• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 34
  • 2
  • 2
  • 1
  • Tagged with
  • 42
  • 42
  • 42
  • 13
  • 13
  • 9
  • 9
  • 8
  • 8
  • 8
  • 7
  • 7
  • 7
  • 7
  • 6
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
21

Nuclear pore membrane glycoprotein 210 as a new marker for epithelial cells

Olsson, Magnus January 2003 (has links)
<p>Epithelial cell polarisation is a prerequisite for the branching morphogenesis in several organs. Differential screening techniques were used to identify genes, which are upregulated during induction of epithelium in early kidney development. This investigation revealed two separate genes, Nuclear localising protein 1 (Nulp1), a previously undescribed gene with sequence characteristics of the basic helix-loop-helix transcription factor family, and glycoprotein 210 (gp210, POM210), an integral membrane protein constituent of the nuclear pore complex (NPC). Of these, gp210 was found to be upreglated during conversion of mesenchyme to epithelium. </p><p>The nuclear envelope, which demarcates the nuclear region in the eukaryotic cell, consists of an inner and an outer membrane that are fused at the locations for NPCs. These large macromolecular assemblages are tube like structures connecting the cytoplasmic and nuclear compartments of the cell. NPCs serve as the only conduits for exchange of molecular information between these cellular rooms. Electron microscopy techniques have revealed detailed information about the NPC architecture. A number of proteins (nucleoporins) have been characterised and embodied as components of the NPC structure. Active, energy dependent nucleocytoplasmic transport of RNAs and proteins is mediated by a group of soluble receptor proteins, collectively termed karyopherins. </p><p>Gp210 has been suggested to be important for nuclear pore formation. Nevertheless, our analyses showed a limited expression pattern of gp210, with its mRNA and protein largely confined to epithelial cells in the mouse embryo. Furthermore, in several cell lines, gp210 was undetectable. The expression pattern of gp210 was not synchronised with some other nucleoporins, indicating NPC heterogeneity. Characterisation of the structure of the human gp210 gene, including its promoter region, gave insight about possible cell-type specific gene regulatory mechanisms. </p><p>Regulation of molecular traffic between the nucleus and the cytoplasm leads to transcriptional control. Cell specific configuration of the NPC structure, due to diffential expression of gp210, could be involved in this control. Gp210 could be of importance for the development of epithelial cell polarisation.</p>
22

Nuclear pore membrane glycoprotein 210 as a new marker for epithelial cells

Olsson, Magnus January 2003 (has links)
Epithelial cell polarisation is a prerequisite for the branching morphogenesis in several organs. Differential screening techniques were used to identify genes, which are upregulated during induction of epithelium in early kidney development. This investigation revealed two separate genes, Nuclear localising protein 1 (Nulp1), a previously undescribed gene with sequence characteristics of the basic helix-loop-helix transcription factor family, and glycoprotein 210 (gp210, POM210), an integral membrane protein constituent of the nuclear pore complex (NPC). Of these, gp210 was found to be upreglated during conversion of mesenchyme to epithelium. The nuclear envelope, which demarcates the nuclear region in the eukaryotic cell, consists of an inner and an outer membrane that are fused at the locations for NPCs. These large macromolecular assemblages are tube like structures connecting the cytoplasmic and nuclear compartments of the cell. NPCs serve as the only conduits for exchange of molecular information between these cellular rooms. Electron microscopy techniques have revealed detailed information about the NPC architecture. A number of proteins (nucleoporins) have been characterised and embodied as components of the NPC structure. Active, energy dependent nucleocytoplasmic transport of RNAs and proteins is mediated by a group of soluble receptor proteins, collectively termed karyopherins. Gp210 has been suggested to be important for nuclear pore formation. Nevertheless, our analyses showed a limited expression pattern of gp210, with its mRNA and protein largely confined to epithelial cells in the mouse embryo. Furthermore, in several cell lines, gp210 was undetectable. The expression pattern of gp210 was not synchronised with some other nucleoporins, indicating NPC heterogeneity. Characterisation of the structure of the human gp210 gene, including its promoter region, gave insight about possible cell-type specific gene regulatory mechanisms. Regulation of molecular traffic between the nucleus and the cytoplasm leads to transcriptional control. Cell specific configuration of the NPC structure, due to diffential expression of gp210, could be involved in this control. Gp210 could be of importance for the development of epithelial cell polarisation.
23

