Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Elife ; 122023 08 21.
Article in English | MEDLINE | ID: mdl-37603466

ABSTRACT

Delta-like homolog 1 (Dlk1), an inhibitor of adipogenesis, controls the cell fate of adipocyte progenitors. Experimental data presented here identify two independent regulatory mechanisms, transcriptional and translational, by which Ifrd1 (TIS7) and its orthologue Ifrd2 (SKMc15) regulate Dlk1 levels. Mice deficient in both Ifrd1 and Ifrd2 (dKO) had severely reduced adipose tissue and were resistant to high-fat diet-induced obesity. Wnt signaling, a negative regulator of adipocyte differentiation, was significantly upregulated in dKO mice. Elevated levels of the Wnt/ß-catenin target protein Dlk1 inhibited the expression of adipogenesis regulators Pparg and Cebpa, and fatty acid transporter Cd36. Although both Ifrd1 and Ifrd2 contributed to this phenotype, they utilized two different mechanisms. Ifrd1 acted by controlling Wnt signaling and thereby transcriptional regulation of Dlk1. On the other hand, distinctive experimental evidence showed that Ifrd2 acts as a general translational inhibitor significantly affecting Dlk1 protein levels. Novel mechanisms of Dlk1 regulation in adipocyte differentiation involving Ifrd1 and Ifrd2 are based on experimental data presented here.


Subject(s)
Adipogenesis , Calcium-Binding Proteins , Immediate-Early Proteins , Membrane Proteins , Animals , Mice , Adipocytes , Adipogenesis/genetics , Adipose Tissue , Calcium-Binding Proteins/genetics , CD36 Antigens , Cell Differentiation , Membrane Proteins/genetics
2.
Genes (Basel) ; 14(7)2023 06 27.
Article in English | MEDLINE | ID: mdl-37510265

ABSTRACT

Congenital glucose-galactose malabsorption is a rare autosomal recessive disorder caused by mutations in SLC5A1 encoding the apical sodium/glucose cotransporter SGLT1. We present clinical and molecular data from eleven affected individuals with congenital glucose-galactose malabsorption from four unrelated, consanguineous Turkish families. Early recognition and timely management by eliminating glucose and galactose from the diet are fundamental for affected individuals to survive and develop normally. We identified novel SLC5A1 missense variants, p.Gly43Arg and p.Ala92Val, which were linked to disease in two families. Stable expression in CaCo-2 cells showed that the p.Ala92Val variant did not reach the plasma membrane, but was retained in the endoplasmic reticulum. The p.Gly43Arg variant, however, displayed processing and plasma membrane localization comparable to wild-type SGLT1. Glycine-43 displays nearly invariant conservation in the relevant structural family of cotransporters and exchangers, and localizes to SGLT1 transmembrane domain TM0. p.Gly43Arg represents the first disease-associated variant in TM0; however, the role of TM0 in the SGLT1 function has not been established. In summary, we are expanding the mutational spectrum of this rare disorder.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Humans , Caco-2 Cells , Carbohydrate Metabolism, Inborn Errors/genetics , Mutation , Glucose/metabolism , Sodium-Glucose Transporter 1/genetics
3.
J Clin Invest ; 132(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35575086

ABSTRACT

Variants in the UNC45A cochaperone have been recently associated with a syndrome combining diarrhea, cholestasis, deafness, and bone fragility. Yet the mechanism underlying intestinal failure in UNC45A deficiency remains unclear. Here, biallelic variants in UNC45A were identified by next-generation sequencing in 6 patients with congenital diarrhea. Corroborating in silico prediction, variants either abolished UNC45A expression or altered protein conformation. Myosin VB was identified by mass spectrometry as client of the UNC45A chaperone and was found misfolded in UNC45AKO Caco-2 cells. In keeping with impaired myosin VB function, UNC45AKO Caco-2 cells showed abnormal epithelial morphogenesis that was restored by full-length UNC45A, but not by mutant alleles. Patients and UNC45AKO 3D organoids displayed altered luminal development and microvillus inclusions, while 2D cultures revealed Rab11 and apical transporter mislocalization as well as sparse and disorganized microvilli. All those features resembled the subcellular abnormalities observed in duodenal biopsies from patients with microvillus inclusion disease. Finally, microvillus inclusions and shortened microvilli were evidenced in enterocytes from unc45a-deficient zebrafish. Taken together, our results provide evidence that UNC45A plays an essential role in epithelial morphogenesis through its cochaperone function of myosin VB and that UNC45A loss causes a variant of microvillus inclusion disease.


