Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters











Publication year range
1.
Neurosci Lett ; 808: 137282, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37127089

ABSTRACT

Two subtypes of alpha (α)subunits, α1and α2, belonging to AP-2 complex have been described in the central nervous system (CNS). The specific role of each subtype is still unclear. In this study, we evaluated the expression and interaction with cell membranes of both subtypes in the postnatal developing cerebral cortex and cerebellum in two rat strains that display distinct developmental features. We observed that α2 displays higher variations than α1 during development, and at lesser extent in the rats with delayed rate of development. Additionally, by in vitro binding assays we evaluated the interaction of α subunits with bovine brain membranes. Both subtypes displayed clear differences in their performance, maximum binding of α1 was higher and α2 reached it faster than α1. In addition, both subtypes displayed different binding to membranes when bivalent cations or nucleotides were added. We conclude that both subtypes interact differently with membranes and that they may play different roles in clathrin-mediated endocytosis in the CNS.


Subject(s)
Adaptor Protein Complex alpha Subunits , Endocytosis , Membrane Proteins , Animals , Cattle , Rats , Cell Membrane/metabolism , Central Nervous System/metabolism , Clathrin/metabolism , Endocytosis/physiology , Membrane Proteins/metabolism , Adaptor Protein Complex alpha Subunits/metabolism
2.
Sci Signal ; 15(759): eadf6187, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36346839
3.
Neuropathol Appl Neurobiol ; 48(2): e12776, 2022 02.
Article in English | MEDLINE | ID: mdl-34820873

ABSTRACT

AIMS: The heterotetrameric assembly protein complex 2 (AP-2) is a central hub for clathrin-dependent endocytosis. The AP-2 α-adaptin subunit has two major isoforms, encoded by two separate genes: AP2A1 and AP2A2. Endocytosis has been implicated in the pathogenesis of neurodegenerative disease, and recent studies linked α-adaptins (gene variants, splicing defects and altered expression) with late-onset Alzheimer's disease (LOAD) risk. Here, we used multiple antibodies to investigate α-adaptin isoforms and their localization in human brains. METHODS: The specificities of 10 different α-adaptin antibodies were evaluated using immunoblots after human AP2A1 and AP2A2 plasmid transfection in cultured cells. Additional immunoblot analyses were then performed on protein homogenates from control and LOAD subjects. Formalin-fixed, paraffin-embedded brain sections from control and LOAD subjects were immunohistochemically stained, and immunofluorescence experiments were performed for quantitation of colocalisation with digital image analysis. RESULTS: Eight of the 10 evaluated antibodies recognised transfected α-adaptin proteins on immunoblots. The α-adaptin subspecies were relatively uniformly expressed in five different human brain regions. The α-adaptins were present in the detergent-insoluble fraction from cognitively impaired, but less so in control, brains. Immunohistochemical analyses showed colocalisation of AP2A1 with tau pathology in LOAD brains. By contrast, AP2A2 colocalised with microglial cells. CONCLUSIONS: These observations provide evidence of isoform-specific changes of α-adaptins in the brains of LOAD subjects. Antibodies that were verified to recognise AP2A1, but not AP2A2, labelled neurofibrillary tangles of LOAD patients. The findings extend our understanding of AP-2 proteins in the human brain in healthy and diseased states.


Subject(s)
Adaptor Protein Complex alpha Subunits/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Neurofibrillary Tangles/metabolism , Protein Isoforms/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Brain/pathology , Female , Humans , Male , Middle Aged , Neurofibrillary Tangles/pathology
4.
Nat Commun ; 12(1): 5812, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608164

