Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell ; 147(5): 1118-31, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22118466

ABSTRACT

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


Subject(s)
Endocytosis , Monomeric Clathrin Assembly Proteins/metabolism , SNARE Proteins/chemistry , Animals , Cell Membrane/metabolism , Crystallography, X-Ray , HeLa Cells , Humans , Mice , Models, Molecular , R-SNARE Proteins/chemistry , R-SNARE Proteins/metabolism , Rats , SNARE Proteins/metabolism , Transport Vesicles/metabolism
2.
Cell ; 141(7): 1220-9, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20603002

ABSTRACT

The AP2 adaptor complex (alpha, beta2, sigma2, and mu2 subunits) crosslinks the endocytic clathrin scaffold to PtdIns4,5P(2)-containing membranes and transmembrane protein cargo. In the "locked" cytosolic form, AP2's binding sites for the two endocytic motifs, YxxPhi on the C-terminal domain of mu2 (C-mu2) and [ED]xxxL[LI] on sigma2, are blocked by parts of beta2. Using protein crystallography, we show that AP2 undergoes a large conformational change in which C-mu2 relocates to an orthogonal face of the complex, simultaneously unblocking both cargo-binding sites; the previously unstructured mu2 linker becomes helical and binds back onto the complex. This structural rearrangement results in AP2's four PtdIns4,5P(2)- and two endocytic motif-binding sites becoming coplanar, facilitating their simultaneous interaction with PtdIns4,5P(2)/cargo-containing membranes. Using a range of biophysical techniques, we show that the endocytic cargo binding of AP2 is driven by its interaction with PtdIns4,5P(2)-containing membranes.


Subject(s)
Adaptor Protein Complex 2/chemistry , Binding Sites , Cell Membrane/chemistry , Ligands , Models, Molecular , Phosphatidylinositols/chemistry , Protein Conformation
3.
Traffic ; 22(1-2): 6-22, 2021 01.
Article in English | MEDLINE | ID: mdl-33225555

ABSTRACT

In eukaryotic cells, clathrin-mediated endocytosis (CME) is a central pathway for the internalization of proteins from the cell surface, thereby contributing to the maintenance of the plasma membrane protein composition. A key component for the formation of endocytic clathrin-coated vesicles (CCVs) is AP-2, as it sequesters cargo membrane proteins, recruits a multitude of other endocytic factors and initiates clathrin polymerization. Here, we inhibited CME by depletion of AP-2 and explored the consequences for the plasma membrane proteome. Quantitative analysis revealed accumulation of major constituents of the endosomal-lysosomal system reflecting a block in retrieval by compensatory CME. The noticeable enrichment of integrins and blockage of their turnover resulted in severely impaired cell migration. Rare proteins such as the anti-cancer drug target CA9 and tumor markers (CD73, CD164, CD302) were significantly enriched. The AP-2 knockdown attenuated the global endocytic capacity, but clathrin-independent entry pathways were still operating, as indicated by persistent internalization of specific membrane-spanning and GPI-anchored receptors (PVR, IGF1R, CD55, TNAP). We hypothesize that blocking AP-2 function and thus inhibiting CME may be a novel approach to identify new druggable targets, or to increase their residence time at the plasma membrane, thereby increasing the probability for efficient therapeutic intervention.


Subject(s)
Endocytosis , Proteome , Cell Membrane , Clathrin , Clathrin-Coated Vesicles
4.
Clin Genet ; 100(4): 486-488, 2021 10.
Article in English | MEDLINE | ID: mdl-34270086

ABSTRACT

Jawad syndrome is a multiple congenital anomaly and intellectual disability syndrome with mutation in RBBP8 reported only in two families. Here, we report on two new families from Pakistan and identified a previously reported variant in RBBP8, NM_002894.3:c.1808-1809delTA. We could show that this mutation impairs splicing resulting in two different abnormal transcripts. Finally, we could verify a shared haplotype among all four families and estimate the founder event to have occurred some 24 generations ago.


