet ; 14(4): e1007363, 2018 04.
Article in English | MEDLINE | ID: mdl-29698489

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 4/deficiency , Adaptor Protein Complex 4/genetics , Autophagy-Related Proteins/metabolism , Membrane Proteins/metabolism , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Protein Complex 4/chemistry , Adaptor Protein Complex Subunits/chemistry , Adaptor Protein Complex Subunits/deficiency , Adaptor Protein Complex Subunits/genetics , Animals , Axons/metabolism , Behavior, Animal/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neurons/metabolism , Protein Aggregates/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Receptors, Glutamate/metabolism , Spastic Paraplegia, Hereditary/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , trans-Golgi Network/metabolism
7.
Curr Opin Neurobiol ; 51: 103-110, 2018 08.
Article in English | MEDLINE | ID: mdl-29558740

ABSTRACT

Selective transport of transmembrane proteins to different intracellular compartments often involves the recognition of sorting signals in the cytosolic domains of the proteins by components of membrane coats. Some of these coats have as their key components a family of heterotetrameric adaptor protein (AP) complexes named AP-1 through AP-5. AP complexes play important roles in all cells, but their functions are most critical in neurons because of the extreme compartmental complexity of these cells. Accordingly, various diseases caused by mutations in AP subunit genes exhibit a range of neurological abnormalities as their most salient features. In this article, we discuss the properties of the different AP complexes, with a focus on their roles in neuronal physiology and pathology.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Neurons/metabolism , Protein Transport/physiology , Action Potentials/genetics , Adaptor Proteins, Vesicular Transport/genetics , Animals , Humans , Mutation/genetics , Nervous System Diseases/genetics
8.
Proc Natl Acad Sci U S A ; 114(50): E10697-E10706, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29180427

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 4/metabolism , Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Protein Complex 4/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Motifs , Animals , Autophagy , Cell Line, Tumor , Gene Knockdown Techniques , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Models, Molecular , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
9.
Proc Natl Acad Sci U S A ; 113(36): E5318-27, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27559088

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Axons/metabolism , Neurons/metabolism , rab5 GTP-Binding Proteins/metabolism , Animals , Axons/pathology , Cell Polarity/genetics , Dynactin Complex/genetics , Dynactin Complex/metabolism , Dyneins/chemistry , Dyneins/metabolism , Endosomes/genetics , Endosomes/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Neurons/physiology , Protein Transport , Rats
10.
Cell ; 166(1): 193-208, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27293189

ABSTRACT

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.


Subject(s)
Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/analysis , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Presenilin-2/analysis , Adaptor Protein Complex 1/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Motifs , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Endosomes/chemistry , Humans , Lysosomes/chemistry , Mice , Presenilin-1/analysis , Presenilin-1/chemistry , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/chemistry , Presenilin-2/genetics , Presenilin-2/metabolism , Rats , Substrate Specificity
11.
Am J Hum Genet ; 97(5): 715-25, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544806

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 4/genetics , Genetic Predisposition to Disease , Mutation/genetics , Phosphoric Diester Hydrolases/genetics , Protein Transport/genetics , Stuttering/genetics , Stuttering/pathology , Asian People , Case-Control Studies , Female , Follow-Up Studies , Genetic Loci , Heterozygote , Humans , Male , Pedigree , Prognosis , trans-Golgi Network
12.
J Biol Chem ; 290(52): 30736-49, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26542808

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Protein Complex 4/chemistry , Adaptor Protein Complex 4/genetics , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Dimerization , Humans , Molecular Sequence Data , Sequence Alignment , trans-Golgi Network/metabolism
13.
PLoS Pathog ; 10(5): e1004107, 2014 May.
Article in English | MEDLINE | ID: mdl-24831812

ABSTRACT

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.


Subject(s)
Cell Polarity/physiology , Neurons/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Embryo, Mammalian , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Neurons/physiology , Nipah Virus , Protein Sorting Signals , Protein Structure, Tertiary , Protein Transport , Rats , Rats, Sprague-Dawley , Viral Envelope Proteins/chemistry , Virus Assembly
14.
Dev Cell ; 27(3): 353-66, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24229647

ABSTRACT

An outstanding question in protein sorting is why polarized epithelial cells express two isoforms of the µ1 subunit of the AP-1 clathrin adaptor complex: the ubiquitous µ1A and the epithelial-specific µ1B. Previous studies led to the notion that µ1A and µ1B mediate basolateral sorting predominantly from the trans-Golgi network (TGN) and recycling endosomes, respectively. Using improved analytical tools, however, we find that µ1A and µ1B largely colocalize with each other. They also colocalize to similar extents with TGN and recycling endosome markers, as well as with basolateral cargoes transiting biosynthetic and endocytic-recycling routes. Instead, the two isoforms differ in their signal-recognition specificity. In particular, µ1B preferentially binds a subset of signals from cargoes that are sorted basolaterally in a µ1B-dependent manner. We conclude that expression of distinct µ1 isoforms in epithelial cells expands the repertoire of signals recognized by AP-1 for sorting of a broader range of cargoes to the basolateral surface.