Late-Onset Triple A Syndrome: A Risk of Overlooked or Delayed Diagnosis and Management

Salmaggi, Andrea, Zirilli, Lucia, Pantaleoni, Chiara, De Joanna, Gabriella, Del Sorbo, Francesca, Köhler, Katrin, Krumbholz, Manuela, Hübner, Angela, Rochira, Vincenzo 19 February 2014 (has links) (PDF)
Background/Aims: A 33-year-old man was referred for the first time to the Division of Neurology because of the presence and progression of neurological symptoms. Dysphagia, weakness, reduced tear production, and nasal speech were present. In order to point the attention of late-onset triple A syndrome we describe this case and review the literature. Methods: Hormonal and biochemical evaluation, Schirmer test, tilt test and genetic testing for AAAS gene mutations. Results: Late-onset triple A syndrome caused by a novel homozygous missense mutation in the AAAS gene (A167V in exon 6) was diagnosed at least 17 years after symptom onset. Conclusions: The association between typical signs and symptoms of triple A syndrome should suggest the diagnosis even if they manifest in adulthood. The diagnosis should be confirmed by Schirmer test, endocrine testing (both basal and dynamic), genetic analysis, and detailed gastroenterological and neurological evaluations. Awareness of the possible late onset of the disease and of diagnosis in adulthood is still poor among clinicians, the acquaintance with the disease is more common among pediatricians. The importance of an adequate multidisciplinary clinical approach, dynamic testing for early diagnosis of adrenal insufficiency and periodical reassessment of adrenal function are emphasized. / Dieser Beitrag ist mit Zustimmung des Rechteinhabers aufgrund einer (DFG-geförderten) Allianz- bzw. Nationallizenz frei zugänglich.
24

Role of ALADIN for Oxidative Stress Response and Microsomal Steroidogenesis in Human Adrenocortical Cells

