Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(4): 1032-1046.e18, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33571428

ABSTRACT

Human immunodeficiency virus (HIV-1) remains a major health threat. Viral capsid uncoating and nuclear import of the viral genome are critical for productive infection. The size of the HIV-1 capsid is generally believed to exceed the diameter of the nuclear pore complex (NPC), indicating that capsid uncoating has to occur prior to nuclear import. Here, we combined correlative light and electron microscopy with subtomogram averaging to capture the structural status of reverse transcription-competent HIV-1 complexes in infected T cells. We demonstrated that the diameter of the NPC in cellulo is sufficient for the import of apparently intact, cone-shaped capsids. Subsequent to nuclear import, we detected disrupted and empty capsid fragments, indicating that uncoating of the replication complex occurs by breaking the capsid open, and not by disassembly into individual subunits. Our data directly visualize a key step in HIV-1 replication and enhance our mechanistic understanding of the viral life cycle.


Subject(s)
Capsid/metabolism , HIV-1/metabolism , Nuclear Pore/metabolism , Active Transport, Cell Nucleus , Capsid/ultrastructure , Cryoelectron Microscopy , HEK293 Cells , HIV Infections/virology , HIV-1/ultrastructure , Humans , Models, Biological , Nuclear Pore/ultrastructure , Nuclear Pore/virology , Reverse Transcription , Virion/metabolism , Virus Internalization , mRNA Cleavage and Polyadenylation Factors/metabolism
2.
Nat Methods ; 17(3): 279-282, 2020 03.
Article in English | MEDLINE | ID: mdl-32066961

ABSTRACT

We introduce an engineered nanobody whose affinity to green fluorescent protein (GFP) can be switched on and off with small molecules. By controlling the cellular localization of GFP fusion proteins, the engineered nanobody allows interrogation of their roles in basic biological processes, an approach that should be applicable to numerous previously described GFP fusions. We also outline how the binding affinities of other nanobodies can be controlled by small molecules.


Subject(s)
Green Fluorescent Proteins/chemistry , Immunoglobulin Fragments/chemistry , Nanoparticles/chemistry , Single-Domain Antibodies/chemistry , Crystallography, X-Ray , DNA/chemistry , Databases, Protein , Escherichia coli , Fluorescence Resonance Energy Transfer , Gene Products, gag/chemistry , HEK293 Cells , HIV-1/chemistry , HeLa Cells , Humans , Kinetics , Ligands , Microscopy, Fluorescence , Mitosis , Protein Domains , nef Gene Products, Human Immunodeficiency Virus/chemistry
3.
Brain ; 144(6): 1833-1852, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33880508

ABSTRACT

Amyloid-ß precursor protein (APP) is sequentially cleaved by secretases and generates amyloid-ß, the major components in senile plaques in Alzheimer's disease. APP is upregulated in human Alzheimer's disease brains. However, the molecular mechanism of how APP contributes to Alzheimer's disease pathogenesis remains incompletely understood. Here we show that truncated APP C586-695 fragment generated by δ-secretase directly binds to CCAAT/enhancer-binding protein beta (CEBPB), an inflammatory transcription factor, and enhances its transcriptional activity, escalating Alzheimer's disease-related gene expression and pathogenesis. The APP C586-695 fragment, but not full-length APP, strongly associates with CEBPB and elicits its nuclear translocation and augments the transcriptional activities on APP itself, MAPT (microtubule-associated protein tau), δ-secretase and inflammatory cytokine mRNA expression, finally triggering Alzheimer's disease pathology and cognitive disorder in a viral overexpression mouse model. Blockade of δ-secretase cleavage of APP by mutating the cleavage sites reduces its stimulatory effect on CEBPB, alleviating amyloid pathology and cognitive dysfunctions. Clearance of APP C586-695 from 5xFAD mice by antibody administration mitigates Alzheimer's disease pathologies and restores cognitive functions. Thus, in addition to the sequestration of amyloid-ß, APP implicates in Alzheimer's disease pathology by activating CEBPB upon δ-secretase cleavage.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation/physiology , Aged , Animals , Cysteine Endopeptidases/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
4.
Int J Mol Sci ; 23(9)2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35563046