Subject(s)
Endodeoxyribonucleases/genetics , Fingers/abnormalities , Founder Effect , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Microcephaly/diagnosis , Microcephaly/genetics , Mutation , RNA Splicing , Toes/abnormalities , Facies , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Pakistan , Pedigree , Phenotype , Sequence Analysis, DNA , Exome Sequencing
5.
PLoS Genet ; 11(4): e1005149, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25875445

ABSTRACT

Mutations in SPAST, encoding spastin, are the most common cause of autosomal dominant hereditary spastic paraplegia (HSP). HSP is characterized by weakness and spasticity of the lower limbs, owing to progressive retrograde degeneration of the long corticospinal axons. Spastin is a conserved microtubule (MT)-severing protein, involved in processes requiring rearrangement of the cytoskeleton in concert to membrane remodeling, such as neurite branching, axonal growth, midbody abscission, and endosome tubulation. Two isoforms of spastin are synthesized from alternative initiation codons (M1 and M87). We now show that spastin-M1 can sort from the endoplasmic reticulum (ER) to pre- and mature lipid droplets (LDs). A hydrophobic motif comprised of amino acids 57 through 86 of spastin was sufficient to direct a reporter protein to LDs, while mutation of arginine 65 to glycine abolished LD targeting. Increased levels of spastin-M1 expression reduced the number but increased the size of LDs. Expression of a mutant unable to bind and sever MTs caused clustering of LDs. Consistent with these findings, ubiquitous overexpression of Dspastin in Drosophila led to bigger and less numerous LDs in the fat bodies and increased triacylglycerol levels. In contrast, Dspastin overexpression increased LD number when expressed specifically in skeletal muscles or nerves. Downregulation of Dspastin and expression of a dominant-negative variant decreased LD number in Drosophila nerves, skeletal muscle and fat bodies, and reduced triacylglycerol levels in the larvae. Moreover, we found reduced amount of fat stores in intestinal cells of worms in which the spas-1 homologue was either depleted by RNA interference or deleted. Taken together, our data uncovers an evolutionarily conserved role of spastin as a positive regulator of LD metabolism and open up the possibility that dysfunction of LDs in axons may contribute to the pathogenesis of HSP.


Subject(s)
Adenosine Triphosphatases/metabolism , Drosophila Proteins/metabolism , Lipid Droplets/metabolism , Lipid Metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Amino Acid Motifs , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Endoplasmic Reticulum/metabolism , Intestinal Mucosa/metabolism , Muscle, Skeletal/metabolism , Neurons/metabolism , Protein Transport , Triglycerides/metabolism
6.
J Biol Chem ; 291(13): 6796-812, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26841862

ABSTRACT

The E3 transcription unit of human species C adenoviruses (Ads) encodes immunomodulatory proteins that mediate direct protection of infected cells. Recently, we described a novel immunomodulatory function for E3/49K, an E3 protein uniquely expressed by species D Ads. E3/49K of Ad19a/Ad64, a serotype that causes epidemic keratokonjunctivitis, is synthesized as a highly glycosylated type I transmembrane protein that is subsequently cleaved, resulting in secretion of its large ectodomain (sec49K). sec49K binds to CD45 on leukocytes, impairing activation and functions of natural killer cells and T cells. E3/49K is localized in the Golgi/trans-Golgi network (TGN), in the early endosomes, and on the plasma membrane, yet the cellular compartment where E3/49K is cleaved and the protease involved remained elusive. Here we show that TGN-localized E3/49K comprises both newly synthesized and recycled molecules. Full-length E3/49K was not detected in late endosomes/lysosomes, but the C-terminal fragment accumulated in this compartment at late times of infection. Inhibitor studies showed that cleavage occurs in a post-TGN compartment and that lysosomotropic agents enhance secretion. Interestingly, the cytoplasmic tail of E3/49K contains two potential sorting motifs, YXXΦ (where Φ represents a bulky hydrophobic amino acid) and LL, that are important for binding the clathrin adaptor proteins AP-1 and AP-2in vitro Surprisingly, mutating the LL motif, either alone or together with YXXΦ, did not prevent proteolytic processing but increased cell surface expression and secretion. Upon brefeldin A treatment, cell surface expression was rapidly lost, even for mutants lacking all known endocytosis motifs. Together with immunofluorescence data, we propose a model for intracellular E3/49K transport whereby cleavage takes place on the cell surface by matrix metalloproteases.