Subject(s)
Diarrhea, Infantile , Malabsorption Syndromes , Mucolipidoses , Myosin Type V , Animals , Caco-2 Cells , Diarrhea, Infantile/metabolism , Diarrhea, Infantile/pathology , Facies , Fetal Growth Retardation , Hair Diseases , Humans , Infant , Intracellular Signaling Peptides and Proteins/metabolism , Malabsorption Syndromes/metabolism , Microvilli/genetics , Microvilli/pathology , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mucolipidoses/pathology , Myosin Type V/genetics , Myosin Type V/metabolism , Phenotype , Zebrafish/genetics , Zebrafish/metabolism
4.
Cells ; 11(9)2022 04 21.
Article in English | MEDLINE | ID: mdl-35563717

ABSTRACT

Inflammatory skin diseases, including atopic dermatitis (AD) and psoriasis, are increasing in populations worldwide. The treatment of patients with AD and other forms of skin inflammation is mainly based on the use of topical corticosteroids or calcineurin inhibitors, which can cause significant side effects with long-term use. Therefore, there is a great need for the development of more effective and less toxic anti-inflammatory agents suitable for the treatment of chronic skin lesions. Here, we screened a number of strains from the ASIB 505 terrestrial algae collection and identified a green algae Chromochloris zofingiensis with pronounced anti-inflammatory properties. We found that a crude nonpolar extract of C. zofingiensis (ID name NAE_2022C), grown upon nitrogen deprivation, acts as a bioactive substance by inhibiting TNFR/NF-κB responses in human skin keratinocyte HaCaT cells. We also found that NAE_2022C suppressed the secretion of pro-inflammatory cytokine tumor necrosis factor α (TNFα) and several Th1- and Th2-related chemokines in a reconstituted human epidermis. The TNFR/NF-κB pathway analysis showed multiple inhibitory effects at different levels and disclosed a direct targeting of IKKß by the extract. Bioassay-guided fractionation followed by high-resolution mass spectrometry detected diacylglyceryl-trimethylhomoserine (DGTS), Lyso-DGTS (LDGTS), 5-phenylvaleric acid, theophylline and oleamide as leading metabolites in the active fraction of NAE_2022C. Further analysis identified betaine lipid DGTS (32:0) as one of the active compounds responsible for the NAE_2022C-mediated NF-κB suppression. Overall, this study presents an approach for the isolation, screening, and identification of anti-inflammatory secondary metabolites produced by soil algae.


Subject(s)
Dermatitis, Atopic , NF-kappa B , Anti-Inflammatory Agents/therapeutic use , Dermatitis, Atopic/pathology , Humans , NF-kappa B/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Soil
5.
Sci Rep ; 11(1): 2331, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504856

ABSTRACT

The nuclear factor kappa B (NF-κB) family of dimeric transcription factors regulates a wide range of genes by binding to their specific DNA regulatory sequences. NF-κB is an important therapeutic target linked to a number of cancers as well as autoimmune and inflammatory diseases. Therefore, effective high-throughput methods for the detection of NF-κB DNA binding are essential for studying its transcriptional activity and for inhibitory drug screening. We describe here a novel fluorescence-based assay for quantitative detection of κB consensus double-stranded (ds) DNA binding by measuring the thermal stability of the NF-κB proteins. Specifically, DNA binding proficient NF-κB probes, consisting of the N-terminal p65/RelA (aa 1-306) and p50 (aa 1-367) regions, were designed using bioinformatic analysis of protein hydrophobicity, folding and sequence similarities. By measuring the SYPRO Orange fluorescence during thermal denaturation of the probes, we detected and quantified a shift in the melting temperatures (ΔTm) of p65/RelA and p50 produced by the dsDNA binding. The increase in Tm was proportional to the concentration of dsDNA with apparent dissociation constants (KD) of 2.228 × 10-6 M and 0.794 × 10-6 M, respectively. The use of withaferin A (WFA), dimethyl fumarate (DMF) and p-xyleneselenocyanate (p-XSC) verified the suitability of this assay for measuring dose-dependent antagonistic effects on DNA binding. In addition, the assay can be used to analyse the direct binding of inhibitors and their effects on structural stability of the protein probe. This may facilitate the identification and rational design of new drug candidates interfering with NF-κB functions.