Subject(s)
Adaptor Protein Complex mu Subunits/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Epithelial Cells/metabolism , Kidney/metabolism , Transcription Factor AP-1/metabolism , trans-Golgi Network/metabolism , ADP-Ribosylation Factor 1/metabolism , Animals , Biomarkers , Cell Polarity , Cells, Cultured , Dogs , Image Processing, Computer-Assisted , Kidney/cytology , Mice , Microscopy, Fluorescence , Protein Isoforms , Protein Transport , Receptors, LDL/metabolism
15.
Neuron ; 75(5): 810-23, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22958822

ABSTRACT

Plasma membranes of the somatodendritic and axonal domains of neurons are known to have different protein compositions, but the molecular mechanisms that determine this polarized protein distribution remain poorly understood. Herein we show that somatodendritic sorting of various transmembrane receptors in rat hippocampal neurons is mediated by recognition of signals within the cytosolic domains of the proteins by the µ1A subunit of the adaptor protein-1 (AP-1) complex. This complex, in conjunction with clathrin, functions in the neuronal soma to exclude somatodendritic proteins from axonal transport carriers. Perturbation of this process affects dendritic spine morphology and decreases the number of synapses. These findings highlight the primary recognition event that underlies somatodendritic sorting and contribute to the evolving view of AP-1 as a global regulator of cell polarity.


Subject(s)
Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Cell Polarity/physiology , Clathrin/physiology , Hippocampus/physiology , Neurons/physiology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex mu Subunits/genetics , Amino Acid Sequence , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Hippocampus/cytology , Humans , Molecular Sequence Data , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/chemistry , Transgenes/physiology
16.
Dev Cell ; 22(4): 811-23, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22516199

ABSTRACT

Clathrin and the epithelial-specific clathrin adaptor AP-1B mediate basolateral trafficking in epithelia. However, several epithelia lack AP-1B, and mice knocked out for AP-1B are viable, suggesting the existence of additional mechanisms that control basolateral polarity. Here, we demonstrate a distinct role of the ubiquitous clathrin adaptor AP-1A in basolateral protein sorting. Knockdown of AP-1A causes missorting of basolateral proteins in MDCK cells, but only after knockdown of AP-1B, suggesting that AP-1B can compensate for lack of AP-1A. AP-1A localizes predominantly to the TGN, and its knockdown promotes spillover of basolateral proteins into common recycling endosomes, the site of function of AP-1B, suggesting complementary roles of both adaptors in basolateral sorting. Yeast two-hybrid assays detect interactions between the basolateral signal of transferrin receptor and the medium subunits of both AP-1A and AP-1B. The basolateral sorting function of AP-1A reported here establishes AP-1 as a major regulator of epithelial polarity.


Subject(s)
Adaptor Protein Complex 1/metabolism , Cell Polarity , Clathrin/metabolism , Endosomes/metabolism , Epithelial Cells/metabolism , trans-Golgi Network/physiology , Adaptor Protein Complex 1/antagonists & inhibitors , Adaptor Protein Complex 1/genetics , Animals , Cell Membrane/metabolism , Cells, Cultured , Dogs , Fluorescent Antibody Technique , Protein Transport , RNA, Small Interfering/genetics , Receptors, LDL/metabolism , Receptors, Transferrin/metabolism , Two-Hybrid System Techniques
17.
Proc Natl Acad Sci U S A ; 109(10): 3820-5, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22343291

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 1/chemistry , Receptors, Virus/chemistry , Adaptor Protein Complex 2/chemistry , Amino Acid Motifs , Animals , Cell Line , Cell Membrane/metabolism , Clathrin/chemistry , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Dogs , Endocytosis , Endosomes/metabolism , Epithelial Cells/cytology , Exocytosis , Fishes , Green Fluorescent Proteins/metabolism , Humans , Mutation , Protein Conformation , Protein Transport , Ranidae
18.
J Biol Chem ; 286(3): 2022-30, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21097499

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Polarity/physiology , Epithelial Cells/metabolism , Multiprotein Complexes/metabolism , Protein Sorting Signals/physiology , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Motifs , Binding Sites , Cell Line , Epithelial Cells/cytology , Humans , Multiprotein Complexes/genetics , Mutation
19.
J Virol ; 83(13): 6578-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19403684

ABSTRACT

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.


Subject(s)
CD4 Antigens/metabolism , Endosomes/metabolism , Lysosomes/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Down-Regulation , HIV-1/metabolism , HeLa Cells , Humans , RNA Interference , Ubiquitination
20.
J Virol ; 83(6): 2518-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129443

ABSTRACT

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.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , CD4 Antigens/metabolism , HIV-1/physiology , Protein Interaction Domains and Motifs , nef Gene Products, Human Immunodeficiency Virus/metabolism , Adaptor Protein Complex alpha Subunits/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation, Missense , Protein Binding , Protein Interaction Mapping , Protein Structure, Quaternary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...