ABSTRACT

The ß- and γ-secretase-driven cleavage of the amyloid precursor protein (APP) gives rise to the amyloid ß peptide, which is believed to be the main driver of neurodegeneration in Alzheimer's disease (AD). As it is prominently detectable in extracellular plaques in post-mortem AD brain samples, research in recent decades focused on the pathological role of extracellular amyloid ß aggregation, widely neglecting the potential meaning of very early generation of amyloid ß inside the cell. In the last few years, the importance of intracellular amyloid ß (iAß) as a strong player in neurodegeneration has been indicated by a rising number of studies. In this review, iAß is highlighted as a crucial APP cleavage fragment, able to manipulate intracellular pathways and foster neurodegeneration. We demonstrate its relevance as a pathological marker and shed light on initial studies aiming to modulate iAß through pharmacological treatment, which has been shown to have beneficial effects on cognitive properties in animal models. Finally, we display the relevance of viral infections on iAß generation and point out future directions urgently needed to manifest the potential relevance of iAß in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Biomarkers , Plaque, Amyloid
5.
Chembiochem ; 22(3): 548-556, 2021 02 02.
Article in English | MEDLINE | ID: mdl-32974998

ABSTRACT

Selective targeting of DNA by means of fluorescent labeling has become a mainstay in the life sciences. While genetic engineering serves as a powerful technique and allows the visualization of nucleic acid by using DNA-targeting fluorescent fusion proteins in a cell-type- and subcellular-specific manner, it relies on the introduction of foreign genes. On the other hand, DNA-binding small fluorescent molecules can be used without genetic engineering, but they are not spatially restricted. Herein, we report a photocaged version of the DNA dye Hoechst33342 (pcHoechst), which can be uncaged by using UV to blue light for the selective staining of chromosomal DNA in subnuclear regions of live cells. Expanding its application to a vertebrate model organism, we demonstrate uncaging in epithelial cells and short-term cell tracking in vivo in zebrafish. We envision pcHoechst as a valuable tool for targeting and interrogating DNA with precise spatiotemporal resolution in living cells and wild-type organisms.


Subject(s)
DNA/chemistry , Fluorescent Dyes/chemistry , Animals , Epithelial Cells/chemistry , HeLa Cells , Humans , Light , Luminescent Proteins/chemistry , Molecular Structure , Photochemical Processes , Recombinant Fusion Proteins/chemistry , Zebrafish
6.
Molecules ; 24(3)2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30700005

ABSTRACT

The replication of a virus within its host cell involves numerous interactions between viral and cellular factors, which have to be tightly controlled in space and time. The intricate interplay between viral exploitation of cellular pathways and the intrinsic host defense mechanisms is difficult to unravel by traditional bulk approaches. In recent years, novel fluorescence microscopy techniques and single virus tracking have transformed the investigation of dynamic virus-host interactions. A prerequisite for the application of these imaging-based methods is the attachment of a fluorescent label to the structure of interest. However, their small size, limited coding capacity and multifunctional proteins render viruses particularly challenging targets for fluorescent labeling approaches. Click chemistry in conjunction with genetic code expansion provides virologists with a novel toolbox for site-specific, minimally invasive labeling of virion components, whose potential has just recently begun to be exploited. Here, we summarize recent achievements, current developments and future challenges for the labeling of viral nucleic acids, proteins, glycoproteins or lipids using click chemistry in order to study dynamic processes in virus-cell interactions.


Subject(s)
Click Chemistry/methods , Virus Replication/physiology , Humans , Microscopy, Fluorescence
8.
PLoS Pathog ; 12(11): e1005964, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27812216

ABSTRACT

Cell-to-cell spread of HIV, a directed mode of viral transmission, has been observed to be more rapid than cell-free infection. However, a mechanism for earlier onset of viral gene expression in cell-to-cell spread was previously uncharacterized. Here we used time-lapse microscopy combined with automated image analysis to quantify the timing of the onset of HIV gene expression in a fluorescent reporter cell line, as well as single cell staining for infection over time in primary cells. We compared cell-to-cell spread of HIV to cell-free infection, and limited both types of transmission to a two-hour window to minimize differences due to virus transit time to the cell. The mean time to detectable onset of viral gene expression in cell-to-cell spread was accelerated by 19% in the reporter cell line and by 35% in peripheral blood mononuclear cells relative to cell-free HIV infection. Neither factors secreted by infected cells, nor contact with infected cells in the absence of transmission, detectably changed onset. We recapitulated the earlier onset by infecting with multiple cell-free viruses per cell. Surprisingly, the acceleration in onset of viral gene expression was not explained by cooperativity between infecting virions. Instead, more rapid onset was consistent with a model where the fastest expressing virus out of the infecting virus pool sets the time for infection independently of the other co-infecting viruses.