ABSTRACT

The advantage of locally applied anesthetics is that they are not associated with the many adverse effects, including addiction liability, of systemically administered analgesics. This therapeutic approach has two inherent pitfalls: specificity and a short duration of action. Here, we identified nociceptor endocytosis as a promising target for local, specific, and long-lasting treatment of inflammatory pain. We observed preferential expression of AP2α2, an α-subunit isoform of the AP2 complex, within CGRP+/IB4- nociceptors in rodents and in CGRP+ dorsal root ganglion neurons from a human donor. We utilized genetic and pharmacological approaches to inhibit nociceptor endocytosis demonstrating its role in the development and maintenance of acute and chronic inflammatory pain. One-time injection of an AP2 inhibitor peptide significantly reduced acute and chronic pain-like behaviors and provided prolonged analgesia. We evidenced sexually dimorphic recovery responses to this pharmacological approach highlighting the importance of sex differences in pain development and response to analgesics.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Chronic Pain/drug therapy , Endocytosis/drug effects , Nociceptors/drug effects , Adaptor Protein Complex 2/antagonists & inhibitors , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/antagonists & inhibitors , Adaptor Protein Complex alpha Subunits/genetics , Adaptor Protein Complex alpha Subunits/metabolism , Animals , Chronic Pain/metabolism , Chronic Pain/physiopathology , Epidermis/innervation , Female , Ganglia, Spinal/metabolism , Humans , Inflammation , Male , Mice , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Nociceptors/metabolism , Nociceptors/physiology , Peptides/administration & dosage , Peptides/metabolism , Peptides/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
5.
Cancer Res ; 81(17): 4455-4470, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34210752

ABSTRACT

In melanoma metastasis, the role of the AP-2α transcription factor, which is encoded by TFAP2A, is controversial as some findings have suggested tumor suppressor activity while other studies have shown high TFAP2A expression in node-positive melanoma associated with poor prognosis. Here we demonstrate that AP-2α facilitates melanoma metastasis through transcriptional activation of genes within the E2F pathway including EZH2. A BioID screen found that AP-2α interacts with members of the nucleosome remodeling and deacetylase (NuRD) complex. Loss of AP-2α removed activating chromatin marks in the promoters of EZH2 and other E2F target genes through activation of the NuRD repression complex. In melanoma cells, treatment with tazemetostat, an FDA-approved and highly specific EZH2 inhibitor, substantially reduced anchorage-independent colony formation and demonstrated heritable antimetastatic effects, which were dependent on AP-2α. Single-cell RNA sequencing analysis of a metastatic melanoma mouse model revealed hyperexpansion of Tfap2a High/E2F-activated cell populations in transformed melanoma relative to progenitor melanocyte stem cells. These findings demonstrate that melanoma metastasis is driven by the AP-2α/EZH2 pathway and suggest that AP-2α expression can be used as a biomarker to predict responsiveness to EZH2 inhibitors for the treatment of advanced melanomas. SIGNIFICANCE: AP-2α drives melanoma metastasis by upregulating E2F pathway genes including EZH2 through inhibition of the NuRD repression complex, serving as a biomarker to predict responsiveness to EZH2 inhibitors.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , E2F Transcription Factors/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Melanoma/metabolism , Animals , Base Sequence , Benzamides/pharmacology , Biomarkers/metabolism , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Epigenesis, Genetic , Humans , Melanocytes , Mice , Mice, Inbred NOD , Mice, SCID , Morpholines/pharmacology , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Second Primary , Promoter Regions, Genetic , Pyridones/pharmacology , Single-Cell Analysis , Transcription Factor AP-2
6.
Traffic ; 21(9): 603-616, 2020 09.
Article in English | MEDLINE | ID: mdl-32657003

ABSTRACT

Clathrin mediated endocytosis (CME) has been extensively studied in living cells by quantitative total internal reflection fluorescence microscopy (TIRFM). Fluorescent protein fusions to subunits of the major coat proteins, clathrin light chains or the heterotetrameric adaptor protein (AP2) complexes, have been used as fiduciary markers of clathrin coated pits (CCPs). However, the functionality of these fusion proteins has not been rigorously compared. Here, we generated stable cells lines overexpressing mRuby-CLCa and/or µ2-eGFP, σ2-eGFP, two markers currently in use, or a novel marker generated by inserting eGFP into the unstructured hinge region of the α subunit (α-eGFP). Using biochemical and TIRFM-based assays, we compared the functionality of the AP2 markers. All of the eGFP-tagged subunits were efficiently incorporated into AP2 and displayed greater accuracy in image-based CCP analyses than mRuby-CLCa. However, overexpression of either µ2-eGFP or σ2-eGFP impaired transferrin receptor uptake. In addition, µ2-eGFP reduced the rates of CCP initiation and σ2-eGFP perturbed AP2 incorporation into CCPs and CCP maturation. In contrast, CME and CCP dynamics were unperturbed in cells overexpressing α-eGFP. Moreover, α-eGFP was a more sensitive and accurate marker of CCP dynamics than mRuby-CLCa. Thus, our work establishes α-eGFP as a robust, fully functional marker for CME.