Subject(s)
Adenoviridae/immunology , Adenovirus E3 Proteins/chemistry , Cell Membrane/immunology , Epithelial Cells/immunology , Fibroblasts/immunology , Adenoviridae/chemistry , Adenoviridae/pathogenicity , Adenovirus E3 Proteins/genetics , Adenovirus E3 Proteins/immunology , Amino Acid Motifs , Brefeldin A/pharmacology , Cell Line, Tumor , Cell Membrane/virology , Endosomes/immunology , Endosomes/virology , Epithelial Cells/drug effects , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Immunomodulation , Jurkat Cells , Lysosomes/immunology , Lysosomes/virology , Molecular Sequence Data , Primary Cell Culture , Protein Structure, Tertiary , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction , Transfection , trans-Golgi Network/immunology , trans-Golgi Network/virology
7.
J Immunol ; 193(7): 3257-61, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25187660

ABSTRACT

Sensing of nucleic acids by TLRs is crucial in the host defense against viruses and bacteria. Unc-93 homolog B1 (UNC93B1) regulates the trafficking of nucleic acid-sensing TLRs from the endoplasmic reticulum to endolysosomes, where the TLRs encounter their respective ligands and become activated. In this article, we show that a carboxyl-terminal tyrosine-based sorting motif (YxxΦ) in UNC93B1 differentially regulates human nucleic acid-sensing TLRs in a receptor- and ligand-specific manner. Destruction of YxxΦ abolished TLR7, TLR8, and TLR9 activity toward nucleic acids in human B cells and monocytes, whereas TLR8 responses toward small molecules remained intact. YxxΦ in UNC93B1 influenced the subcellular localization of human UNC93B1 via both adapter protein complex (AP)1- and AP2-dependent trafficking pathways. However, loss of AP function was not causal for altered TLR responses, suggesting AP-independent functions of YxxΦ in UNC93B1.


Subject(s)
Adaptor Protein Complex 1/immunology , Adaptor Protein Complex 2/immunology , B-Lymphocytes/immunology , Membrane Transport Proteins/immunology , Monocytes/immunology , Toll-Like Receptors/immunology , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 2/genetics , Amino Acid Motifs , B-Lymphocytes/cytology , Cell Line, Tumor , HEK293 Cells , Humans , Membrane Transport Proteins/genetics , Monocytes/cytology , Protein Transport/genetics , Protein Transport/immunology , Toll-Like Receptors/genetics
8.
Proc Natl Acad Sci U S A ; 110(47): E4482-91, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24194549

ABSTRACT

Mutations in either syntaxin 11 (Stx11) or Munc18-2 abolish cytotoxic T lymphocytes (CTL) and natural killer cell (NK) cytotoxicity, and give rise to familial hemophagocytic lymphohistiocytosis (FHL4 or FHL5, respectively). Although Munc18-2 is known to interact with Stx11, little is known about the molecular mechanisms governing the specificity of this interaction or how in vitro IL-2 activation leads to compensation of CTL and NK cytotoxicity. To understand how mutations in Munc18-2 give rise to disease, we have solved the structure of human Munc18-2 at 2.6 Å resolution and mapped 18 point mutations. The four surface mutations identified (R39P, L130S, E132A, P334L) map exclusively to the predicted syntaxin and soluble N-ethylmaleimide-sensitive factor accessory protein receptor binding sites of Munc18-2. We find that Munc18-2 binds the N-terminal peptide of Stx11 with a ~20-fold higher affinity than Stx3, suggesting a potential role in selective binding. Upon IL-2 activation, levels of Stx3 are increased, favoring Munc18-2 binding when Stx11 is absent. Similarly, Munc18-1, expressed in IL-2-activated CTL, is capable of binding Stx11. These findings provide potential explanations for restoration of Munc18-Stx function and cytotoxicity in IL-2-activated cells.