Subject(s)
Gene Expression Regulation, Viral/physiology , HIV Infections/genetics , HIV Infections/transmission , HIV Infections/virology , HIV-1/genetics , Cells, Cultured , Flow Cytometry , Humans , Time-Lapse Imaging
9.
J Cell Biochem ; 118(9): 2752-2763, 2017 09.
Article in English | MEDLINE | ID: mdl-28176357

ABSTRACT

BRI2 is a ubiquitously expressed type II transmembrane phosphoprotein. BRI2 undergoes proteolytic processing into secreted fragments and during the maturation process it suffers post-translational modifications. Of particular relevance, BRI2 is a protein phosphatase 1 (PP1) interacting protein, where PP1 is able to dephosphorylate the former. Further, disruption of the BRI2:PP1 complex, using BRI2 PP1 binding motif mutants, leads to increased BRI2 phosphorylation levels. However, the physiological function of BRI2 remains elusive; although findings suggest a role in neurite outgrowth and neuronal differentiation. In the work here presented, BRI2 expression during neuronal development was investigated. This increases during neuronal differentiation and an increase in its proteolytic processing is also evident. To elucidate the importance of BRI2 phosphorylation for both proteolytic processing and neuritogenesis, SH-SY5Y cells were transfected with the BRI2 PP1 binding motif mutant constructs. For the first time, it was possible to show that BRI2 phosphorylation is an important regulatory mechanism for its proteolytic processing and its neuritogenic role. Furthermore, by modulating BRI2 processing using an ADAM10 inhibitor, a dual role for BRI2 in neurite outgrowth is suggested: phosphorylated full-length BRI2 appears to be important for the formation of neuritic processes, and BRI2 NTF promotes neurite elongation. This work significantly contributed to the understanding of the physiological function of BRI2 and its regulation by protein phosphorylation. J. Cell. Biochem. 118: 2752-2763, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation , Membrane Glycoproteins/metabolism , Multiprotein Complexes/metabolism , Neurites/metabolism , Protein Processing, Post-Translational , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Humans , Membrane Glycoproteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Multiprotein Complexes/genetics , Phosphorylation/genetics , Protein Phosphatase 1/genetics , Proteolysis , Rats , Rats, Wistar
10.
Biochim Biophys Acta Proteins Proteom ; 1865(6): 703-714, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28377147

ABSTRACT

BACKGROUND: Human hippocampal area Cornu Ammonis (CA) 1 is one of the first fields in the human telencephalon showing Alzheimer disease (AD)-specific neuropathological changes. In contrast, CA2 and CA3 are far later affected pointing to functional differences, which may be accompanied by differences in proteome endowment and changes. METHODS: Human pyramidal cell layers of hippocampal areas CA1, CA2, and CA3 from neurologically unaffected individuals were excised using laser microdissection. The proteome of each individual sample was analyzed and differentially abundant proteins were validated by immuno-histochemistry. RESULTS: Comparison of CA1 to CA2 revealed 223, CA1 to CA3 197 proteins with differential abundance, among them we found motor proteins MYO5A and DYNC1H1. Extension of the study to human hippocampus slices from AD patients revealed extensive depletion of these proteins in CA1 area compared to unaffected controls. CONCLUSION: High abundance of motor proteins in pyramidal cell layers CA1 compared to CA2 and CA3 points the specific vulnerability of this hippocampal area to transport-associated changes based on microtubule dysfunction and destabilization in AD.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Proteomics , Aged , Aged, 80 and over , Chromatography, Liquid , Female , Humans , Immunohistochemistry , Male , Middle Aged , Tandem Mass Spectrometry
11.
Angew Chem Int Ed Engl ; 56(49): 15737-15741, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28960788