Subject(s)
Clathrin , Coated Pits, Cell-Membrane , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Endocytosis , Protein Binding
7.
J Invest Dermatol ; 140(9): 1743-1752.e4, 2020 09.
Article in English | MEDLINE | ID: mdl-32027876

ABSTRACT

IGF-1 receptor (IGF1R) signaling promotes keratinocyte proliferation, migration, and survival. However, the mechanism of IGF1R endocytosis in normal keratinocytes remains unclear. Confocal, super resolution structured illumination microscopy, total internal reflection fluorescence microscopy, and coimmunoprecipitation studies reveal that IGF1R associates with flotillin-1 (Flot-1), which currently has no known role in normal receptor tyrosine kinase endocytosis, under basal conditions in monolayer keratinocyte cultures. Ligand stimulation of IGF1R promotes its clathrin-dependent endocytosis, mediated by two distinct adaptors, Flot-1 in noncaveolar lipid rafts and the AP2A1/2 complex in clathrin vesicles. Concurrent, but not individual, short hairpin RNA knockdown of FLOT1/2 and AP2A1/2 reduced IGF1R association with clathrin, internalization, and pathway activation by more than 50% (of phosphorylated IGF1R, phosphorylated protein kinase B, and phosphorylated MAPK kinase), suggesting the complementarity of these two adaptor-specific pathways. The Flot-1 pathway is more responsive to low IGF-1 concentrations, whereas the AP2A1/2 pathway predominates at higher IGF-1 concentrations. Selective association of IGF1R-Flot-1-clathrin with Rab4, but IGF1R-AP2A1/2-clathrin with Rab11, implicates Flot-1 as the adaptor for faster recycling and AP2A1/2 as the adaptor for slower IGF1R recycling. These dual pathways, particularly flotillin-dependent, clathrin-mediated endocytosis, provide a new avenue for drug targeting in disorders with aberrant regulation of IGF1R signaling.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism , Receptor, IGF Type 1/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/genetics , Cells, Cultured , Clathrin/metabolism , Endocytosis , Gene Knockdown Techniques , Humans , Intravital Microscopy , Membrane Proteins/genetics , Microscopy, Fluorescence , Primary Cell Culture
8.
FASEB J ; 33(12): 13267-13279, 2019 12.
Article in English | MEDLINE | ID: mdl-31533003

ABSTRACT

Adipose tissue plays a major role in the regulation of systemic metabolic homeostasis, with the AP2 adaptor complex being important in clathrin-mediated endocytosis (CME) of various cell surface receptors, including glucose transporter 4, the insulin receptor, and ß-adrenergic receptors (ARs). One of the AP2 subunits, adaptor-related protein complex 2, α2 subunit (Ap2a2), has recently been identified as a peroxisome proliferator-activated receptor (PPAR)α target gene. The effects of PPARα on the AP2 adaptor complex and CME are unknown. We generated adipocyte-specific Ap2a2 knockout mice and investigated their metabolism when fed a standard chow or high-fat diet, without and with supplementation with the PPARα-agonist WY-14643 (WY). Although Ap2a2 deletion had only minor effects on glycaemic control, it led to substantial impairment in ß-adrenergic activation of lipolysis, as evidenced by a loss of cAMP response, PKA activation, and glycerol/fatty acid release. These differences were related to increased cell surface localization of the ß2- and ß3-ARs. Lipolytic defects were accompanied by impaired WY-mediated loss of fat mass and whole-body fat oxidation. This study demonstrates a novel role for PPARα in ß-adrenergic regulation of adipose tissue lipolysis and for adipose tissue in supplying adequate substrate to other peripheral tissues to accommodate the increase in systemic fatty acid oxidation that occurs upon treatment with PPARα agonists.-Montgomery, M. K., Bayliss, J., Keenan, S., Rhost, S., Ting, S. B., Watt, M. J. The role of Ap2a2 in PPARα-mediated regulation of lipolysis in adipose tissue.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Adipose Tissue/metabolism , PPAR alpha/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/genetics , Adipocytes/metabolism , Animals , Immunoblotting , Lipolysis/genetics , Lipolysis/physiology , Mice , Mice, Knockout
9.
Cell Rep ; 28(1): 11-20.e9, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31269433

