Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 17(6): e1009607, 2021 06.
Article in English | MEDLINE | ID: mdl-34081703

ABSTRACT

Early endosomes are the sorting hub on the endocytic pathway, wherein sorting nexins (SNXs) play important roles for formation of the distinct membranous microdomains with different sorting functions. Tubular endosomes mediate the recycling of clathrin-independent endocytic (CIE) cargoes back toward the plasma membrane. However, the molecular mechanism underlying the tubule formation is still poorly understood. Here we screened the effect on the ARF-6-associated CIE recycling endosomal tubules for all the SNX members in Caenorhabditis elegans (C. elegans). We identified SNX-3 as an essential factor for generation of the recycling tubules. The loss of SNX-3 abolishes the interconnected tubules in the intestine of C. elegans. Consequently, the surface and total protein levels of the recycling CIE protein hTAC are strongly decreased. Unexpectedly, depletion of the retromer components VPS-26/-29/-35 has no similar effect, implying that the retromer trimer is dispensable in this process. We determined that hTAC is captured by the ESCRT complex and transported into the lysosome for rapid degradation in snx-3 mutants. Interestingly, EEA-1 is increasingly recruited on early endosomes and localized to the hTAC-containing structures in snx-3 mutant intestines. We also showed that SNX3 and EEA1 compete with each other for binding to phosphatidylinositol-3-phosphate enriching early endosomes in Hela cells. Our data demonstrate for the first time that PX domain-only C. elegans SNX-3 organizes the tubular endosomes for efficient recycling and retrieves the CIE cargo away from the maturing sorting endosomes by competing with EEA-1 for binding to the early endosomes. However, our results call into question how SNX-3 couples the cargo capture and membrane remodeling in the absence of the retromer trimer complex.


Subject(s)
ADP-Ribosylation Factors/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Endosomes/metabolism , Epithelial Cells/metabolism , Sorting Nexins/genetics , Vesicular Transport Proteins/genetics , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Biological Transport , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Membrane/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Cells/cytology , Gene Expression Regulation , HeLa Cells , Humans , Intestines/cytology , Lysosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , Signal Transduction , Sorting Nexins/deficiency , Vesicular Transport Proteins/metabolism
2.
Nature ; 589(7842): 456-461, 2021 01.
Article in English | MEDLINE | ID: mdl-33328639

ABSTRACT

Autophagy, a process of degradation that occurs via the lysosomal pathway, has an essential role in multiple aspects of immunity, including immune system development, regulation of innate and adaptive immune and inflammatory responses, selective degradation of intracellular microorganisms, and host protection against infectious diseases1,2. Autophagy is known to be induced by stimuli such as nutrient deprivation and suppression of mTOR, but little is known about how autophagosomal biogenesis is initiated in mammalian cells in response to viral infection. Here, using genome-wide short interfering RNA screens, we find that the endosomal protein sorting nexin 5 (SNX5)3,4 is essential for virus-induced, but not for basal, stress- or endosome-induced, autophagy. We show that SNX5 deletion increases cellular susceptibility to viral infection in vitro, and that Snx5 knockout in mice enhances lethality after infection with several human viruses. Mechanistically, SNX5 interacts with beclin 1 and ATG14-containing class III phosphatidylinositol-3-kinase (PI3KC3) complex 1 (PI3KC3-C1), increases the lipid kinase activity of purified PI3KC3-C1, and is required for endosomal generation of phosphatidylinositol-3-phosphate (PtdIns(3)P) and recruitment of the PtdIns(3)P-binding protein WIPI2 to virion-containing endosomes. These findings identify a context- and organelle-specific mechanism-SNX5-dependent PI3KC3-C1 activation at endosomes-for initiation of autophagy during viral infection.