ABSTRACT

The characterization of low-affinity protein complexes is challenging due to their dynamic nature. Here, we present a method to stabilize transient protein complexes in vivo by generating a covalent and conformationally flexible bridge between the interaction partners. A highly active pyrrolysyl tRNA synthetase mutant directs the incorporation of unnatural amino acids bearing bromoalkyl moieties (BrCnK) into proteins. We demonstrate for the first time that low-affinity protein complexes between BrCnK-containing proteins and their binding partners can be stabilized in vivo in bacterial and mammalian cells. Using this approach, we determined the crystal structure of a transient GDP-bound complex between a small G-protein and its nucleotide exchange factor. We envision that this approach will prove valuable as a general tool for validating and characterizing protein-protein interactions in vitro and in vivo.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , GTP-Binding Protein Regulators/metabolism , GTP-Binding Proteins/metabolism , Green Fluorescent Proteins/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , GTP-Binding Protein Regulators/chemistry , GTP-Binding Proteins/chemistry , Green Fluorescent Proteins/chemistry , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Binding , Protein Stability
12.
Mol Cell Proteomics ; 13(2): 475-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284412

ABSTRACT

FE65 is a cytosolic adapter protein and an important binding partner of amyloid precursor protein. Dependent on Thr668 phosphorylation in amyloid precursor protein, which influences amyloidogenic amyloid precursor protein processing, FE65 undergoes nuclear translocation, thereby transmitting a signal from the cell membrane to the nucleus. As this translocation may be relevant in Alzheimer disease, and as FE65 consists of three protein-protein interaction domains able to bind and affect a variety of other proteins and downstream signaling pathways, the identification of the FE65 interactome is of central interest in Alzheimer disease research. In this study, we identified 121 proteins as new potential FE65 interacting proteins in a pulldown/mass spectrometry approach using human post-mortem brain samples as protein pools for recombinantly expressed FE65. Co-immunoprecipitation assays further validated the interaction of FE65 with the candidates SV2A and SERCA2. In parallel, we investigated the whole cell proteome of primary hippocampal neurons from FE65/FE65L1 double knockout mice. Notably, the validated FE65 binding proteins were also found to be differentially abundant in neurons derived from the FE65 knockout mice relative to wild-type control neurons. SERCA2 is an important player in cellular calcium homeostasis, which was found to be up-regulated in double knockout neurons. Indeed, knock-down of FE65 in HEK293T cells also evoked an elevated sensitivity to thapsigargin, a stressor specifically targeting the activity of SERCA2. Thus, our results suggest that FE65 is involved in the regulation of intracellular calcium homeostasis. Whereas transfection of FE65 alone caused a typical dot-like phenotype in the nucleus, co-transfection of SV2A significantly reduced the percentage of FE65 dot-positive cells, pointing to a possible role for SV2A in the modulation of FE65 intracellular targeting. Given that SV2A has a signaling function at the presynapse, its effect on FE65 intracellular localization suggests that the SV2A/FE65 interaction might play a role in synaptic signal transduction.


Subject(s)
Brain/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Interaction Maps , Animals , Brain/pathology , Cells, Cultured , Embryo, Mammalian , HEK293 Cells , Humans , Immunoprecipitation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/isolation & purification , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/genetics , Protein Binding , Protein Interaction Maps/genetics , Synapses/genetics , Synapses/metabolism
13.
Traffic ; 14(12): 1290-301, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24034674

ABSTRACT

Peroxisomal matrix protein import is facilitated by cycling receptors that recognize their cargo proteins in the cytosol by a peroxisomal targeting sequence (PTS) and ferry them to the peroxisomal membrane. Subsequently, the cargo is translocated into the peroxisomal lumen, whereas the receptor is released to the cytosol for further rounds of protein import. This cycle is controlled by the ubiquitination status of the receptor, which is best understood for the PTS1-receptor. While polyubiquitination of PTS-receptors results in their proteasomal degradation, the monoubiquitinated PTS-receptors are exported to the cytosol and recycled for further rounds of protein import. Here, we describe the identification of two ubiquitination cascades acting on the PTS2 co-receptor Pex18p. Using in vivo and in vitro approaches, we demonstrate that the polyubiquitination of Pex18p requires the ubiquitin-conjugating enzyme (E2) Ubc4p, which cooperates with the RING (really interesting new gene)-type ubiquitin-protein ligases (E3) Pex2p as well as Pex10p. Monoubiquitination of Pex18p depends on the E2 enzyme Pex4p (Ubc10p), which functions in concert with the E3 enzymes Pex12p and Pex10p. Our findings for the PTS2-pathway complement the data on PTS1-receptor ubiquitination and add up to a unified concept of the ubiquitin-based regulation of peroxisomal import.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitination , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peroxins , Peroxisomal Targeting Signal 2 Receptor , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
J Cell Sci ; 126(Pt 11): 2480-92, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23572515