Subject(s)
Evolution, Molecular , Killer Cells, Natural/immunology , Lymphohistiocytosis, Hemophagocytic/genetics , Models, Molecular , Munc18 Proteins/chemistry , Qa-SNARE Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Blotting, Western , Crystallization , HEK293 Cells , Humans , Immunohistochemistry , Killer Cells, Natural/metabolism , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Point Mutation/genetics , Protein Binding , Sf9 Cells , Spodoptera , T-Lymphocytes, Cytotoxic/metabolism
9.
Sci Adv ; 8(17): eabn2018, 2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35486718

ABSTRACT

Clathrin-mediated endocytosis (CME) is the main mechanism by which mammalian cells control their cell surface proteome. Proper operation of the pivotal CME cargo adaptor AP2 requires membrane-localized Fer/Cip4 homology domain-only proteins (FCHO). Here, live-cell enhanced total internal reflection fluorescence-structured illumination microscopy shows that FCHO marks sites of clathrin-coated pit (CCP) initiation, which mature into uniform-sized CCPs comprising a central patch of AP2 and clathrin corralled by an FCHO/Epidermal growth factor potential receptor substrate number 15 (Eps15) ring. We dissect the network of interactions between the FCHO interdomain linker and AP2, which concentrates, orients, tethers, and partially destabilizes closed AP2 at the plasma membrane. AP2's subsequent membrane deposition drives its opening, which triggers FCHO displacement through steric competition with phosphatidylinositol 4,5-bisphosphate, clathrin, cargo, and CME accessory factors. FCHO can now relocate toward a CCP's outer edge to engage and activate further AP2s to drive CCP growth/maturation.

10.
HGG Adv ; 3(3): 100111, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35571680

ABSTRACT

CSNK2B encodes for casein kinase II subunit beta (CK2ß), the regulatory subunit of casein kinase II (CK2), which is known to mediate diverse cellular pathways. Variants in this gene have been recently identified as a cause of Poirier-Bienvenu neurodevelopmental syndrome (POBINDS), but functional evidence is sparse. Here, we report five unrelated individuals: two of them manifesting POBINDS, while three are identified to segregate a new intellectual disability-craniodigital syndrome (IDCS), distinct from POBINDS. The three IDCS individuals carried two different de novo missense variants affecting the same codon of CSNK2B. Both variants, NP_001311.3; p.Asp32His and NP_001311.3; p.Asp32Asn, lead to an upregulation of CSNK2B expression at transcript and protein level, along with global dysregulation of canonical Wnt signaling. We found impaired interaction of the two key players DVL3 and ß-catenin with mutated CK2ß. The variants compromise the kinase activity of CK2 as evident by a marked reduction of phosphorylated ß-catenin and consequent absence of active ß-catenin inside nuclei of the patient-derived lymphoblastoid cell lines (LCLs). In line with these findings, whole-transcriptome profiling of patient-derived LCLs harboring the NP_001311.3; p.Asp32His variant confirmed a marked difference in expression of genes involved in the Wnt signaling pathway. In addition, whole-phosphoproteome analysis of the LCLs of the same subject showed absence of phosphorylation for 313 putative CK2 substrates, enriched in the regulation of nuclear ß-catenin and transcription of the target genes. Our findings suggest that discrete variants in CSNK2B cause dominant-negative perturbation of the canonical Wnt signaling pathway, leading to a new craniodigital syndrome distinguishable from POBINDS.

11.
Nat Cell Biol ; 4(2): 154-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11802162

ABSTRACT

Adaptors are heterotetrameric complexes that mediate the incorporation of cargo into transport vesicles by interacting with sorting signals present in the cytosolic domain of transmembrane proteins. Four adaptors, AP-1 (beta 1, gamma, mu 1A or mu 1B, sigma 1), AP-2 (beta 2, alpha, mu 2, sigma 2), AP-3 (beta 3 , delta, mu 3, sigma 3) or AP-4 (beta 4, epsilon, mu 4, sigma 4), have been characterized. AP-1 and AP-3 mediate sorting events at the level of the TGN and/or endosomes, whereas AP-2 functions in endocytic clathrin coated vesicle formation; no function is known so far for AP-4. Here, we show that AP-4 can bind different types of cytosolic signals known to mediate basolateral transport in epithelial cells. Furthermore, in MDCK cells with depleted mu 4 protein levels, several basolateral proteins are mis-sorted to the apical surface, showing that AP-4 participates in basolateral sorting in epithelial cells.