ABSTRACT

Myosin VI is an actin-based cytoskeletal motor implicated in various steps of membrane trafficking. Here, we investigated whether this myosin is crucial for synaptic function and plasticity in neurons. We find that myosin VI localizes at cerebellar parallel fiber to Purkinje cell synapses and that the myosin is indispensable for long-term depression of AMPA-receptor-mediated synaptic signal transmission at this synapse. Moreover, direct visualization of GluA2-containing AMPA receptors in Purkinje cells reveals that the myosin drives removal of AMPA receptors from the surface of dendritic spines in an activity-dependent manner. Co-immunoprecipitation and super-resolution microscopy indicate that specifically the interaction of myosin VI with the clathrin adaptor component α-adaptin is important during long-term depression. Together, these data suggest that myosin VI directly promotes clathrin-mediated endocytosis of AMPA receptors in Purkinje cells to mediate cerebellar long-term depression. Our results provide insights into myosin VI function and the molecular mechanisms underlying synaptic plasticity.


Subject(s)
Cerebellum/metabolism , Long-Term Synaptic Depression , Myosin Heavy Chains/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Clathrin/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Endocytosis/genetics , Endocytosis/physiology , Hippocampus/cytology , Hippocampus/metabolism , Long-Term Synaptic Depression/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myosin Heavy Chains/antagonists & inhibitors , Myosin Heavy Chains/genetics , Purkinje Cells/metabolism , Receptors, AMPA/agonists , Receptors, AMPA/chemistry , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Synaptic Transmission/physiology
10.
Cell Prolif ; 52(1): e12522, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30443989

ABSTRACT

OBJECTIVES: Bone regeneration by bone tissue engineering is a therapeutic option for bone defects. Improving the osteogenic differentiation of mesenchymal stem cells (MSCs) is essential for successful bone regeneration. We previously showed that AP2a enhances the osteogenic differentiation in MSCs. The present study investigated the mechanism of how AP2a regulates the direct differentiation. MATERIALS AND METHODS: Co-immunoprecipitation and ChIP assays were carried out to investigate the underlying mechanism in MSCs differentiation. The osteogenic differentiation potential was determined by mineralization ability and the expression of osteogenic marker in vitro and the in vivo bone-like tissue generation in nude mice. RESULTS: We show that AP2a can compete with RUNX2, a key transcription factor in osteogenic differentiation, to recruit YAP and release the inhibition of RUNX2 activity from YAP by forming YAP-AP2a protein complex. YAP-AP2a protein complex also interacts with the BARX1 promoter through AP2a, inhibit the transcription of BARX1. Moreover, BARX1 inhibits osteogenic differentiation of MSCs. CONCLUSIONS: Our discoveries revealed that AP2a may regulate the osteogenic differentiation in an indirect way through competing with RUNX2 to relieve the RUNX2 activity which inhibited by YAP, and also in a direct way via targeting the BARX1 and directly repressed its transcription. Thus, our discoveries shed new light on the mechanism of direct differentiation of MSCs and provide candidate targets for improving the osteogenic differentiation and enhancing bone tissue regeneration.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Bone Regeneration/physiology , Core Binding Factor Alpha 1 Subunit/metabolism , Homeodomain Proteins/biosynthesis , Mesenchymal Stem Cells/cytology , Nuclear Proteins/metabolism , Osteogenesis/physiology , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Animals , Calcium Phosphates/metabolism , Cell Cycle Proteins , Cell Line , Durapatite/metabolism , Female , Gene Expression Regulation , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , RNA, Small Interfering/genetics , Tissue Engineering/methods , Tooth/cytology , Transcription Factors/genetics , Transcription, Genetic/genetics
11.
PLoS Biol ; 16(8): e2004506, 2018 08.
Article in English | MEDLINE | ID: mdl-30142146