ABSTRACT

The intracellular domain of the amyloid precursor protein (AICD) is generated following cleavage of the precursor by the γ-secretase complex and is involved in membrane to nucleus signaling, for which the binding of AICD to the adapter protein FE65 is essential. Here we show that FE65 knockdown causes a downregulation of the protein Bloom syndrome protein (BLM) and the minichromosome maintenance (MCM) protein family and that elevated nuclear levels of FE65 result in stabilization of the BLM protein in nuclear mobile spheres. These spheres are able to grow and fuse, and potentially correspond to the nuclear domain 10. BLM plays a role in DNA replication and repair mechanisms and FE65 was also shown to play a role in DNA damage response in the cell. A set of proliferation assays in our work revealed that FE65 knockdown in HEK293T cells reduced cell replication. On the basis of these results, we hypothesize that nuclear FE65 levels (nuclear FE65/BLM containing spheres) may regulate cell cycle re-entry in neurons as a result of increased interaction of FE65 with BLM and/or an increase in MCM protein levels. Thus, FE65 interactions with BLM and MCM proteins may contribute to the neuronal cell cycle re-entry observed in brains affected by Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Cell Cycle , Cell Nucleus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , RecQ Helicases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Cell Line , Cell Nucleus/genetics , Gene Knockdown Techniques , Humans , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , RecQ Helicases/genetics
15.
Mol Cell Proteomics ; 12(1): 215-27, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115302

ABSTRACT

Filaminopathy is a subtype of myofibrillar myopathy caused by mutations in FLNC, the gene encoding filamin C, and histologically characterized by pathologic accumulation of several proteins within skeletal muscle fibers. With the aim to get new insights in aggregate composition, we collected aggregates and control tissue from skeletal muscle biopsies of six myofibrillar myopathy patients harboring three different FLNC mutations by laser microdissection and analyzed the samples by a label-free mass spectrometry approach. A total of 390 proteins were identified, and 31 of those showed significantly higher spectral indices in aggregates compared with patient controls with a ratio >1.8. These proteins included filamin C, other known myofibrillar myopathy associated proteins, and a striking number of filamin C binding partners. Across the patients the patterns were extremely homogeneous. Xin actin-binding repeat containing protein 2, heat shock protein 27, nebulin-related-anchoring protein, and Rab35 could be verified as new filaminopathy biomarker candidates. In addition, further experiments identified heat shock protein 27 and Xin actin-binding repeat containing protein 2 as novel filamin C interaction partners and we could show that Xin actin-binding repeat containing protein 2 and the known interaction partner Xin actin-binding repeat containing protein 1 simultaneously associate with filamin C. Ten proteins showed significant lower spectral indices in aggregate samples compared with patient controls (ratio <0.56) including M-band proteins myomesin-1 and myomesin-2. Proteomic findings were consistent with previous and novel immunolocalization data. Our findings suggest that aggregates in filaminopathy have a largely organized structure of proteins also interacting under physiological conditions. Different filamin C mutations seem to lead to almost identical aggregate compositions. The finding that filamin C was detected as highly abundant protein in aggregates in filaminopathy indicates that our proteomic approach may be suitable to identify new candidate genes among the many MFM patients with so far unknown mutation.