Subject(s)
Carrier Proteins/metabolism , Epithelial Cells/metabolism , Membrane Proteins/metabolism , Monomeric Clathrin Assembly Proteins , Protein Transport/physiology , Adaptor Proteins, Vesicular Transport , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Fractionation , Cell Line , Cell Polarity , Dogs , Epithelial Cells/ultrastructure , Humans , Kidney/cytology , Membrane Proteins/genetics , Molecular Sequence Data , Protein Binding , Protein Subunits , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
12.
FASEB J ; 24(11): 4443-58, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20624928

ABSTRACT

Hypoxia-inducible protein 2 (HIG2) has been implicated in canonical Wnt signaling, both as target and activator. The potential link between hypoxia and an oncogenic signaling pathway might play a pivotal role in renal clear-cell carcinoma characterized by constitutive activation of hypoxia-inducible factors (HIFs), and hence prompted us to analyze HIG2 regulation and function in detail. HIG2 was up-regulated by hypoxia and HIF inducers in all cell types and mouse organs investigated and abundantly expressed in renal clear-cell carcinomas. Promoter analyses, gel shifts, and siRNA studies revealed that HIG2 is a direct and specific target of HIF-1, but not responsive to HIF-2. Surprisingly, HIG2 was not secreted, and HIG2 overexpression neither stimulated proliferation nor activated Wnt signaling. Instead, we show that HIG2 decorates the hemimembrane of lipid droplets, whose number and size increase on hypoxic inhibition of fatty acid ß-oxidation, and colocalizes with the lipid droplet proteins adipophilin and TIP47. Normoxic overexpression of HIG2 was sufficient to increase neutral lipid deposition in HeLa cells and stimulated cytokine expression. HIG2 could be detected in atherosclerotic arteries and fatty liver disease, suggesting that this ubiquitously inducible HIF-1 target gene may play an important functional role in diseases associated with pathological lipid accumulation.


Subject(s)
Carcinoma, Renal Cell/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Animals , Carcinoma, Renal Cell/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Kidney Neoplasms/pathology , Lipid Metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Signal Transduction , Transcriptional Activation/genetics , Wnt1 Protein/metabolism
13.
Genes (Basel) ; 12(5)2021 05 13.
Article in English | MEDLINE | ID: mdl-34068194

ABSTRACT

Congenital microcephaly is the clinical presentation of significantly reduced head circumference at birth. It manifests as both non-syndromic-microcephaly primary hereditary (MCPH)-and syndromic forms and shows considerable inter- and intrafamilial variability. It has been hypothesized that additional genetic variants may be responsible for this variability, but data are sparse. We have conducted deep phenotyping and genotyping of five Pakistani multiplex families with either MCPH (n = 3) or Seckel syndrome (n = 2). In addition to homozygous causal variants in ASPM or CENPJ, we discovered additional heterozygous modifier variants in WDR62, CEP63, RAD50 and PCNT-genes already known to be associated with neurological disorders. MCPH patients carrying an additional heterozygous modifier variant showed more severe phenotypic features. Likewise, the phenotype of Seckel syndrome caused by a novel CENPJ variant was aggravated to microcephalic osteodysplastic primordial dwarfism type II (MOPDII) in conjunction with an additional PCNT variant. We show that the CENPJ missense variant impairs splicing and decreases protein expression. We also observed centrosome amplification errors in patient cells, which were twofold higher in MOPDII as compared to Seckel cells. Taken together, these observations advocate for consideration of additional variants in related genes for their role in modifying the expressivity of the phenotype and need to be considered in genetic counseling and risk assessment.