Subject(s)
Escherichia coli/metabolism , NF-kappa B/metabolism , Chromatography, Affinity , Computational Biology , DNA/metabolism , Drug Discovery , Electrophoretic Mobility Shift Assay
6.
Mol Genet Genomic Med ; 9(3): e1607, 2021 03.
Article in English | MEDLINE | ID: mdl-33471954

ABSTRACT

BACKGROUND: The epileptic encephalopathies display extensive locus and allelic heterogeneity. Biallelic truncating DOCK7 variants were recently reported in five children with early-onset epilepsy, intellectual disability, and cortical blindness, indicating that DOCK7 deficiency causes a specific type of epileptic encephalopathy. METHODS: We identified 23- and 27-year-old siblings with the clinical pattern reported for DOCK7 deficiency, and conducted genome-wide linkage analysis and WES. The consequences of a DOCK7 variant were analyzed on the transcript and protein level in patients' fibroblasts. RESULTS: We identified a novel homozygous DOCK7 frameshift variant, an intragenic tandem duplication of 124-kb, previously missed by CGH array, in adult patients. Patients display atrophy in the occipital lobe and pontine hypoplasia with marked pontobulbar sulcus, and focal atrophy of occasional cerebellar folia is a novel finding. Recognizable dysmorphic features include normo-brachycephaly, narrow forehead, low anterior and posterior hairlines, prominent ears, full cheeks, and long eyelashes. Our patients function on the level of 4-year-old children, never showed signs of regression, and seizures are largely controlled with multi-pharmacotherapy. Studies of patients' fibroblasts showed nonsense-mediated RNA decay and lack of DOCK7 protein. CONCLUSION: DOCK7 deficiency causes a definable clinical entity, a recognizable type of epileptic encephalopathy.


Subject(s)
Blindness, Cortical/genetics , Craniofacial Abnormalities/genetics , Epilepsy/genetics , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Adult , Blindness, Cortical/pathology , Cells, Cultured , Craniofacial Abnormalities/pathology , Epilepsy/pathology , Female , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Homozygote , Humans , Mutation , Syndrome
7.
Front Immunol ; 11: 2010, 2020.
Article in English | MEDLINE | ID: mdl-32922405

ABSTRACT

Dendritic cells (DCs) possess intrinsic cellular defense mechanisms to specifically inhibit HIV-1 replication. In turn, HIV-1 has evolved strategies to evade innate immune sensing by DCs resulting in suboptimal maturation and poor antiviral immune responses. We previously showed that complement-opsonized HIV-1 (HIV-C) was able to efficiently infect various DC subsets significantly higher than non-opsonized HIV-1 (HIV) and therefore also mediate a higher antiviral immunity. Thus, complement coating of HIV-1 might play a role with respect to viral control occurring early during infection via modulation of DCs. To determine in detail which complement receptors (CRs) expressed on DCs was responsible for infection and superior pro-inflammatory and antiviral effects, we generated stable deletion mutants for the α-chains of CR3, CD11b, and CR4, CD11c using CRISPR/Cas9 in THP1-derived DCs. We found that CD11c deletion resulted in impaired DC infection as well as antiviral and pro-inflammatory immunity upon exposure to complement-coated HIV-1. In contrast, sole expression of CD11b on DCs shifted the cells to an anti-inflammatory, regulatory DC type. We here illustrated that CR4 comprised of CD11c and CD18 is the major player with respect to DC infection associated with a potent early pro-inflammatory immune response. A more detailed characterization of CR3 and CR4 functions using our powerful tool might open novel avenues for early therapeutic intervention during HIV-1 infection.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/physiology , Integrin alphaXbeta2/metabolism , Macrophage-1 Antigen/metabolism , CD11b Antigen/genetics , CD11c Antigen/genetics , CD18 Antigens/genetics , CRISPR-Cas Systems , Complement System Proteins/metabolism , Humans , Immunity , Integrin alphaXbeta2/genetics , Macrophage-1 Antigen/genetics , Sequence Deletion/genetics , Signal Transduction , THP-1 Cells
8.
Clin Genet ; 98(3): 282-287, 2020 09.
Article in English | MEDLINE | ID: mdl-32557621