Subject(s)
Contractile Proteins/genetics , Microfilament Proteins/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/metabolism , Proteome/analysis , Adult , Biomarkers, Tumor/analysis , DNA-Binding Proteins/analysis , Female , Filamins , HSP27 Heat-Shock Proteins/analysis , Heat-Shock Proteins , Humans , LIM Domain Proteins/analysis , Male , Mass Spectrometry , Middle Aged , Molecular Chaperones , Muscle Proteins/analysis , Muscle, Skeletal/metabolism , Muscular Dystrophies/etiology , Muscular Dystrophies/genetics , Mutation , Nuclear Proteins/analysis , Proteomics , rab GTP-Binding Proteins/analysis
16.
Biochim Biophys Acta ; 1834(1): 387-94, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22902274

ABSTRACT

The ternary complex consisting of AICD/FE65/TIP60 is thought to play a role in gene expression and was suggested to have a crucial impact in Alzheimer's disease. AICD is the intracellular subdomain of the amyloid precursor protein (APP) and able to bind the adapter protein FE65 and the histone acetyltransferase TIP60 setting up a nuclear dot-like phenotype. Within this work we readdressed the generation of the complex as a function of its compartments. Subsequently, we studied the proteome of AFT expressing cells vs. controls and identified Stathmin1 significantly down-regulated in AFT cells. Stathmin1 functions as an important regulatory protein of microtubule dynamics and was found associated with neurofibrillary tangles in brains of Alzheimer's disease patients. We validated our results using an independent label-free mass spectrometry based method using the same cell culture model. In a reversal model with diminished APP expression, caused by simultaneous knock-down of all three members of the APP family, we further confirmed our results, as Stathmin1 was regulated in an opposite fashion. We hypothesize that AICD-dependent deregulation of Stathmin1 causes microtubule disorganization, which might play an important role for the pathophysiology of Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Down-Regulation , Histone Acetyltransferases/metabolism , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Stathmin/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Cell Line , Histone Acetyltransferases/genetics , Humans , Lysine Acetyltransferase 5 , Microtubules/genetics , Microtubules/metabolism , Multiprotein Complexes/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Protein Structure, Tertiary , Stathmin/genetics
17.
Mol Cell Proteomics ; 11(11): 1274-88, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22879628

ABSTRACT

Central hallmark of Alzheimer's disease are senile plaques mainly composed of ß-amyloid, which is a cleavage product of the amyloid precursor protein (APP). The physiological function of APP and its family members APLP1 and APLP2 is poorly understood. In order to fill this gap, we established a cell-culture based model with simultaneous knockdown of all members of the family. A comprehensive proteome study of the APP/APLP1/APLP2 knockdown cell lysates versus controls revealed significant protein abundance changes of more than 30 proteins. Targeted validation of selected candidates by immunoblotting supported the significant down-regulation of the methionine adenosyltransferase II, alpha (MAT2A) as well as of peroxiredoxin 4 in the knockdown cells. Moreover, MAT2A was significantly down-regulated at the mRNA level as well. MAT2A catalyzes the production of S-adenosylmethionine from methionine and ATP, which plays a pivotal role in the methylation of neurotransmitters, DNA, proteins, and lipids. MAT2A-dependent significant up-regulation of S-adenosylmethionine was also detectable in the knockdown cells compared with controls. Our results point to a role of the APP family proteins in cellular methylation mechanisms and fit to findings of disturbed S-adenosylmethionine levels in tissue and CSF of Alzheimer disease patients versus controls. Importantly, methylation plays a central role for neurotransmitter generation like acetylcholine pointing to a crucial relevance of our findings for Alzheimer's disease. In addition, we identified differential gene expression of BACE1 and PSEN1 in the knockdown cells, which is possibly a consequence of MAT2A deregulation and may indicate a self regulatory mechanism.


Subject(s)
Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/genetics , Gene Expression Regulation , Methionine Adenosyltransferase/metabolism , Presenilin-1/genetics , S-Adenosylmethionine/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Down-Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Models, Biological , Nerve Tissue Proteins/metabolism , Peroxiredoxins , Presenilin-1/metabolism , Proteome/metabolism , Reproducibility of Results , Staining and Labeling
18.
Cells ; 13(6)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38534351