ABSTRACT

Refinement of the nervous system depends on selective removal of excessive axons/dendrites, a process known as pruning. Drosophila ddaC sensory neurons prune their larval dendrites via endo-lysosomal degradation of the L1-type cell adhesion molecule (L1-CAM), Neuroglian (Nrg). Here, we have identified a novel gene, pruning defect 1 (prd1), which governs dendrite pruning of ddaC neurons. We show that Prd1 colocalizes with the clathrin adaptor protein α-Adaptin (α-Ada) and the kinesin-3 immaculate connections (Imac)/Uncoordinated-104 (Unc-104) in dendrites. Moreover, Prd1 physically associates with α-Ada and Imac, which are both critical for dendrite pruning. Prd1, α-Ada, and Imac promote dendrite pruning via the regulation of endo-lysosomal degradation of Nrg. Importantly, genetic interactions among prd1, α-adaptin, and imac indicate that they act in the same pathway to promote dendrite pruning. Our findings indicate that Prd1, α-Ada, and Imac act together to regulate discrete distribution of α-Ada/clathrin puncta, facilitate endo-lysosomal degradation, and thereby promote dendrite pruning in sensory neurons.


Subject(s)
Adaptor Protein Complex alpha Subunits/genetics , Cell Adhesion Molecules, Neuronal/genetics , Dendrites/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Kinesins/genetics , Neural Cell Adhesion Molecule L1/genetics , Neuronal Plasticity/genetics , Adaptor Protein Complex alpha Subunits/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Dendrites/ultrastructure , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Endosomes/metabolism , Endosomes/ultrastructure , Gene Expression Regulation, Developmental , Kinesins/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Lysosomes/metabolism , Lysosomes/ultrastructure , Metamorphosis, Biological/genetics , Neural Cell Adhesion Molecule L1/metabolism , Protein Binding , Proteolysis , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Signal Transduction
12.
Cell Host Microbe ; 23(6): 786-795.e5, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29902442

ABSTRACT

Listeriolysin O (LLO) is a cholesterol-dependent cytolysin that mediates escape of Listeria monocytogenes from a phagosome, enabling growth of the bacteria in the host cell cytosol. LLO contains a PEST-like sequence that prevents it from killing infected cells, but the mechanism involved is unknown. We found that the LLO PEST-like sequence was necessary to mediate removal of LLO from the interior face of the plasma membrane, where it coalesces into discrete puncta. LLO interacts with Ap2a2, an adaptor protein involved in endocytosis, via its PEST-like sequence, and Ap2a2-dependent endocytosis is required to prevent LLO-induced cytotoxicity. An unrelated PEST-like sequence from a human G protein-coupled receptor (GPCR), which also interacts with Ap2a2, could functionally complement the PEST-like sequence in L. monocytogenes LLO. These data revealed that LLO co-opts the host endocytosis machinery to protect the integrity of the host plasma membrane during L. monocytogenes infection.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Endocytosis/physiology , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/genetics , Animals , Colony Count, Microbial , Cytosol/metabolism , Cytosol/microbiology , Disease Models, Animal , Female , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/physiology , Humans , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Phagosomes/metabolism , Receptors, G-Protein-Coupled , Spleen/microbiology
13.
J Invest Dermatol ; 138(12): 2578-2588, 2018 12.
Article in English | MEDLINE | ID: mdl-29913133

ABSTRACT

Variants in IRF6 can lead to Van der Woude syndrome and popliteal pterygium syndrome. Furthermore, genes upstream and downstream of IRF6, including GRHL3 and TP63, are also associated with orofacial clefting. Additionally, a variant in an enhancer (MCS9.7) that regulates IRF6 is associated with risk for isolated orofacial clefting. This variant (rs642961) abrogates AP2A protein binding at MCS9.7. Here, we found that AP2A protein regulates MCS9.7 enhancer activity in vivo and IRF6 protein expression in epidermal development. In addition, loss of IRF6 leads to supra-basal expression of AP2A protein. Finally, using an IRF6 allelic series, we found that either increasing or decreasing IRF6 protein expression can destabilize AP2A protein expression in vivo. These data suggest that IRF6 regulates AP2A protein level in epidermal development. Therefore, we conclude that IRF6 and TFAP2A are part of a genetic regulatory network that is critical in epithelial development, with implications for both orofacial and cutaneous tissues. Our work provides in vivo, functional data to explain the relationship between AP2A protein binding and the MCS9.7 enhancer in orofacial clefting. This work is important because the MCS9.7 enhancer element contains a variant that abrogates AP2A protein binding and increases risk for orofacial clefting worldwide.