ABSTRACT

Biallelic loss-of-function mutations in the centrosomal pericentrin gene (PCNT) cause microcephalic osteodysplastic primordial dwarfism type II (MOPDII), which is characterized by extreme growth retardation, microcephaly, skeletal dysplasia, and dental anomalies. Life expectancy is reduced due to a high risk of cerebral vascular anomalies. Here, we report two siblings with MOPDII and attenuated growth restriction, and pachygyria. Compound heterozygosity for two novel truncated PCNT variants was identified. Both truncated PCNT proteins were expressed in patient's fibroblasts, with a reduced total protein amount compared to control. Patient's fibroblasts showed impaired cell cycle progression. As a novel finding, 20% of patient's fibroblasts were shown to express PCNT comparable to control. This was associated with normal mitotic morphology and normal co-localization of mutated PCNT with centrosome-associated proteins γ-tubulin and centrin 3, suggesting some residual function of truncated PCNT proteins. These data expand the clinical and molecular spectrum of MOPDII and indicate that residual PCNT function might be associated with attenuated growth restriction in MOPDII.


Subject(s)
Antigens/genetics , Dwarfism/genetics , Fetal Growth Retardation/genetics , Genetic Predisposition to Disease , Lissencephaly/genetics , Microcephaly/genetics , Osteochondrodysplasias/genetics , Adolescent , Adult , Alleles , Centrosome/metabolism , Child , Child, Preschool , Dwarfism/pathology , Female , Fetal Growth Retardation/pathology , Fibroblasts/metabolism , Humans , Lissencephaly/pathology , Loss of Function Mutation/genetics , Male , Microcephaly/pathology , Osteochondrodysplasias/pathology , Siblings , Tubulin/genetics , Young Adult
9.
Hum Mutat ; 41(4): 753-758, 2020 04.
Article in English | MEDLINE | ID: mdl-31898838

ABSTRACT

ACTB encodes ß-cytoplasmic actin, an essential component of the cytoskeleton. Based on chromosome 7p22.1 deletions that include the ACTB locus and on rare truncating ACTB variants, a phenotype resulting from ACTB haploinsufficiency was recently proposed. We report putative ACTB loss-of-function variants in four patients. To the best of our knowledge, we report the first 7p22.1 microdeletion confined to ACTB and the second ACTB frameshifting mutation that predicts mRNA decay. A de-novo ACTB p.(Gly302Ala) mutation affects ß-cytoplasmic actin distribution. All four patients share a facial gestalt that is distinct from that of individuals with dominant-negative ACTB variants in Baraitser-Winter cerebrofrontofacial syndrome. Two of our patients had strikingly thin and sparse scalp hair. One patient had sagittal craniosynostosis and hypospadias. All three affected male children have attention deficits and mild global developmental delay. Mild intellectual disability was present in only one patient. Heterozygous ACTB deletion can allow for normal psychomotor function.


Subject(s)
Actins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Loss of Function Mutation , Actins/chemistry , Adult , Child , Child, Preschool , Facies , Female , Genetic Association Studies/methods , Genetic Loci , Humans , Magnetic Resonance Imaging , Male , Models, Molecular , Phenotype , Protein Conformation , Structure-Activity Relationship
10.
Front Mol Neurosci ; 12: 73, 2019.
Article in English | MEDLINE | ID: mdl-30983969