ABSTRACT

Genome editing, notably CRISPR (cluster regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9), has revolutionized genetic engineering allowing for precise targeted modifications. This technique's combination with human induced pluripotent stem cells (hiPSCs) is a particularly valuable tool in cerebral organoid (CO) research. In this study, CRISPR/Cas9-generated fluorescently labeled hiPSCs exhibited no significant morphological or growth rate differences compared with unedited controls. However, genomic aberrations during gene editing necessitate efficient genome integrity assessment methods. Optical genome mapping, a high-resolution genome-wide technique, revealed genomic alterations, including chromosomal copy number gain and losses affecting numerous genes. Despite these genomic alterations, hiPSCs retain their pluripotency and capacity to generate COs without major phenotypic changes but one edited cell line showed potential neuroectodermal differentiation impairment. Thus, this study highlights optical genome mapping in assessing genome integrity in CRISPR/Cas9-edited hiPSCs emphasizing the need for comprehensive integration of genomic and morphological analysis to ensure the robustness of hiPSC-based models in cerebral organoid research.


Subject(s)
Gene Editing , Induced Pluripotent Stem Cells , Humans , Gene Editing/methods , CRISPR-Cas Systems , Induced Pluripotent Stem Cells/metabolism , Genomics , Brain , Chromosome Mapping
19.
Cell Rep ; 43(3): 113941, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38478523

ABSTRACT

Resting CD4 T cells resist productive HIV-1 infection. The HIV-2/simian immunodeficiency virus protein viral accessory protein X (Vpx) renders these cells permissive to infection, presumably by alleviating blocks at cytoplasmic reverse transcription and subsequent nuclear import of reverse-transcription/pre-integration complexes (RTC/PICs). Here, spatial analyses using quantitative virus imaging techniques reveal that HIV-1 capsids containing RTC/PICs are readily imported into the nucleus, recruit the host dependency factor CPSF6, and translocate to nuclear speckles in resting CD4 T cells. Reverse transcription, however, remains incomplete, impeding proviral integration and viral gene expression. Vpx or pharmacological inhibition of the deoxynucleotide triphosphohydrolase (dNTPase) activity of the restriction factor SAM domain and HD domain-containing protein 1 (SAMHD1) increases levels of nuclear reverse-transcribed cDNA and facilitates HIV-1 integration. Nuclear import and intranuclear transport of viral complexes therefore do not pose important blocks to HIV-1 in resting CD4 T cells, and the limitation to reverse transcription by SAMHD1's dNTPase activity constitutes the main pre-integration block to infection.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Monomeric GTP-Binding Proteins , Animals , Humans , HIV-1/genetics , CD4-Positive T-Lymphocytes/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , HIV-2/genetics , Viral Regulatory and Accessory Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , HEK293 Cells
20.
BMC Evol Biol ; 13: 242, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24195737

ABSTRACT

BACKGROUND: Pseudogenes are traditionally considered "dead" genes, therefore lacking biological functions. This view has however been challenged during the last decade. This is the case of the Protein phosphatase 1 regulatory subunit 2 (PPP1R2) or inhibitor-2 gene family, for which several incomplete copies exist scattered throughout the genome. RESULTS: In this study, the pseudogenization process of PPP1R2 was analyzed. Ten PPP1R2-related pseudogenes (PPP1R2P1-P10), highly similar to PPP1R2, were retrieved from the human genome assembly present in the databases. The phylogenetic analysis of mammalian PPP1R2 and related pseudogenes suggested that PPP1R2P7 and PPP1R2P9 retroposons appeared before the great mammalian radiation, while the remaining pseudogenes are primate-specific and retroposed at different times during Primate evolution. Although considered inactive, four of these pseudogenes seem to be transcribed and possibly possess biological functions. Given the role of PPP1R2 in sperm motility, the presence of these proteins was assessed in human sperm, and two PPP1R2-related proteins were detected, PPP1R2P3 and PPP1R2P9. Signatures of negative and positive selection were also detected in PPP1R2P9, further suggesting a role as a functional protein. CONCLUSIONS: The results show that contrary to initial observations PPP1R2-related pseudogenes are not simple bystanders of the evolutionary process but may rather be at the origin of genes with novel functions.


Subject(s)
Evolution, Molecular , Genome, Human , Phylogeny , Protein Phosphatase 1/genetics , Pseudogenes , Animals , Genome , Humans , Male , Mammals/genetics , Retroelements , Sperm Motility
SELECTION OF CITATIONS
SEARCH DETAIL