Subject(s)
Abnormalities, Multiple/genetics , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Cleft Lip/genetics , Cleft Palate/genetics , Cysts/genetics , Enhancer Elements, Genetic/genetics , Epidermis/physiology , Eye Abnormalities/genetics , Fingers/abnormalities , Interferon Regulatory Factors/genetics , Knee Joint/abnormalities , Lip/abnormalities , Lower Extremity Deformities, Congenital/genetics , Syndactyly/genetics , Urogenital Abnormalities/genetics , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/genetics , Alleles , Animals , Cells, Cultured , Gene Regulatory Networks , Humans , Mice , Mice, Transgenic , Organogenesis/genetics , Polymorphism, Single Nucleotide , Protein Binding , Protein Stability
14.
J Biol Chem ; 293(11): 4149-4158, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29382713

ABSTRACT

Traffic of cargo across membranes helps establish, maintain, and reorganize distinct cellular compartments and is fundamental to many metabolic processes. The cargo-selective endocytic adaptor Numb participates in clathrin-dependent endocytosis by attaching cargoes to the clathrin adaptor α-adaptin. The phosphorylation of Numb at Ser265 and Ser284 recruits the regulatory protein 14-3-3, accompanied by the dissociation of Numb from α-adaptin and Numb's translocation from the cortical membrane to the cytosol. However, the molecular mechanisms underlying the Numb-α-adaptin interaction and its regulation by Numb phosphorylation and 14-3-3 recruitment remain poorly understood. Here, biochemical and structural analyses of the Numb·14-3-3 complex revealed that Numb phosphorylation at both Ser265 and Ser284 is required for Numb's efficient interaction with 14-3-3. We also discovered that an RQFRF motif surrounding Ser265 in Numb functions together with the canonical C-terminal DPF motif, required for Numb's interaction with α-adaptin, to form a stable complex with α-adaptin. Of note, we provide evidence that the phosphorylation-induced binding of 14-3-3 to Numb directly competes with the binding of α-adaptin to Numb. Our findings suggest a potential mechanism governing the dynamic assembly of Numb with α-adaptin or 14-3-3. This dual-site recognition of Numb by α-adaptin may have implications for other α-adaptin targets. We propose that the newly identified α-adaptin-binding site surrounding Ser265 in Numb functions as a triggering mechanism for the dynamic dissociation of the Numb·α-adaptin complex.


Subject(s)
14-3-3 Proteins/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Endocytosis/physiology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , 14-3-3 Proteins/chemistry , Adaptor Protein Complex alpha Subunits/chemistry , Animals , Binding Sites , Catalysis , Catalytic Domain , Crystallography, X-Ray , Mice , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Structure-Activity Relationship
15.
Hum Mol Genet ; 27(5): 901-911, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29325022

ABSTRACT

Mutations of the sigma subunit of the heterotetrameric adaptor-related protein complex 2 (AP2σ) impair signalling of the calcium-sensing receptor (CaSR), and cause familial hypocalciuric hypercalcaemia type 3 (FHH3). To date, FHH3-associated AP2σ mutations have only been identified at one residue, Arg15. We hypothesized that additional rare AP2σ variants may also be associated with altered CaSR function and hypercalcaemia, and sought for these by analysing >111 995 exomes (>60 706 from ExAc and dbSNP, and 51 289 from the Geisinger Health System-Regeneron DiscovEHR dataset, which also contains clinical data). This identified 11 individuals to have 9 non-synonymous AP2σ variants (Arg3His, Arg15His (x3), Ala44Thr, Phe52Tyr, Arg61His, Thr112Met, Met117Ile, Glu122Gly and Glu142Lys) with 3 of the 4 individuals who had Arg15His and Met117Ile AP2σ variants having mild hypercalcaemia, thereby indicating a prevalence of FHH3-associated AP2σ mutations of ∼7.8 per 100 000 individuals. Structural modelling of the novel eight AP2σ variants (Arg3His, Ala44Thr, Phe52Tyr, Arg61His, Thr112Met, Met117Ile, Glu122Gly and Glu142Lys) predicted that the Arg3His, Thr112Met, Glu122Gly and Glu142Lys AP2σ variants would disrupt polar contacts within the AP2σ subunit or affect the interface between the AP2σ and AP2α subunits. Functional analyses of all eight AP2σ variants in CaSR-expressing cells demonstrated that the Thr112Met, Met117Ile and Glu142Lys variants, located in the AP2σ α4-α5 helical region that forms an interface with AP2α, impaired CaSR-mediated intracellular calcium (Cai2+) signalling, consistent with a loss of function, and this was rectified by treatment with the CaSR positive allosteric modulator cinacalcet. Thus, our studies demonstrate another potential class of FHH3-causing AP2σ mutations located at the AP2σ-AP2α interface.