ABSTRACT

Sprouty proteins act ubiquitously as signaling integrators and inhibitors of receptor tyrosine kinase (RTK) activated pathways. Among the four Sprouty isoforms, Sprouty2 is a key regulator of growth factor signaling in several neurological disorders. High protein levels correlate with reduced survival of glioma patients. We recently demonstrated that abrogating its function inhibits tumor growth by overstimulation of ERK and induction of DNA replication stress. The important role of Sprouty2 in the proliferation of malignant glioma cells prompted us to investigate its subcellular localization applying super-resolution fluorescence and immunoelectron microscopy. We found that cytoplasmic Sprouty2 is not homogenously distributed but localized to small spots (<100 nm) partly attached to vimentin filaments and co-localized with activated ERK. The protein is associated with early, late and recycling endosomes in response to but also independently of growth factor stimulation. The subcellular localization of Sprouty2 in all areas exhibiting strong RTK activities may reflect a protective response of glioma cells to limit excessive ERK activation and to prevent cellular senescence and apoptosis.

11.
BMC Biol ; 14(1): 95, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27782840

ABSTRACT

BACKGROUND: TPA Induced Sequence 7 acts as a transcriptional co-regulator controlling the expression of genes involved in differentiation of various cell types, including skeletal myoblasts. We and others have shown that TIS7 regulates adult myogenesis through MyoD, one of the essential myogenic regulatory factors. RESULTS: Here, we present data identifying ICln as the specific, novel protein downstream of TIS7 controlling myogenesis. We show that TIS7/ICln epigenetically regulate myoD expression controlling protein methyl transferase activity. In particular, ICln regulates MyoD expression via its interaction with PRMT5 by an epigenetic modification that utilizes symmetrical di-methylation of histone H3 on arginine 8. We provide multiple evidences that TIS7 directly binds DNA, which is a functional feature necessary for its role in transcriptional regulation. CONCLUSION: We present here a molecular insight into TIS7-specific control of MyoD gene expression and thereby skeletal muscle differentiation.


Subject(s)
Cell Differentiation/physiology , Immediate-Early Proteins/metabolism , Membrane Proteins/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Immunoblotting , Membrane Proteins/genetics , Mice , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Protein Binding , Real-Time Polymerase Chain Reaction , Surface Plasmon Resonance , Transcription, Genetic/genetics
12.
Proc Natl Acad Sci U S A ; 113(16): 4326-31, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27051065

ABSTRACT

Nuclear factor kappa B (NF-κB) is an inducible transcription factor that plays critical roles in immune and stress responses and is often implicated in pathologies, including chronic inflammation and cancer. Although much has been learned about NF-κB-activating pathways, the specific repression of NF-κB is far less well understood. Here we identified the type I protein arginine methyltransferase 1 (PRMT1) as a restrictive factor controlling TNFα-induced activation of NF-κB. PRMT1 forms a cellular complex with NF-κB through direct interaction with the Rel homology domain of RelA. We demonstrate that PRMT1 methylates RelA at evolutionary conserved R30, located in the DNA-binding L1 loop, which is a critical residue required for DNA binding. Asymmetric R30 dimethylation inhibits the binding of RelA to DNA and represses NF-κB target genes in response to TNFα. Molecular dynamics simulations of the DNA-bound RelA:p50 predicted structural changes in RelA caused by R30 methylation or a mutation that interferes with the stability of the DNA-NF-κB complex. Our findings provide evidence for the asymmetric arginine dimethylation of RelA and unveil a unique mechanism controlling TNFα/NF-κB signaling.


Subject(s)
Arginine/analogs & derivatives , Signal Transduction/physiology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Arginine/genetics , Arginine/metabolism , Cell Line , Humans , Methylation , Mice , Mice, Knockout , Molecular Dynamics Simulation , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/genetics
13.
Biol Open ; 3(5): 397-407, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24771621

ABSTRACT

The myc protooncogene encodes the Myc transcription factor which is the essential part of the Myc-Max network controlling fundamental cellular processes. Deregulation of myc leads to tumorigenesis and is a hallmark of many human cancers. We have recently identified homologs of myc (myc1, myc2) and max in the early diploblastic cnidarian Hydra and have characterized myc1 in detail. Here we show that myc2 is transcriptionally activated in the interstitial stem cell system. Furthermore, in contrast to myc1, myc2 expression is also detectable in proliferating epithelial stem cells throughout the gastric region. myc2 but not myc1 is activated in cycling precursor cells during early oogenesis and spermatogenesis, suggesting that the Hydra Myc2 protein has a possible non-redundant function in cell cycle progression. The Myc2 protein displays the principal design and properties of vertebrate Myc proteins. In complex with Max, Myc2 binds to DNA with similar affinity as Myc1-Max heterodimers. Immunoprecipitation of Hydra chromatin revealed that both Myc1 and Myc2 bind to the enhancer region of CAD, a classical Myc target gene in mammals. Luciferase reporter gene assays showed that Myc1 but not Myc2 transcriptionally activates the CAD promoter. Myc2 has oncogenic potential when tested in primary avian fibroblasts but to a lower degree as compared to Myc1. The identification of an additional myc gene in Cnidaria, a phylum that diverged prior to bilaterians, with characteristic expression patterns in tissue homeostasis and developmental processes suggests that principle functions of myc genes have arisen very early in metazoan evolution.

14.
Sci Rep ; 3: 3444, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24309437

ABSTRACT

The c-myc protooncogene encodes the Myc transcription factor, a global regulator of fundamental cellular processes. Deregulation of c-myc leads to tumorigenesis, and c-myc is an important driver in human cancer. Myc and its dimerization partner Max are bHLH-Zip DNA binding proteins involved in transcriptional regulation of target genes. Non-transcriptional functions have also been attributed to the Myc protein, notably direct interaction with the pre-replicative complex (pre-RC) controlling the initiation of DNA replication. A key component of the pre-RC is the Cdt1 protein, an essential factor in origin licensing. Here we present data suggesting that the CDT1 gene is a transcriptional target of the Myc-Max complex. Expression of the CDT1 gene in v-myc-transformed cells directly correlates with myc expression. Also, human tumor cells with elevated c-myc expression display increased CDT1 expression. Occupation of the CDT1 promoter by Myc-Max is demonstrated by chromatin immunoprecipitation, and transactivation by Myc-Max is shown in reporter assays. Ectopic expression of CDT1 leads to cell transformation. Our results provide a possible direct mechanistic link of Myc's canonical function as a transcription factor to DNA replication. Furthermore, we suggest that aberrant transcriptional activation of CDT1 by deregulated myc alleles contributes to the genomic instabilities observed in tumor cells.


Subject(s)
DNA Replication , Gene Expression Regulation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Chickens , Gene Order , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Transcriptional Activation
15.
Proc Natl Acad Sci U S A ; 110(21): 8531-6, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23657011

ABSTRACT

Activated G protein-coupled receptors (GPCRs) and receptor tyrosine kinases relay extracellular signals through spatial and temporal controlled kinase and GTPase entities. These enzymes are coordinated by multifunctional scaffolding proteins for precise intracellular signal processing. The cAMP-dependent protein kinase A (PKA) is the prime example for compartmentalized signal transmission downstream of distinct GPCRs. A-kinase anchoring proteins tether PKA to specific intracellular sites to ensure precision and directionality of PKA phosphorylation events. Here, we show that the Rho-GTPase Rac contains A-kinase anchoring protein properties and forms a dynamic cellular protein complex with PKA. The formation of this transient core complex depends on binary interactions with PKA subunits, cAMP levels and cellular GTP-loading accounting for bidirectional consequences on PKA and Rac downstream signaling. We show that GTP-Rac stabilizes the inactive PKA holoenzyme. However, ß-adrenergic receptor-mediated activation of GTP-Rac-bound PKA routes signals to the Raf-Mek-Erk cascade, which is critically implicated in cell proliferation. We describe a further mechanism of how cAMP enhances nuclear Erk1/2 signaling: It emanates from transphosphorylation of p21-activated kinases in their evolutionary conserved kinase-activation loop through GTP-Rac compartmentalized PKA activities. Sole transphosphorylation of p21-activated kinases is not sufficient to activate Erk1/2. It requires complex formation of both kinases with GTP-Rac1 to unleash cAMP-PKA-boosted activation of Raf-Mek-Erk. Consequently GTP-Rac functions as a dual kinase-tuning scaffold that favors the PKA holoenzyme and contributes to potentiate Erk1/2 signaling. Our findings offer additional mechanistic insights how ß-adrenergic receptor-controlled PKA activities enhance GTP-Rac-mediated activation of nuclear Erk1/2 signaling.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , MAP Kinase Signaling System/physiology , Multienzyme Complexes/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line, Tumor , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Multienzyme Complexes/genetics , Phosphorylation/physiology , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , rac1 GTP-Binding Protein/genetics , raf Kinases/genetics , raf Kinases/metabolism
16.
Nat Methods ; 10(4): 339-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455924