Subject(s)
Autophagy/immunology , Sorting Nexins/metabolism , Viruses/immunology , Animals , Autophagy/genetics , Autophagy-Related Proteins/metabolism , Beclin-1/metabolism , Cell Line , Class III Phosphatidylinositol 3-Kinases/metabolism , Endosomes/metabolism , Female , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Sorting Nexins/deficiency , Sorting Nexins/genetics , Vesicular Transport Proteins/metabolism
3.
Circ Res ; 127(4): 534-549, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32316875

ABSTRACT

RATIONALE: SNX10 (sorting nexin 10) has been reported to play a critical role in regulating macrophage function and lipid metabolism. OBJECTIVE: To investigate the precise role of SNX10 in atherosclerotic diseases and the underlying mechanisms. METHODS AND RESULTS: SNX10 expression was compared between human healthy vessels and carotid atherosclerotic plaques. Myeloid cell-specific SNX10 knockdown mice were crossed onto the APOE-/- (apolipoprotein E) background and atherogenesis (high-cholesterol diet-induced) was monitored for 16 weeks. We found that SNX10 expression was increased in atherosclerotic lesions of aortic specimens from humans and APOE-/- mice. Myeloid cell-specific SNX10 deficiency (Δ knockout [KO]) attenuated atherosclerosis progression in APOE-/- mice. The population of anti-inflammatory monocytes/macrophages was increased in the peripheral blood and atherosclerotic lesions of ΔKO mice. In vitro experiments showed that SNX10 deficiency-inhibited foam cell formation through interrupting the internalization of CD36, which requires the interaction of SNX10 and Lyn-AKT (protein kinase B). The reduced Lyn-AKT activation by SNX10 deficiency promoted the nuclear translocation of TFEB (transcription factor EB), thereby enhanced lysosomal biogenesis and LAL (lysosomal acid lipase) activity, resulting in an increase of free fatty acids to fuel mitochondrial fatty acid oxidation. This further promoted the reprogramming of macrophages and shifted toward the anti-inflammatory phenotype. CONCLUSIONS: Our data demonstrate for the first time that SNX10 plays a crucial role in diet-induced atherogenesis via the previously unknown link between the Lyn-Akt-TFEB signaling pathway and macrophage reprogramming, suggest that SNX10 may be a potentially promising therapeutic target for atherosclerosis treatment.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cellular Reprogramming/physiology , Macrophages/physiology , Sorting Nexins/physiology , Animals , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , CD36 Antigens/metabolism , Cell Nucleus/metabolism , Disease Progression , Fatty Acids, Nonesterified/metabolism , Foam Cells/cytology , Humans , Lysosomes/physiology , Macrophages/cytology , Mice , Mitochondria/metabolism , Monocytes/cytology , Oxidation-Reduction , Proto-Oncogene Proteins c-akt/metabolism , Sorting Nexins/deficiency , Sorting Nexins/genetics , Sterol Esterase/metabolism
4.
Autophagy ; 16(4): 735-749, 2020 04.
Article in English | MEDLINE | ID: mdl-31208298

ABSTRACT

The non-receptor tyrosine kinase SRC is a key mediator of cellular protumorigenic signals. SRC is aberrantly over-expressed and activated in more than 80% of colorectal cancer (CRC) patients, therefore regulation of its stability and activity is essential. Here, we report a significant down regulation of SNX10 (sorting nexin 10) in human CRC tissues, which is closely related to tumor differentiation, TNM stage, lymph node metastasis and survival period. SNX10 deficiency in normal and neoplastic colorectal epithelial cells promotes initiation and progression of CRC in mice. SNX10 controls SRC levels by mediating autophagosome-lysosome fusion and SRC recruitment for autophagic degradation. These mechanisms ensure proper controlling of the activities of SRC-STAT3 and SRC-CTNNB1 signaling pathways by up-regulating SNX10 expression under stress conditions. These findings suggest that SNX10 acts as a tumor suppressor in CRC and it could be a potential therapeutic target for future development.Abbreviations: ACTB: actin beta; ATG5: autophagy related 5; ATG12: autophagy related 12; CQ: chloroquine; CRC: colorectal cancer; CTNNB1: catenin beta 1; EBSS: Earle's balanced salt solution; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LAMP2: lysosomal associated membrane protein 2; MAP1LC3: microtubule associated protein 1 light chain 3; MKI67: marker of proliferation Ki-67; mRNA: messenger RNA; PX: phox homology; RT-qPCR: real time quantitative polymerase chain reaction; siRNA: small interfering RNA; SNX10: sorting nexin 10; SQSTM1: sequestosome 1; SRC: SRC proto-oncogene, non-receptor tyrosine kinase; STAT3: signal transducer and activator of transcription 3; WT: wild type.


Subject(s)
Autophagy/physiology , Colorectal Neoplasms/metabolism , Sorting Nexins/metabolism , Animals , Autophagy/genetics , Cell Movement/physiology , Colorectal Neoplasms/pathology , Down-Regulation , Humans , Lysosomes/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Mas , Sorting Nexins/deficiency , Sorting Nexins/genetics , Up-Regulation
5.
Cell Death Dis ; 9(6): 666, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29867114

ABSTRACT

Amino-acid metabolism plays a vital role in mammalian target of rapamycin (mTOR) signaling, which is the pivot in colorectal cancer (CRC). Upregulated chaperone-mediated autophagy (CMA) activity contributes to the regulation of metabolism in cancer cells. Previously, we found that sorting nexin 10 (SNX10) is a critical regulator in CMA activation. Here we investigated the role of SNX10 in regulating amino-acid metabolism and mTOR signaling pathway activation, as well as the impact on the tumor progression of mouse CRC. Our results showed that SNX10 deficiency promoted colorectal tumorigenesis in male FVB mice and CRC cell proliferation and survival. Metabolic pathway analysis of gas chromatography-mass spectrometry (GC-MS) data revealed unique changes of amino-acid metabolism by SNX10 deficiency. In HCT116 cells, SNX10 knockout resulted in the increase of CMA and mTOR activation, which could be abolished by chloroquine treatment or reversed by SNX10 overexpression. By small RNA interference (siRNA), we found that the activation of mTOR was dependent on lysosomal-associated membrane protein type-2A (LAMP-2A), which is a limiting factor of CMA. Similar results were also found in Caco-2 and SW480 cells. Ultra-high-performance liquid chromatography-quadrupole time of flight (UHPLC-QTOF) and GC-MS-based untargeted metabolomics revealed that 10 amino-acid metabolism in SNX10-deficient cells were significantly upregulated, which could be restored by LAMP-2A siRNA. All of these amino acids were previously reported to be involved in mTOR activation. In conclusion, this work revealed that SNX10 controls mTOR activation through regulating CMA-dependent amino-acid metabolism, which provides potential target and strategy for treating CRC.


Subject(s)
Amino Acids/metabolism , Colorectal Neoplasms/metabolism , Sorting Nexins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy , Caco-2 Cells , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cell Survival , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Lysosomal-Associated Membrane Protein 2/metabolism , Male , Metabolomics , Mice, Knockout , Models, Biological , Molecular Chaperones/metabolism , Multivariate Analysis , Nucleotides/metabolism , Signal Transduction , Sorting Nexins/deficiency , Up-Regulation
6.
Hum Mol Genet ; 27(11): 1927-1940, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29635513

ABSTRACT

Mutations in SNX14 cause the autosomal recessive cerebellar ataxia 20 (SCAR20). Mutations generally result in loss of protein although several coding region deletions have also been reported. Patient-derived fibroblasts show disrupted autophagy, but the precise function of SNX14 is unknown. The yeast homolog, Mdm1, functions in endoplasmic reticulum (ER)-lysosome/vacuole inter-organelle tethering, but functional conservation in mammals is still required. Here, we show that loss of SNX14 alters but does not block autophagic flux. In addition, we find that SNX14 is an ER-associated protein that functions in neutral lipid homeostasis and inter-organelle crosstalk. SNX14 requires its N-terminal transmembrane helices for ER localization, while the Phox homology (PX) domain is dispensable for subcellular localization. Both SNX14-mutant fibroblasts and SNX14KO HEK293 cells accumulate aberrant cytoplasmic vacuoles, suggesting defects in endolysosomal homeostasis. However, ER-late endosome/lysosome contact sites are maintained in SNX14KO cells, indicating that it is not a prerequisite for ER-endolysosomal tethering. Further investigation of SNX14- deficiency indicates general defects in neutral lipid metabolism. SNX14KO cells display distinct perinuclear accumulation of filipin in LAMP1-positive lysosomal structures indicating cholesterol accumulation. Consistent with this, SNX14KO cells display a slight but detectable decrease in cholesterol ester levels, which is exacerbated with U18666A. Finally, SNX14 associates with ER-derived lipid droplets (LD) following oleate treatment, indicating a role in ER-LD crosstalk. We therefore identify an important role for SNX14 in neutral lipid homeostasis between the ER, lysosomes and LDs that may provide an early intervention target to alleviate the clinical symptoms of SCAR20.


Subject(s)
Endoplasmic Reticulum/genetics , Lipid Metabolism/genetics , Sorting Nexins/genetics , Spinocerebellar Ataxias/genetics , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Endosomes , Gene Knockout Techniques , HEK293 Cells , Homeostasis/drug effects , Humans , Intermediate Filament Proteins/genetics , Lipid Droplets/metabolism , Lysosomes/drug effects , Lysosomes/genetics , Mutation , Oleic Acid/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sorting Nexins/deficiency , Sorting Nexins/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/physiopathology
7.
Proc Natl Acad Sci U S A ; 114(49): 13000-13005, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29180417

ABSTRACT

IFNγ is a cytokine that plays a key role in host defense against intracellular pathogens. In addition to the canonical JAK-STAT1 pathway, IFNγ also activates an IKKß-mediated noncanonical signaling pathway that is essential for induction of a subset of downstream effector genes. The molecular mechanisms and functional significance of this IFNγ-triggered noncanonical pathway remains enigmatic. Here, we identified sorting nexin 8 (SNX8) as an important component of the IFNγ-triggered noncanonical signaling pathway. SNX8-deficiency impaired IFNγ-triggered induction of a subset of downstream genes. Snx8-/- mice infected with Listeria monocytogenes exhibited lower serum cytokine levels and higher bacterial loads in the livers and spleens, resulting in higher lethality. Mechanistically, SNX8 interacted with JAK1 and IKKß and promoted their association. IFNγ induced JAK1-mediated phosphorylation of SNX8 at Tyr95 and Tyr126, which promoted the recruitment of IKKß to the JAK1 complex. SNX8-deficiency impaired IFNγ-induced oligomerization and autophosphorylation of IKKß at Ser177, which is critical for selective induction of downstream genes. Our findings suggest that SNX8 acts as a link for IFNγ-triggered noncanonical signaling pathway, which induces a subset of downstream genes important for host defense against L. monocytogenes infection.


Subject(s)
I-kappa B Kinase/immunology , Janus Kinase 1/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Sorting Nexins/immunology , Animals , Bacterial Load , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Janus Kinase 1/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/mortality , Liver/immunology , Liver/microbiology , Mice , Mice, Knockout , Peptidoglycan/administration & dosage , Phosphorylation , Signal Transduction , Sorting Nexins/deficiency , Sorting Nexins/genetics , Spleen/immunology , Spleen/microbiology , Survival Analysis , THP-1 Cells
8.
Elife ; 62017 01 30.
Article in English | MEDLINE | ID: mdl-28134614