Subject(s)
Adaptor Protein Complex alpha Subunits/metabolism , Adaptor Protein Complex sigma Subunits/genetics , Mutation , Receptors, Calcium-Sensing/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex sigma Subunits/metabolism , Cinacalcet/pharmacology , Databases, Genetic , Exome , Female , Humans , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Male , Middle Aged , Models, Molecular , Protein Conformation , Signal Transduction , Exome Sequencing
16.
J Cell Biol ; 216(1): 167-179, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-28003333

ABSTRACT

The critical initiation phase of clathrin-mediated endocytosis (CME) determines where and when endocytosis occurs. Heterotetrameric adaptor protein 2 (AP2) complexes, which initiate clathrin-coated pit (CCP) assembly, are activated by conformational changes in response to phosphatidylinositol-4,5-bisphosphate (PIP2) and cargo binding at multiple sites. However, the functional hierarchy of interactions and how these conformational changes relate to distinct steps in CCP formation in living cells remains unknown. We used quantitative live-cell analyses to measure discrete early stages of CME and show how sequential, allosterically regulated conformational changes activate AP2 to drive both nucleation and subsequent stabilization of nascent CCPs. Our data establish that cargoes containing Yxxφ motif, but not dileucine motif, play a critical role in the earliest stages of AP2 activation and CCP nucleation. Interestingly, these cargo and PIP2 interactions are not conserved in yeast. Thus, we speculate that AP2 has evolved as a key regulatory node to coordinate CCP formation and cargo sorting and ensure high spatial and temporal regulation of CME.


Subject(s)
Adaptor Protein Complex 2/metabolism , Clathrin-Coated Vesicles/metabolism , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Endocytosis , Retinal Pigment Epithelium/metabolism , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/genetics , Adaptor Protein Complex alpha Subunits/metabolism , Adaptor Protein Complex mu Subunits/genetics , Adaptor Protein Complex mu Subunits/metabolism , Amino Acid Motifs , Cell Line , Humans , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Protein Transport , RNA Interference , Signal Transduction , Structure-Activity Relationship , Time Factors , Transfection
17.
Sci Rep ; 6: 19565, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26822536

ABSTRACT

FCHo1, FCHo2, and SGIP1 are key regulators of clathrin-mediated endocytosis. Their µ homology domains (µHDs) interact with the C-terminal region of an endocytic scaffold protein, Eps15, containing fifteen Asp-Pro-Phe (DPF) motifs. Here, we show that the high-affinity µHD-binding site in Eps15 is a region encompassing six consecutive DPF motifs, while the minimal µHD-binding unit is two consecutive DPF motifs. We present the crystal structures of the SGIP1 µHD in complex with peptides containing two DPF motifs. The peptides bind to a novel ligand-binding site of the µHD, which is distinct from those of other distantly related µHD-containing proteins. The two DPF motifs, which adopt three-dimensional structures stabilized by sequence-specific intramotif and intermotif interactions, are extensively recognized by the µHD and are both required for binding. Thus, consecutive and singly scattered DPF motifs play distinct roles in µHD binding.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Structural Homology, Protein , Adaptor Protein Complex alpha Subunits/chemistry , Adaptor Protein Complex alpha Subunits/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acids/metabolism , Binding Sites , Calorimetry , Crystallography, X-Ray , Humans , Ligands , Membrane Proteins , Models, Molecular , Protein Binding , Protein Domains , Structure-Activity Relationship
18.
J Cell Biol ; 210(6): 933-50, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26370500