ABSTRACT

To accelerate high-density interactome mapping, we developed a yeast two-hybrid interaction screening approach involving short-read second-generation sequencing (Y2H-seq) with improved sensitivity and a quantitative scoring readout allowing rapid interaction validation. We applied Y2H-seq to investigate enzymes involved in protein methylation, a largely unexplored post-translational modification. The reported network of 523 interactions involving 22 methyltransferases or demethylases is comprehensively annotated and validated through coimmunoprecipitation experiments and defines previously undiscovered cellular roles of nonhistone protein methylation.


Subject(s)
Methyltransferases/metabolism , Protein Interaction Mapping/methods , Two-Hybrid System Techniques , Chromatography, Liquid , Escherichia coli , Gene Expression Regulation, Enzymologic/physiology , HEK293 Cells , Humans , Methyltransferases/genetics , Sensitivity and Specificity , Sequence Analysis, DNA/methods , Tandem Mass Spectrometry
17.
Chemistry ; 17(16): 4460-9, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21433149

ABSTRACT

Electron detachment dissociation (EDD) is an emerging mass spectrometry (MS) technique for the primary structure analysis of peptides, carbohydrates, and oligonucleotides. Herein, we explore the potential of EDD for sequencing of proteins of up to 147 amino acid residues by using top-down MS. Sequence coverage ranged from 72% for Melittin, which lacks carboxylic acid functionalities, to 19% for an acidic 147-residue protein, to 12% for Ferredoxin, which showed unusual backbone fragmentation next to cysteine residues. A limiting factor for protein sequencing by EDD is the facile loss of small molecules from amino acid side chains, in particular CO(2). Based on the types of fragments observed and fragmentation patterns found, we propose detailed mechanisms for protein backbone cleavage and side chain dissociation in EDD. The insights from this study should further the development of EDD for top-down MS of acidic proteins.


Subject(s)
Electrons , Mass Spectrometry/methods , Proteins/chemistry , Amino Acids/chemistry , Ferredoxins/chemistry , Hydrogen-Ion Concentration , Melitten/chemistry , Models, Molecular , Molecular Structure
18.
Biochem J ; 434(2): 253-63, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21158742

ABSTRACT

NF-κB (nuclear factor κB) controls diverse cellular processes and is frequently misregulated in chronic immune diseases or cancer. The activity of NF-κB is regulated by IκB (inhibitory κB) proteins which control nuclear-cytoplasmic shuttling and DNA binding of NF-κB. In the present paper, we describe a novel role for p65 as a critical regulator of the cellular localization and functions of NF-κB and its inhibitor IκBß. In genetically modified p65-/- cells, the localization of ectopic p65 is not solely regulated by IκBα, but is largely dependent on the NLS (nuclear localization signal) and the NES (nuclear export signal) of p65. Furthermore, unlike IκBα, IκBß does not contribute to the nuclear export of p65. In fact, the cellular localization and degradation of IκBß is controlled by the p65-specific NLS and NES. The results of our present study also reveal that, in addition to stimulus-induced redistribution of NF-κB, changes in the constitutive localization of p65 and IκBß specifically modulate activation of inflammatory genes. This is a consequence of differences in the DNA-binding activity and signal responsiveness between the nuclear and cytoplasmic NF-κB-IκBß complexes. Taken together, the findings of the present study indicate that the p65 subunit controls transcriptional competence of NF-κB by regulating the NF-κB/IκBß pathway.