Subject(s)
Genes, Modifier , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Acid Anhydride Hydrolases/genetics , Adult , Antigens/genetics , Cell Cycle Proteins/genetics , Child , DNA-Binding Proteins/genetics , Female , Heterozygote , Humans , Male , Microcephaly/pathology , Microtubule-Associated Proteins/metabolism , Mutation , Pedigree , Phenotype
14.
Traffic ; 9(7): 1157-72, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18410487

ABSTRACT

The adaptor complexes AP-1 and AP-3 are localized to endosomes and/or the trans Golgi network (TGN). Because of limitations in analysing intracellular adaptor function directly, their site of function is a matter of ongoing uncertainty. To overcome this problem and to analyse adaptor sorting at the TGN, we reconstituted vesicle formation from Golgi/TGN-enriched membranes in a novel in vitro budding assay. Melanocytes were metabolically labelled followed by a 19 degrees C temperature block to accumulate newly synthesized proteins in Golgi membranes, which were then enriched by subcellular fractionation and used as donor membranes for vesicle formation in vitro. The incorporation of the melanosomal proteins tyrosinase and tyrosinase-related protein 1 (TRP-1) as well as Lamp-1 and 46 kDa mannose-6-phosphate receptor (MPR46) into Golgi/TGN-derived vesicles was temperature, nucleotide, cytosol, ADP ribosylation factor 1 and adaptor dependent. We show that sorting of TRP-1 and MPR46 was AP-1 dependent, while budding of tyrosinase and Lamp-1 required AP-3. Depletion of clathrin inhibited sorting of all four cargo proteins, suggesting that AP-1 and AP-3 are involved in the formation of distinct types of clathrin-coated vesicles, each of which is characterized by the incorporation of specific cargo membrane proteins.


Subject(s)
Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex 3/metabolism , Gene Expression Regulation , Golgi Apparatus/metabolism , Lysosomes/metabolism , Animals , Cell Membrane/metabolism , Clathrin/metabolism , Fibroblasts/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , Models, Biological , Subcellular Fractions/metabolism , Swine
15.
J Biol Chem ; 284(40): 27646-54, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19651769

ABSTRACT

Clathrin-coated vesicles (CCVs) originating from the trans-Golgi network (TGN) provide a major transport pathway from the secretory system to endosomes/lysosomes. Herein we describe paralogous Sec14 domain-bearing proteins, clavesin 1/CRALBPL and clavesin 2, identified through a proteomic analysis of CCVs. Clavesins are enriched on CCVs and form a complex with clathrin heavy chain (CHC) and adaptor protein-1, major coat components of TGN-derived CCVs. The proteins co-localize with markers of endosomes and the TGN as well as with CHC and adaptor protein-1. A membrane mimic assay using the Sec14 domain of clavesin 1 reveals phosphatidylinositol 3,5-bisphosphate as a specific lipid partner. Phosphatidylinositol 3,5-bisphosphate is localized to late endosomes/lysosomes, and interestingly, isoform-specific knockdown of clavesins in neurons using lentiviral delivery of interfering RNA leads to enlargement of a lysosome-associated membrane protein 1-positive membrane compartment with no obvious influence on the CCV machinery at the TGN. Since clavesins are expressed exclusively in neurons, this new protein family appears to provide a unique neuron-specific regulation of late endosome/lysosome morphology.


Subject(s)
Carrier Proteins/metabolism , Clathrin/metabolism , Lipid Metabolism , Lysosomes/metabolism , Neurons/cytology , Neurons/metabolism , Adaptor Protein Complex 1/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Membrane/metabolism , Endosomes/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , Molecular Sequence Data , Organ Specificity , Phosphatidylinositol Phosphates/metabolism , Protein Structure, Tertiary , Protein Transport , trans-Golgi Network/metabolism
16.
J Exp Med ; 198(8): 1133-46, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-14557411

ABSTRACT

Relatively little is known about the pathway leading to the presentation of glycolipids by CD1 molecules. Here we show that the adaptor protein complex 3 (AP-3) is required for the efficient presentation of glycolipid antigens that require internalization and processing. AP-3 interacts with mouse CD1d, and cells from mice deficient for AP-3 have increased cell surface levels of CD1d and decreased expression in late endosomes. Spleen cells from AP-3-deficient mice have a reduced ability to present glycolipids to natural killer T (NKT) cells. Furthermore, AP-3-deficient mice have a significantly reduced NKT cell population, although this is not caused by self-tolerance that might result from increased CD1d surface levels. These data suggest that the generation of the endogenous ligand that selects NKT cells may also be AP-3 dependent. However, the function of MHC class II-reactive CD4+ T lymphocytes is not altered by AP-3 deficiency. Consistent with this divergence from the class II pathway, NKT cell development and antigen presentation by CD1d are not reduced by invariant chain deficiency. These data demonstrate that the AP-3 requirement is a particular attribute of the CD1d pathway in mice and that, although MHC class II molecules and CD1d are both found in late endosomes or lysosomes, different pathways mediate their intracellular trafficking.


Subject(s)
Adaptor Protein Complex 3/immunology , Antigens, CD1/immunology , Glycosphingolipids/immunology , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigens, CD1/genetics , Antigens, CD1d , CD4-Positive T-Lymphocytes/immunology , Cell Count , Complementarity Determining Regions/chemistry , Flow Cytometry , Liver/immunology , Lysosomes/metabolism , Mice , Mice, Mutant Strains , Recombinant Fusion Proteins/immunology , T-Lymphocyte Subsets/chemistry , Thymus Gland/immunology
17.
Life Sci Alliance ; 3(6)2020 06.
Article in English | MEDLINE | ID: mdl-32321733

ABSTRACT

Lipid droplets (LDs) are metabolic organelles that store neutral lipids and dynamically respond to changes in energy availability by accumulating or mobilizing triacylglycerols (TAGs). How the plastic behavior of LDs is regulated is poorly understood. Hereditary spastic paraplegia is a central motor axonopathy predominantly caused by mutations in SPAST, encoding the microtubule-severing protein spastin. The spastin-M1 isoform localizes to nascent LDs in mammalian cells; however, the mechanistic significance of this targeting is not fully explained. Here, we show that tightly controlled levels of spastin-M1 are required to inhibit LD biogenesis and TAG accumulation. Spastin-M1 maintains the morphogenesis of the ER when TAG synthesis is prevented, independent from microtubule binding. Moreover, spastin plays a microtubule-dependent role in mediating the dispersion of LDs from the ER upon glucose starvation. Our results reveal a dual role of spastin to shape ER tubules and to regulate LD movement along microtubules, opening new perspectives for the pathogenesis of hereditary spastic paraplegia.


Subject(s)
Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Microtubules/metabolism , Signal Transduction/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastin/deficiency , Animals , Cell Line, Tumor , Fibroblasts/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Isoenzymes , Mice , Motor Neurons/metabolism , Mutation , Spastic Paraplegia, Hereditary/genetics , Spastin/genetics , Transfection , Triglycerides/metabolism
18.
Nat Microbiol ; 5(2): 354-367, 2020 02.
Article in English | MEDLINE | ID: mdl-31873204

ABSTRACT

The cytosolic appearance and propagation of bacteria cause overwhelming cellular stress responses that induce apoptosis under normal conditions. Therefore, successful bacterial colonization depends on the ability of intracellular pathogens to block apoptosis and to safeguard bacterial replicative niches. Here, we show that the cytosolic Gram-negative bacterium Shigella flexneri stalls apoptosis by inhibiting effector caspase activity. Our data identified lipopolysaccharide (LPS) as a bona fide effector caspase inhibitor that directly binds caspases by involving its O-antigen (O Ag) moiety. Bacterial strains that lacked the O Ag or failed to replicate within the cytosol were incapable of blocking apoptosis and exhibited reduced virulence in a murine model of bacterial infection. Our findings demonstrate how Shigella inhibits pro-apoptotic caspase activity, effectively delays coordinated host-cell demise and supports its intracellular propagation. Next to the recently discovered pro-inflammatory role of cytosolic LPS, our data reveal a distinct mode of LPS action that, through the disruption of the early coordinated non-lytic cell death response, ultimately supports the inflammatory breakdown of infected cells at later time points.


Subject(s)
Apoptosis/physiology , Caspase Inhibitors/metabolism , Caspases, Effector/metabolism , Gram-Negative Bacteria/pathogenicity , Lipopolysaccharides/metabolism , Shigella flexneri/pathogenicity , Animals , Cytosol/microbiology , Female , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/physiology , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , O Antigens/metabolism , Shigella flexneri/genetics , Shigella flexneri/physiology , Virulence
19.
Mol Genet Genomic Med ; 8(9): e1408, 2020 09.
Article in English | MEDLINE | ID: mdl-32677750