ABSTRACT

SNX6 is a ubiquitously expressed PX-BAR protein that plays important roles in retromer-mediated retrograde vesicular transport from endosomes. Here we report that CNS-specific Snx6 knockout mice exhibit deficits in spatial learning and memory, accompanied with loss of spines from distal dendrites of hippocampal CA1 pyramidal cells. SNX6 interacts with Homer1b/c, a postsynaptic scaffold protein crucial for the synaptic distribution of other postsynaptic density (PSD) proteins and structural integrity of dendritic spines. We show that SNX6 functions independently of retromer to regulate distribution of Homer1b/c in the dendritic shaft. We also find that Homer1b/c translocates from shaft to spines by protein diffusion, which does not require SNX6. Ablation of SNX6 causes reduced distribution of Homer1b/c in distal dendrites, decrease in surface levels of AMPAR and impaired AMPAR-mediated synaptic transmission. These findings reveal a physiological role of SNX6 in CNS excitatory neurons.


Subject(s)
CA1 Region, Hippocampal/physiology , Pyramidal Cells/physiology , Sorting Nexins/deficiency , Spatial Memory , Synapses/physiology , Animals , Homer Scaffolding Proteins/metabolism , Mice, Knockout , Receptors, Glutamate/metabolism , Sorting Nexins/metabolism
9.
Glia ; 64(8): 1437-60, 2016 08.
Article in English | MEDLINE | ID: mdl-27270750

ABSTRACT

The G protein-coupled receptor 17 (GPR17) plays crucial roles in myelination. It is highly expressed during transition of oligodendrocyte progenitor cells to immature oligodendrocytes, but, after this stage, it must be down-regulated to allow generation of mature myelinating cells. After endocytosis, GPR17 is sorted into lysosomes for degradation or recycled to the plasma membrane. Balance between degradation and recycling is important for modulation of receptor levels at the cell surface and thus for the silencing/activation of GPR17-signaling pathways that, in turn, affect oligodendrocyte differentiation. The molecular mechanisms at the basis of these processes are still partially unknown and their characterization will allow a better understanding of myelination and provide cues to interpret the consequences of GPR17 dysfunction in diseases. Here, we demonstrate that the endocytic trafficking of GPR17 is mediated by the interaction of a type I PDZ-binding motif located at the C-terminus of the receptor and SNX27, a recently identified protein of the endosome-associated retromer complex and whose functions in oligodendrocytes have never been studied. SNX27 knock-down significantly reduces GPR17 plasma membrane recycling in differentiating oligodendrocytes while accelerating cells' terminal maturation. Interestingly, trisomy-linked down-regulation of SNX27 expression in the brain of Ts65Dn mice, a model of Down syndrome, correlates with a decrease in GPR17(+) cells and an increase in mature oligodendrocytes, which, however, fail in reaching full maturation, eventually leading to hypomyelination. Our data demonstrate that SNX27 modulates GPR17 plasma membrane recycling and stability, and that disruption of the SNX27/GPR17 interaction might contribute to pathological oligodendrocyte differentiation defects. GLIA 2016. GLIA 2016;64:1437-1460.


Subject(s)
Biological Transport/physiology , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Sorting Nexins/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Differentiation/physiology , Cell Line , Cell Membrane/metabolism , Cell Membrane/pathology , Disease Models, Animal , Down Syndrome/metabolism , Down Syndrome/pathology , Endocytosis/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendroglia/pathology , Sorting Nexins/deficiency , Sorting Nexins/genetics
10.
Ann Rheum Dis ; 75(6): 1211-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26141367