Subject(s)
I-kappa B Proteins/analysis , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Mice , Nuclear Localization Signals/metabolism , Signal Transduction , Transcription Factor RelA/genetics
19.
Proc Natl Acad Sci U S A ; 107(9): 4051-6, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20142507

ABSTRACT

The c-myc protooncogene encodes a transcription factor (Myc) with oncogenic potential. Myc and its dimerization partner Max are bHLH-Zip DNA binding proteins controlling fundamental cellular processes. Deregulation of c-myc leads to tumorigenesis and is a hallmark of many human cancers. We have identified and extensively characterized ancestral forms of myc and max genes from the early diploblastic cnidarian Hydra, the most primitive metazoan organism employed so far for the structural, functional, and evolutionary analysis of these genes. Hydra myc is specifically activated in all stem cells and nematoblast nests which represent the rapidly proliferating cell types of the interstitial stem cell system and in proliferating gland cells. In terminally differentiated nerve cells, nematocytes, or epithelial cells, myc expression is not detectable by in situ hybridization. Hydra max exhibits a similar expression pattern in interstitial cell clusters. The ancestral Hydra Myc and Max proteins display the principal design of their vertebrate derivatives, with the highest degree of sequence identities confined to the bHLH-Zip domains. Furthermore, the 314-amino acid Hydra Myc protein contains basic forms of the essential Myc boxes I through III. A recombinant Hydra Myc/Max complex binds to the consensus DNA sequence CACGTG with high affinity. Hybrid proteins composed of segments from the retroviral v-Myc oncoprotein and the Hydra Myc protein display oncogenic potential in cell transformation assays. Our results suggest that the principal functions of the Myc master regulator arose very early in metazoan evolution, allowing their dissection in a simple model organism showing regenerative ability but no senescence.


Subject(s)
Genes, myc , Hydra/physiology , Proto-Oncogene Proteins c-myc/metabolism , Stem Cells/cytology , Amino Acid Sequence , Animals , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Lineage , Hydra/genetics , Molecular Sequence Data , Proto-Oncogene Proteins c-myc/chemistry , Sequence Homology, Amino Acid
20.
Proc Natl Acad Sci U S A ; 106(14): 5604-9, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19297618

ABSTRACT

Cell transformation by the Myc oncoprotein involves transcriptional activation or suppression of specific target genes with intrinsic oncogenic or tumor-suppressive potential, respectively. We have identified the BASP1 (CAP-23, NAP-22) gene as a novel target suppressed by Myc. The acidic 25-kDa BASP1 protein was originally isolated as a cortical cytoskeleton-associated protein from rat and chicken brain, but has also been found in other tissues and subcellular locations. BASP1 mRNA and protein expression is specifically suppressed in fibroblasts transformed by the v-myc oncogene, but not in cells transformed by other oncogenic agents. The BASP1 gene encompasses 2 exons separated by a 58-kbp intron and a Myc-responsive regulatory region at the 5' boundary of untranslated exon 1. Bicistronic expression of BASP1 and v-myc from a retroviral vector blocks v-myc-induced cell transformation. Furthermore, ectopic expression of BASP1 renders fibroblasts resistant to subsequent cell transformation by v-myc, and exogenous delivery of the BASP1 gene into v-myc-transformed cells leads to significant attenuation of the transformed phenotype. The inhibition of v-myc-induced cell transformation by BASP1 also prevents the transcriptional activation or repression of known Myc target genes. Mutational analysis showed that the basic N-terminal domain containing a myristoylation site, a calmodulin binding domain, and a putative nuclear localization signal is essential for the inhibitory function of BASP1. Our results suggest that down-regulation of the BASP1 gene is a necessary event in myc-induced oncogenesis and define the BASP1 protein as a potential tumor suppressor.


Subject(s)
Calmodulin-Binding Proteins/physiology , Cell Transformation, Neoplastic , Nerve Tissue Proteins/physiology , Proto-Oncogene Proteins c-myc/physiology , Tumor Suppressor Proteins/physiology , Animals , Binding Sites , Calmodulin , Chickens , Gene Expression Regulation, Neoplastic , Molecular Sequence Data , Myristic Acid , Nuclear Localization Signals , Quail
SELECTION OF CITATIONS
SEARCH DETAIL
...