ABSTRACT

During mammalian T cell development, the requirement for expansion of many individual T cell clones, rather than merely expansion of the entire T cell population, suggests a possible role for asymmetric cell division (ACD). We show that ACD of developing T cells controls cell fate through differential inheritance of cell fate determinants Numb and α-Adaptin. ACD occurs specifically during the ß-selection stage of T cell development, and subsequent divisions are predominantly symmetric. ACD is controlled by interaction with stromal cells and chemokine receptor signaling and uses a conserved network of polarity regulators. The disruption of polarity by deletion of the polarity regulator, Scribble, or the altered inheritance of fate determinants impacts subsequent fate decisions to influence the numbers of DN4 cells arising after the ß-selection checkpoint. These findings indicate that ACD enables the thymic microenvironment to orchestrate fate decisions related to differentiation and self-renewal.


Subject(s)
Asymmetric Cell Division , Cell Proliferation , Thymocytes/metabolism , Thymus Gland/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Animals , Cell Communication , Cell Death , Cell Differentiation , Cell Polarity , Cells, Cultured , Cellular Microenvironment , Coculture Techniques , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Nerve Tissue Proteins/metabolism , Phosphorylation , Protein Kinase C/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Stromal Cells/immunology , Stromal Cells/metabolism , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Time Factors , Transfection
19.
Article in Chinese | MEDLINE | ID: mdl-25966562

ABSTRACT

OBJECTIVE: To investigate the expression of adaptin-2(AP-2) in mice cochlea and to discuss the probable role in the endocytosis of hair cells. METHOD: Laser scanning confocal microscopy and immune-fluroscence histochemistry were performed in this study. RESULT: In mature mice cochlea, the immunoreactivity for AP-2 was found in the inner hair cells cytoplasm. This protein mainly expressed in the hair cells basal part and nearby the ribbon synapse. CONCLUSION: AP-2 protein mainly expressed in the hair cells synaptic activity zone , which suggested that AP-2 could play an important role in the synaptic vesicle endocytosis. This finding built the foundation for the further research involved in the physiological and pathological role of AP-2.


Subject(s)
Adaptor Protein Complex alpha Subunits/metabolism , Hair Cells, Auditory, Inner/metabolism , Animals , Cochlea , Hair Cells, Auditory , Mice , Microscopy, Confocal , Synapses
20.
J Cell Biol ; 204(7): 1141-56, 2014 Mar 31.
Article in English | MEDLINE | ID: mdl-24662566

ABSTRACT

Dynamin is a well-known regulator of synaptic endocytosis. Temperature-sensitive dynamin (shi(ts1)) mutations in Drosophila melanogaster or deletion of some of the mammalian Dynamins causes the accumulation of invaginated endocytic pits at synapses, sometimes also on bulk endosomes, indicating impaired membrane scission. However, complete loss of dynamin function has not been studied in neurons in vivo, and whether Dynamin acts in different aspects of synaptic vesicle formation remains enigmatic. We used acute photoinactivation and found that loss of Dynamin function blocked membrane recycling and caused the buildup of huge membrane-connected cisternae, in contrast to the invaginated pits that accumulate in shi(ts1) mutants. Moreover, photoinactivation of Dynamin in shi(ts1) animals converted these pits into bulk cisternae. Bulk membrane retrieval has also been seen upon Clathrin photoinactivation, and superresolution imaging indicated that acute Dynamin photoinactivation blocked Clathrin and α-adaptin relocalization to synaptic membranes upon nerve stimulation. Hence, our data indicate that Dynamin is critically involved in the stabilization of Clathrin- and AP2-dependent endocytic pits.


Subject(s)
Adaptor Protein Complex alpha Subunits/metabolism , Cell Membrane/metabolism , Clathrin/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/cytology , Dynamins/physiology , Adaptor Protein Complex 2/metabolism , Animals , Cell Membrane/ultrastructure , Cells, Cultured , Endocytosis , Fluorescein/chemistry , Larva/cytology , Neurons/physiology , Neurons/ultrastructure , Photochemical Processes , Protein Transport , Synaptic Vesicles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL