Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Plant Cell ; 35(3): 975-993, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36660928

ABSTRACT

Elucidating enzyme-substrate relationships in posttranslational modification (PTM) networks is crucial for understanding signal transduction pathways but is technically difficult because enzyme-substrate interactions tend to be transient. Here, we demonstrate that TurboID-based proximity labeling (TbPL) effectively and specifically captures the substrates of kinases and phosphatases. TbPL-mass spectrometry (TbPL-MS) identified over 400 proximal proteins of Arabidopsis thaliana BRASSINOSTEROID-INSENSITIVE2 (BIN2), a member of the GLYCOGEN SYNTHASE KINASE 3 (GSK3) family that integrates signaling pathways controlling diverse developmental and acclimation processes. A large portion of the BIN2-proximal proteins showed BIN2-dependent phosphorylation in vivo or in vitro, suggesting that these are BIN2 substrates. Protein-protein interaction network analysis showed that the BIN2-proximal proteins include interactors of BIN2 substrates, revealing a high level of interactions among the BIN2-proximal proteins. Our proteomic analysis establishes the BIN2 signaling network and uncovers BIN2 functions in regulating key cellular processes such as transcription, RNA processing, translation initiation, vesicle trafficking, and cytoskeleton organization. We further discovered significant overlap between the GSK3 phosphorylome and the O-GlcNAcylome, suggesting an evolutionarily ancient relationship between GSK3 and the nutrient-sensing O-glycosylation pathway. Our work presents a powerful method for mapping PTM networks, a large dataset of GSK3 kinase substrates, and important insights into the signaling network that controls key cellular functions underlying plant growth and acclimation.


Subject(s)
Protein Kinases , Proteomics , Signal Transduction , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Biotin/chemistry , Biotinylation , Brassinosteroids/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Proteomics/methods , Signal Transduction/physiology
2.
Mol Cell Proteomics ; 19(6): 1005-1016, 2020 06.
Article in English | MEDLINE | ID: mdl-32284353

ABSTRACT

Posttranslational modifications play a critical and diverse role in regulating cellular activities. Despite their fundamentally important role in cellular function, there has been no report to date of an effective generalized approach to the targeting, extraction, and characterization of the critical c-terminal regions of natively prenylated proteins. Various chemical modification and metabolic labeling strategies in cell culture have been reported. However, their applicability is limited to cell culture systems and does not allow for analysis of tissue samples. The chemical characteristics (hydrophobicity, low abundance, highly basic charge) of many of the c-terminal regions of prenylated proteins have impaired the use of standard proteomic workflows. In this context, we sought a direct approach to the problem in order to examine these proteins in tissue without the use of labeling. Here we demonstrate that prenylated proteins can be captured on chromatographic resins functionalized with mixed disulfide functions. Protease treatment of resin-bound proteins using chymotryptic digestion revealed peptides from many known prenylated proteins. Exposure of the protease-treated resin to reducing agents and hydro organic mixtures released c-terminal peptides with intact prenyl groups along with other enzymatic modifications expected in this protein family. Database and search parameters were selected to allow for c-terminal modifications unique to these molecules such as CAAX box processing and c-terminal methylation. In summary, we present a direct approach to enrich and obtain information at a molecular level of detail about prenylation of proteins from tissue and cell extracts using high-performance LC-MS without the need for metabolic labeling and derivatization.


Subject(s)
Chromatography, Liquid/methods , Peptides/analysis , Proteins/analysis , Proteomics/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Brain/metabolism , Databases, Protein , Mice , Peptide Hydrolases/chemistry , Peptides/chemistry , Protein Prenylation , Proteins/chemistry , Sepharose/analogs & derivatives , Sepharose/chemistry
3.
Proc Natl Acad Sci U S A ; 113(11): E1536-44, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26903630

ABSTRACT

In humans, the connection between sleep and mood has long been recognized, although direct molecular evidence is lacking. We identified two rare variants in the circadian clock gene PERIOD3 (PER3-P415A/H417R) in humans with familial advanced sleep phase accompanied by higher Beck Depression Inventory and seasonality scores. hPER3-P415A/H417R transgenic mice showed an altered circadian period under constant light and exhibited phase shifts of the sleep-wake cycle in a short light period (photoperiod) paradigm. Molecular characterization revealed that the rare variants destabilized PER3 and failed to stabilize PERIOD1/2 proteins, which play critical roles in circadian timing. Although hPER3-P415A/H417R-Tg mice showed a mild depression-like phenotype, Per3 knockout mice demonstrated consistent depression-like behavior, particularly when studied under a short photoperiod, supporting a possible role for PER3 in mood regulation. These findings suggest that PER3 may be a nexus for sleep and mood regulation while fine-tuning these processes to adapt to seasonal changes.


Subject(s)
Affect/physiology , Period Circadian Proteins/genetics , Seasonal Affective Disorder/genetics , Aged , Amino Acid Sequence , Animals , Circadian Clocks/genetics , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Period Circadian Proteins/metabolism , Photoperiod , Protein Stability , Sleep Disorders, Circadian Rhythm/genetics
4.
Mol Cell Proteomics ; 14(8): 2103-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25995273

ABSTRACT

This manuscript describes the enrichment and mass spectrometric analysis of intact glycopeptides from mouse liver, which yielded site-specific N- and O-glycosylation data for ∼ 130 proteins. Incorporation of different sialic acid variants in both N- and O-linked glycans was observed, and the importance of using both collisional activation and electron transfer dissociation for glycopeptide analysis was illustrated. The N-glycan structures of predicted lysosomal, endoplasmic reticulum (ER), secreted and transmembrane proteins were compared. The data suggest that protein N-glycosylation differs depending on cellular location. The glycosylation patterns of several mouse liver and mouse brain glycopeptides were compared. Tissue-specific differences in glycosylation were observed between sites within the same protein: Some sites displayed a similar spectrum of glycan structures in both tissues, whereas for others no overlap was observed. We present comparative brain/liver glycosylation data on 50 N-glycosylation sites from 34 proteins and 13 O-glycosylation sites from seven proteins.


Subject(s)
Glycopeptides/metabolism , Organ Specificity , Animals , Brain/metabolism , Endoplasmic Reticulum/metabolism , Glycopeptides/chemistry , Glycosylation , Liver/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice
5.
Anal Chem ; 87(5): 3064-71, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25654559

ABSTRACT

Beam-type collision-induced dissociation (CID) data of intact glycopeptides isolated from mouse liver tissue are presented to illustrate characteristic fragmentation of differentially sialylated glycopeptides. Eight glycoforms of an O-linked glycopeptide from Nucleobindin-1 are distinguished on the basis of the precursor masses and characteristic oxonium ions. We report that all sialic acid variants are prone to neutral loss from the charge reduced species in electron-transfer dissociation (ETD) fragmentation. We show how changes in sialic acid composition affect reverse phase chromatographic retention times: sialic acid addition increases glycopeptide retention times significantly; replacing the N-acetylneuraminic acid with the N-glycolyl variant leads to slightly reduced retention times, while O-acetylated sialic acid-containing glycoforms are retained longer. We then demonstrate how MS-Filter in Protein Prospector can use these diagnostic oxonium ions to find glycopeptides, by showing that a wealth of different glycopeptides can be found in a published phosphopeptide data set.


Subject(s)
Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/chemistry , Glycopeptides/analysis , Liver/metabolism , N-Acetylneuraminic Acid/chemistry , Nerve Tissue Proteins/chemistry , Protein Processing, Post-Translational , Tandem Mass Spectrometry/methods , Animals , Chromatography, Affinity , Chromatography, Liquid , Glycopeptides/isolation & purification , Glycosylation , Mice , Nucleobindins , Oxonic Acid/chemistry
6.
Cell Metab ; 17(2): 291-302, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395175

ABSTRACT

Posttranslational modifications play central roles in myriad biological pathways including circadian regulation. We employed a circadian proteomic approach to demonstrate that circadian timing of phosphorylation is a critical factor in regulating complex GSK3ß-dependent pathways and identified O-GlcNAc transferase (OGT) as a substrate of GSK3ß. Interestingly, OGT activity is regulated by GSK3ß; hence, OGT and GSK3ß exhibit reciprocal regulation. Modulating O-GlcNAcylation levels alter circadian period length in both mice and Drosophila; conversely, protein O-GlcNAcylation is circadianly regulated. Central clock proteins, Clock and Period, are reversibly modified by O-GlcNAcylation to regulate their transcriptional activities. In addition, O-GlcNAcylation of a region in PER2 known to regulate human sleep phase (S662-S674) competes with phosphorylation of this region, and this interplay is at least partly mediated by glucose levels. Together, these results indicate that O-GlcNAcylation serves as a metabolic sensor for clock regulation and works coordinately with phosphorylation to fine-tune circadian clock.


Subject(s)
Acetylglucosamine/metabolism , Circadian Clocks , Glucose/metabolism , Adenosine Triphosphate/analogs & derivatives , Amino Acid Sequence , Animals , CLOCK Proteins/chemistry , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Glycosylation , Humans , Mice , Molecular Sequence Data , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , Substrate Specificity , Transcription, Genetic , Transfection
7.
J Biol Chem ; 286(11): 9063-70, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21228270

ABSTRACT

Glucocorticoids rapidly and robustly induce cell fate decisions in various multipotent cells, although the precise mechanisms of these important cellular events are not understood. Here we showed that glucocorticoids repressed Per3 expression and that this repression was critical for advancing mesenchymal stem cells to the adipocyte fate. Exogenous expression of Per3 inhibited adipogenesis, whereas knocking out Per3 enhanced that fate. Moreover, we found that PER3 formed a complex with PPARγ and inhibited PPARγ-mediated transcriptional activation via Pparγ response elements. Consistent with these findings, Per3 knock-out mice displayed alterations in body composition, with both increased adipose and decreased muscle tissue compared with wild-type mice. Our findings identify Per3 as potent mediator of cell fate that functions by altering the transcriptional activity of PPARγ.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , PPAR gamma/biosynthesis , Period Circadian Proteins/metabolism , Response Elements/physiology , 3T3-L1 Cells , Adipocytes/cytology , Animals , COS Cells , Chlorocebus aethiops , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Mice , PPAR gamma/genetics , Period Circadian Proteins/genetics
8.
Hered Cancer Clin Pract ; 8(1): 4, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20380699

ABSTRACT

BACKGROUND: The aim of this study was to identify BRCA1 and BRCA2 mutations in the Estonian population. We analyzed genetic data and questionnaire from 64 early-onset (< 45 y) breast cancer patients, 47 familial cases (patients with breast or ovarian cancer and a case of these cancers in the family), and 33 predictive cases (patients without breast or ovarian cancer, with a family history of such diseases) from Estonia for mutations in the BRCA1 gene. A sub-set of familial cases and predictive cases were also analyzed for mutations in the BRCA2 gene. METHODS: For mutation detection, we used the Polymerase Chain Reaction-Single Stranded Conformation Polymorphism Heteroduplex Analysis (PCR-SSCP-HD), followed by direct DNA sequencing. RESULTS: We identified three clinically important mutations in the BRCA1 gene, including seven occurrences of the c.5382insC mutation, three of c.4154delA, and one instance of c.3881_3882delGA. We also detected six polymorphisms: c.2430T>C, c.3232A>G, c.4158A>G, c.4427T>C, c.4956A>G, and c.5002T>C. Four sequence alterations were detected in introns: c.560+64delT, c.560+ [36-38delCTT, 52-63del12], c.666-58delT, and c.5396+60insGTATTCCACTCC. In the BRCA2 gene, two clinically important mutations were found: c.9610C>T and c.6631delTTAAATG. Additionally, two alterations (c.7049G>T and c.7069+80delTTAG) with unknown clinical significance were detected. CONCLUSIONS: In our dataset, the overall frequency of clinically important BRCA1 mutations in early-onset patients, familial cases, and predictive testing was 7.6% (144 cases, 11 mutation carriers). Pathogenic mutations were identified in 4 of the 64 early-onset breast cancer cases (6.3%). In familial cases, clinically important mutations in the BRCA1 gene were found in 6 of the 47 individuals analyzed (12.8%). In predictive cases, 1 clinically important mutation was detected in 33 individuals studied (3%). The occurrence of clinically important mutations in BRCA2 in familial cases of breast cancer was 2 of the 16 individuals analyzed (12.5%).

9.
Am J Hum Genet ; 83(1): 43-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18589395

ABSTRACT

Fragile X syndrome results from the absence of the fragile X mental retardation 1 (FMR1) gene product (FMRP). FMR1 has two paralogs in vertebrates: fragile X related gene 1 and 2 (FXR1 and FXR2). Here we show that Fmr1/Fxr2 double knockout (KO) and Fmr1 KO/Fxr2 heterozygous animals exhibit a loss of rhythmic activity in a light:dark (LD) cycle, and that Fmr1 or Fxr2 KO mice display a shorter free-running period of locomotor activity in total darkness (DD). Molecular analysis and in vitro electrophysiological studies suggest essentially normal function of cells in the suprachiasmatic nucleus (SCN) in Fmr1/Fxr2 double KO mice. However, the cyclical patterns of abundance of several core clock component messenger (m) RNAs are altered in the livers of double KO mice. Furthermore, FXR2P alone or FMRP and FXR2P together can increase PER1- or PER2-mediated BMAL1-Neuronal PAS2 (NPAS2) transcriptional activity in a dose-dependent manner. These data collectively demonstrate that FMR1 and FXR2 are required for the presence of rhythmic circadian behavior in mammals and suggest that this role may be relevant to sleep and other behavioral alterations observed in fragile X patients.


Subject(s)
Behavior, Animal , Circadian Rhythm/genetics , Gene Expression Regulation , RNA-Binding Proteins/genetics , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cryptochromes , Electrophysiology , Flavoproteins/metabolism , Fragile X Syndrome , Gene Expression Regulation/genetics , Heterozygote , In Situ Hybridization , Liver/chemistry , Male , Mammals/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Period Circadian Proteins , RNA, Messenger/metabolism , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Nature ; 439(7074): 340-3, 2006 Jan 19.
Article in English | MEDLINE | ID: mdl-16421573

ABSTRACT

Environmental light is the 'zeitgeber' (time-giver) of circadian behaviour. Constant darkness is considered a 'free-running' circadian state. Mammals encounter constant darkness during hibernation. Ablation of the master clock synchronizer, the suprachiasmatic nucleus, abolishes torpor, a hibernation-like state, implicating the circadian clock in this phenomenon. Here we report a mechanism by which constant darkness regulates the gene expression of fat catabolic enzymes in mice. Genes for murine procolipase (mClps) and pancreatic lipase-related protein 2 (mPlrp2) are activated in a circadian manner in peripheral organs during 12 h dark:12 h dark (DD) but not light-dark (LD) cycles. This mechanism is deregulated in circadian-deficient mPer1-/-/mPer2m/m mice. We identified circadian-regulated 5'-AMP, which is elevated in the blood of DD mice, as a key mediator of this response. Synthetic 5'-AMP induced torpor and mClps expression in LD animals. Torpor induced by metabolic stress was associated with elevated 5'-AMP levels in DD mice. Levels of glucose and non-esterified fatty acid in the blood are reversed in DD and LD mice. Induction of mClps expression by 5'-AMP in LD mice was reciprocally linked to blood glucose levels. Our findings uncover a circadian metabolic rhythm in mammals.


Subject(s)
Circadian Rhythm/physiology , Darkness , Gene Expression Regulation , Lipid Metabolism , Adenosine Monophosphate/blood , Animals , Blood Glucose/analysis , Cell Cycle Proteins , Circadian Rhythm/genetics , Colipases/genetics , Colipases/metabolism , Enzyme Precursors , Fatty Acids, Nonesterified/blood , Feeding Behavior , Female , Light , Lipase/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Period Circadian Proteins , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological/metabolism , Transcription Factors/genetics
11.
Nature ; 430(6998): 467-71, 2004 Jul 22.
Article in English | MEDLINE | ID: mdl-15269772

ABSTRACT

The circadian clock is the central timing system that controls numerous physiological processes. In mammals, one such process is haem biosynthesis, which the clock controls through regulation of the rate-limiting enzyme aminolevulinate synthase 1 (Alas1). Several members of the core clock mechanism are PAS domain proteins, one of which, neuronal PAS 2 (NPAS2), has a haem-binding motif. Indeed, haem controls activity of the BMAL1-NPAS2 transcription complex in vitro by inhibiting DNA binding in response to carbon monoxide. Here we show that haem differentially modulates expression of the mammalian Period genes mPer1 and mPer2 in vivo by a mechanism involving NPAS2 and mPER2. Further experiments show that mPER2 positively stimulates activity of the BMAL1-NPAS2 transcription complex and, in turn, NPAS2 transcriptionally regulates Alas1. Vitamin B12 and haem compete for binding to NPAS2 and mPER2, but they have opposite effects on mPer2 and mPer1 expression in vivo. Our data show that the circadian clock and haem biosynthesis are reciprocally regulated and suggest that porphyrin-containing molecules are potential targets for therapy of circadian disorders.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Gene Expression Regulation , Heme/biosynthesis , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors , Biological Clocks/drug effects , Biological Clocks/genetics , COS Cells , Cell Cycle Proteins , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Gene Expression Regulation/drug effects , Heme/metabolism , Heme/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Motor Activity/physiology , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Period Circadian Proteins , Protein Binding , Substrate Specificity , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Vitamin B 12/metabolism
12.
Hum Mol Genet ; 13(4): 367-78, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14709593

ABSTRACT

Contiguous gene syndromes (CGS) refer to a group of disorders associated with chromosomal rearrangements in which the phenotype is thought to result from altered copy number of physically linked dosage-sensitive genes. Smith-Magenis syndrome and [dup(17)(p11.2p11.2)] are CGS associated with a heterozygous deletion or duplication of band p11.2 of chromosome 17, respectively. We previously constructed animal models for these CGSs by engineering rearranged chromosomes carrying a deletion/deficiency [Df(11)17] (Del mutant) or a duplication [Dp(11)17 ] (Dup mutant) of the syntenic region on mouse chromosome 11. Here we present a behavioral analysis of these models indicating that heterozygous male mice carrying the engineered deletion or the duplication are hypoactive or hyperactive, respectively. In addition, male Dup mutant mice, but not Del mutant mice, have impaired contextual fear conditioning. Circadian rhythm studies revealed period length differences in Del mutant mice, but not Dup mutant mice. These results indicate that some of the behavioral abnormalities are gene dosage sensitive, whereas other behavioral abnormalities are specific to mice carrying the deletion or the duplication and can be observed in a sex preferential manner. Our findings suggest that there is a gene(s) present in this defined genomic interval that is responsible for behavioral abnormalities in the mouse, as has been shown for the human syntenic region.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 17/genetics , Gene Deletion , Gene Duplication , Mental Disorders/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Syndrome
13.
J Biol Chem ; 277(14): 12128-36, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11823458

ABSTRACT

Estrogen reduces the risk of Alzheimer's disease in post-menopausal women, beta-amyloid (Abeta) burden in animal models of Alzheimer's disease, and secretion of Abeta from neuronal cultures. The biological basis for these effects remains unknown. Here, utilizing cell-free systems derived from both neuroblastoma cells and primary neurons, we demonstrate that 17beta-estradiol (17beta-E2) stimulates formation of vesicles containing the beta-amyloid precursor protein (betaAPP) from the trans-Golgi network (TGN). Accelerated betaAPP trafficking precludes maximal Abeta generation within the TGN. 17beta-E2 appears to modulate TGN phospholipid levels, particularly those of phosphatidylinositol, and to recruit soluble trafficking factors, such as Rab11, to the TGN. Together, these results suggest that estrogen may exert its anti-Abeta effects by regulating betaAPP trafficking within the late secretory pathway. These results suggest a novel mechanism through which 17beta-E2 may act in estrogen-responsive tissues and illustrate how altering the kinetics of the transport of a protein can influence its metabolic fate.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Estrogens/pharmacology , Golgi Apparatus/metabolism , trans-Golgi Network/metabolism , Animals , Biotinylation , Cell Membrane/metabolism , Cell-Free System , Chromatography, Thin Layer , Cytosol/metabolism , Estrogens/metabolism , Humans , Lipid Metabolism , Mice , Mutation , Neuroblastoma/metabolism , Neurons/metabolism , Precipitin Tests , Protein Binding , Protein Transport , Rats , Signal Transduction , Sucrose/pharmacology , Temperature , Time Factors , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...