ABSTRACT

OBJECTIVE: Periarticular and subchondral bone erosion in rheumatoid arthritis caused by osteoclast differentiation and activation is a critical index for diagnosis, therapy and monitoring of the disease. Sorting nexin (SNX) 10, a member of the SNX family which functions in regulation of endosomal sorting, has been implicated to play an important clinical role in malignant osteopetrosis. Here we studied the roles and precise mechanisms of SNX10 in the bone destruction of collagen-induced arthritis (CIA) mice. METHODS: The role of SNX10 in bone destruction was evaluated by a CIA mice model which was induced in male SNX10(-/-) mice and wild type littermates. The mechanism was explored in osteoclasts induced by receptor activator of nuclear factor κB ligand from bone marrow mononuclear cells of wild type and SNX10(-/-) mice. RESULTS: SNX10 knockout prevented bone loss and joint destruction in CIA mice with reduced serum levels of TNF-α, interleukin 1ß and anticollagen IgG 2α antibody. SNX10 deficiency did not block osteoclastogenesis, but significantly impaired osteoclast maturation and bone-resorption function by disturbing the formation of actin belt. The production of TRAP, CtsK and MMP9 in SNX10(-/-) osteoclasts was significantly inhibited, and partially restored by SNX10 overexpression. We further demonstrated that the degradation of NFATc1 was accelerated in SNX10(-/-) osteoclasts causing an inhibition of integrin ß3-Src-PYK2 signalling. CONCLUSIONS: Our study discloses a crucial role and novel mechanism for SNX10 in osteoclast function, and provides evidence for SNX10 as a promising novel therapeutic target for suppression of immune inflammation and bone erosion in rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/complications , Bone Resorption/prevention & control , NFATC Transcription Factors/metabolism , Sorting Nexins/physiology , Animals , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Bone Resorption/diagnostic imaging , Bone Resorption/etiology , Bone Resorption/pathology , Integrin beta3/physiology , Male , Mice, Knockout , Osteoclasts/pathology , Osteoclasts/physiology , Osteogenesis/physiology , Signal Transduction/physiology , Sorting Nexins/deficiency , Synovial Membrane/pathology , Tomography, X-Ray Computed
11.
Neurogenetics ; 16(3): 215-221, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25894286

ABSTRACT

The composition of the neuronal cell surface dictates synaptic plasticity and thereby cognitive development. This remodeling of the synapses is governed by the endocytic network which internalize transmembrane proteins, then sort them back to the cell surface or carry them to the lysosome for degradation. The multi-protein retromer complex is central to this selection, capturing specific transmembrane proteins and remodeling the cell membrane to form isolated cargo-enriched transport carriers. We investigated a consanguineous family with four patients who presented in infancy with intractable myoclonic epilepsy and lack of psychomotor development. Using exome analysis, we identified a homozygous deleterious mutation in SNX27, which encodes sorting nexin 27, a retromer cargo adaptor. In western analysis of patient fibroblasts, the encoded mutant protein was expressed at an undetectable level when compared with a control sample. The patients' presentation and clinical course recapitulate that reported for the SNX27 knock-out mouse. Since the cargo proteins for SNX27-mediated sorting include subunits of ionotropic glutamate receptors and endosome-to-cell surface synaptic insertion of AMPA receptors is severely perturbed in SNX27(-/-) neurons, it is proposed that at least part of the neurological aberrations observed in the patients is attributed to defective sorting of ionotropic glutamate receptors. SNX27 deficiency is now added to the growing list of neurodegenerative disorders associated with retromer dysfunction.


Subject(s)
Epilepsies, Myoclonic/genetics , Neurodegenerative Diseases/genetics , Sorting Nexins/deficiency , Sorting Nexins/genetics , Brain/pathology , Brain/physiopathology , Female , Fibroblasts/metabolism , Humans , Infant , Infant, Newborn , Male , Mutation , Pedigree
12.
Neuron ; 82(3): 659-69, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24811384

ABSTRACT

The subcellular pathways that regulate G protein-gated inwardly rectifying potassium (GIRK or Kir3) channels are important for controlling the excitability of neurons. Sorting nexin 27 (SNX27) is a PDZ-containing protein known to bind GIRK2c/GIRK3 channels, but its function in vivo is poorly understood. Here, we investigated the role of SNX27 in regulating GIRK currents in dopamine (DA) neurons of the ventral tegmental area (VTA). Mice lacking SNX27 in DA neurons exhibited reduced GABABR-activated GIRK currents but had normal Ih currents and DA D2R-activated GIRK currents. Expression of GIRK2a, an SNX27-insensitive splice variant, restored GABABR-activated GIRK currents in SNX27-deficient DA neurons. Remarkably, mice with significantly reduced GABABR-activated GIRK currents in only DA neurons were hypersensitive to cocaine and could be restored to a normal locomotor response with GIRK2a expression. These results identify a pathway for regulating excitability of VTA DA neurons, highlighting SNX27 as a promising target for treating addiction.