ABSTRACT

BACKGROUND: Primary microcephaly (MCPH) is a congenital neurodevelopmental disorder manifesting as small brain and intellectual disability. It underlies isolated reduction of the cerebral cortex that is reminiscent of early hominids which makes it suitable model disease to study the hominin-specific volumetric expansion of brain. Mutations in 25 genes have been reported to cause this disorder. Although majority of these genes were discovered in the Pakistani population, still a significant proportion of these families remains uninvestigated. METHODS: We studied a cohort of 32 MCPH families from different regions of Pakistan. For disease gene identification, genome-wide linkage analysis, Sanger sequencing, gene panel, and whole-exome sequencing were performed. RESULTS: By employing these techniques individually or in combination, we were able to discern relevant disease-causing DNA variants. Collectively, 15 novel mutations were observed in five different MCPH genes; ASPM (10), WDR62 (1), CDK5RAP2 (1), STIL (2), and CEP135 (1). In addition, 16 known mutations were also verified. We reviewed the literature and documented the published mutations in six MCPH genes. Intriguingly, our cohort also revealed a recurrent mutation, c.7782_7783delGA;p.(Lys2595Serfs*6), of ASPM reported worldwide. Drawing from this collective data, we propose two founder mutations, ASPM:c.9557C>G;p.(Ser3186*) and CENPJ:c.18delC;p.(Ser7Profs*2), in the Pakistani population. CONCLUSIONS: We discovered novel DNA variants, impairing the function of genes indispensable to build a proper functioning brain. Our study expands the mutational spectra of known MCPH genes and also provides supporting evidence to the pathogenicity of previously reported mutations. These novel DNA variants will be helpful for the clinicians and geneticists for establishing reliable diagnostic strategies for MCPH families.


Subject(s)
Genetic Loci , Microcephaly/genetics , Mutation , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Consanguinity , Female , Founder Effect , Gene Frequency , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Microcephaly/pathology , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Pedigree
20.
Hum Mutat ; 30(4): 641-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19177549

ABSTRACT

We extend the spectrum of phenotypes caused by mutations in the Wnt/Norrin coreceptor low-density lipoprotein receptor-related protein 5 (LRP5) by identifying two novel types of mutation in related individuals whose presenting features were profound muscle hypotonia, mild mental retardation, blindness, and growth retardation. One mutation removes 6 out of 9 consecutive leucine residues in the LRP5 signal peptide (c.43_60del or p.Leu15_Leu20del), which impairs polypeptide entry into the endoplasmic reticulum (ER), trafficking to the cell membrane, and signal transduction. The second mutation resulted from nonhomologous recombination between Alu repeat sequences, which deleted exons 14-16 and would produce a nonfunctional, truncated, and frameshifted polypeptide, if expressed [chr11:g.(13871447_1387511)_(13879636_13879700)del (NW_925106.1) or p.Pro1010GlnfsX38]. We confirmed that the length of the LRP5 signal peptide poly-leucine repeat is polymorphic in the general population, and, importantly, we were able to demonstrate in independent in vitro assays that different allele sizes affect receptor processing and signal transduction. Consequently, this polymorphism may have physiologic effects in vivo. This latter finding is relevant since through a genomewide search we identified nearly 400 human proteins that contain poly-leucine repeats within their signal peptide. We chose 18 of these proteins and genotyped the underlying trinucleotide repeat in healthy Caucasian individuals. More than one length allele was observed in one-half of the proteins. We therefore propose that natural variation in poly-leucine-stretches within signal peptides constitutes a currently unrecognized source of variability in protein translation and expression.


Subject(s)
Abnormalities, Multiple/genetics , LDL-Receptor Related Proteins/genetics , Mutation , Osteoporosis/pathology , Protein Sorting Signals/genetics , Trinucleotide Repeats/genetics , Abnormalities, Multiple/pathology , Amino Acid Sequence , Base Sequence , Blotting, Western , Cell Line , Eye Diseases/pathology , Family Health , Female , Gene Frequency , Genotype , Humans , Leucine/genetics , Low Density Lipoprotein Receptor-Related Protein-5 , Luciferases/genetics , Luciferases/metabolism , Male , Pedigree , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Syndrome , Turkey
SELECTION OF CITATIONS
SEARCH DETAIL