Subject(s)
Cocaine/toxicity , G Protein-Coupled Inwardly-Rectifying Potassium Channels/biosynthesis , Sorting Nexins/physiology , Animals , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Organ Culture Techniques , Protein Binding/genetics , Signal Transduction/genetics , Sorting Nexins/deficiency , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
14.
PLoS One ; 8(3): e58511, 2013.
Article in English | MEDLINE | ID: mdl-23526992

ABSTRACT

Sorting nexin 5 (Snx5) has been posited to regulate the degradation of epidermal growth factor receptor and the retrograde trafficking of cation-independent mannose 6-phosphate receptor/insulin-like growth factor II receptor. Snx5 has also been suggested to interact with Mind bomb-1, an E3 ubiquitin ligase that regulates the activation of Notch signaling. However, the in vivo functions of Snx5 are largely unknown. Here, we report that disruption of the Snx5 gene in mice (Snx5(-/-) mice) resulted in partial perinatal lethality; 40% of Snx5(-/-) mice died shortly after birth due to cyanosis, reduced air space in the lungs, and respiratory failure. Histological analysis revealed that Snx5(-/-) mice exhibited thickened alveolar walls associated with undifferentiated alveolar epithelial type I cells. In contrast, alveolar epithelial type II cells were intact, exhibiting normal surfactant synthesis and secretion. Although the expression levels of surfactant proteins and saturated phosphatidylcholine in the lungs of Snx5(-/-) mice were comparable to those of Snx5(+/+) mice, the expression levels of T1α, Aqp5, and Rage, markers for distal alveolar epithelial type I cells, were significantly decreased in Snx5 (-/-) mice. These results demonstrate that Snx5 is necessary for the differentiation of alveolar epithelial type I cells, which may underlie the adaptation to air breathing at birth.


Subject(s)
Alveolar Epithelial Cells/physiology , Respiratory Insufficiency/etiology , Sorting Nexins/deficiency , Alveolar Epithelial Cells/classification , Alveolar Epithelial Cells/pathology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Pulmonary Alveoli/abnormalities , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Respiratory Insufficiency/pathology , Respiratory Insufficiency/physiopathology , Sorting Nexins/genetics , Sorting Nexins/physiology
15.
Mol Cell Biol ; 31(8): 1734-47, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300787

ABSTRACT

Phox (PX) domain-containing sorting nexins (SNXs) are emerging as important regulators of endocytic trafficking. Sorting nexin 27 (SNX27) is unique, as it contains a PDZ (Psd-95/Dlg/ZO1) domain. We show here that SNX27 is primarily targeted to the early endosome by interaction of its PX domain with PtdIns(3)P. Although targeted ablation of the SNX27 gene in mice did not significantly affect growth and survival during embryonic development, SNX27 plays an essential role in postnatal growth and survival. N-Methyl-d-aspartate (NMDA) receptor 2C (NR2C) was identified as a novel SNX27-interacting protein, and this interaction is mediated by the PDZ domain of SNX27 and the C-terminal PDZ-binding motif of NR2C. Increased NR2C expression levels, together with impaired NR2C endocytosis in SNX27(-/-) neurons, indicate that SNX27 may function to regulate endocytosis and/or endosomal sorting of NR2C. This is consistent with a role of SNX27 as a general regulator for sorting of membrane proteins containing a PDZ-binding motif, and its absence may alter the trafficking of these proteins, leading to growth and survival defects.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Sorting Nexins/metabolism , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Evolution, Molecular , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Sequence Alignment , Sorting Nexins/deficiency , Sorting Nexins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...