Jühlen, Ramona 12 January 2016 (has links) (PDF)
Autosomal recessive triple A syndrome is caused by mutations in the AAAS gene encoding the protein ALADIN. The disorder manifests with the triad of adrenocorticotropin-resistant adrenal insufficiency, achalasia of the stomach cardia and impaired tear production (alacrima) in combination with progressive neurological impairment of the central, peripheral and autonomic nervous systems. ALADIN is part of the nuclear pore complex acting as a scaffold nucleoporin. In this work the role of ALADIN in the human adrenocortical tumour cell line NCI-H295R1 was investigated. These cells were engineered to either over-express or down-regulate AAAS by inducible stable transfection. Alterations in steroidogenic gene expression and functional consequences were determined. In addition, the role of ALADIN on cell viability and oxidative stress response was analysed. Using both the human adrenal NCI-H295R1-TR AAAS knock-down and over-expression models the potential impairment of the nuclear import of aprataxin, DNA ligase 1 and ferritin heavy chain 1 was investigated. For this YFP-specific vectors transiently transfected into the cell lines were employed. The findings indicate that AAAS knock-down induces a down-regulation of genes coding for type II microsomal cytochrome P450 hydroxylases CYP17A1 and CYP21A2 and their electron donor enzyme cytochrome P450 oxidoreductase, thereby decreasing biosynthesis of precursor metabolites required for glucocorticoid and androgen production. Furthermore I demonstrate that ALADIN deficiency leads to increased susceptibility to oxidative stress and alteration in redox homeostasis after paraquat treatment. Finally, I show significantly impaired nuclear import of DNA ligase 1, aprataxin and ferritin heavy chain 1 in ALADIN knock-down cells. I conclude that down-regulating ALADIN results in decreased oxidative stress response leading to alteration in steroidogenesis, highlighting the knock-down cell model as an important in vitro tool for studying the adrenal phenotype in triple A syndrome. In an approach to identify new interaction partners of ALADIN, co-immunoprecipitation followed by proteome analyses using mass spectrometry was conducted in a GFP-ALADIN over-expression model using the human adrenocortical tumor cell line NCI-H295R. These results were verified in co-immunoprecipitation assays of endogenous ALADIN using NCI-H295R wild-type cells. The results suggest a possible interaction between ALADIN and microsomal flavoprotein cytochrome P450 oxidoreductase and progesterone receptor membrane compartment 2. Co-localisation analyses of these findings were done using immunofluorescence. The data are suggestive for an involvement of ALADIN in the export of nuclear-encoded mitochondrial proteins. Regulation of adrenocortical steroidogenesis is complex and there is increasing evidence that oxidative stress due to ROS accumulation and mitochondria are significantly involved. Furthermore, there may be an important cross-talk between functional organelles comprising nucleus, ER and mitochondria which presumably involves lipid metabolism. The goal of this work was to elucidate the function of ALADIN for the cellular oxidative stress response and its possible consequences for adrenocortical steroidogenesis in triple A syndrome patients. / Mutationen im AAAS Gen verursachen die autosomal rezessive Krankheit Triple-A-Syndrom. AAAS kodiert das Nukleoporin ALADIN, welches Bestandteil des nukleären Porenkomplexes ist. Phänotypische Charakteristika des Triple-A-Syndroms sind Nebennierenrinden-Insuffizienz, Achalasie des unteren Speiseröhrenschließmuskels und eine fehlende Tränenproduktion (Alakrimie). Diese Symptome sind kombiniert mit progredienten neurologischen Störungen des zentralen, peripheren und autonomen Nervensystems. In dieser Arbeit wurde die Rolle von ALADIN in der humanen Karzinom-Zelllinie NCI-H295R1 untersucht. Diese Nebennierenrinden-Zellen wurden stabil transfiziert und mit einem induzierbaren Expressionssystem modifiziert, so dass sie AAAS entweder überexprimierten oder herunterregulierten. In NCI-H295R1-Zellen wurden Veränderungen der Genexpression von Enzymen der Steroidogenese und funktionelle Konsequenzen der Überexpression oder Herunterregulation von ALADIN gemessen. Des Weiteren wurde die Rolle von ALADIN auf die Zellviabilität und die Redox-Homöostase analysiert. ALADIN überexprimierende und herunterregulierte Zellen wurden verwendet, um die potentielle Behinderung des nukleären Imports von Proteinen zu untersuchen, welche den Zellkern gegen oxidativen Stress schützen (z.B. Aprataxin, DNA-Ligase 1 und Ferritin Heavy Chain 1). Dazu wurden YFP-spezifische Vektoren transient in diese Zellen gebracht. Mit den Ergebnissen dieser Arbeit wurde gezeigt, dass die Herunterregulation von AAAS eine Verminderung der Genexpression von CYP17A1 und CYP21A2 und deren Elektronendonor Cytochrom P450 Oxidoreduktase bewirken. Die Biosynthese der Vorläufermetabolite von Kortisol und Aldosteron ist in diesen Zellen ebenfalls vermindert. Des Weiteren zeigen die ALADIN-defizienten NCIH295R1-Zellen eine erhöhte Sensitivität gegenüber oxidativem Stress und eine veränderte Redox-Homöostase nach der Behandlung mit Paraquat. Darüber hinaus konnte in dieser Studie auch gezeigt werden, dass herunterregulierte ALADIN NCI-H295R1-Zellen einen verminderten Zellkernimport von Aprataxin, DNA-Ligase 1 und Ferritin heavy chain 1 besitzen. Aus diesen Ergebnissen kann geschlussfolgert werden, dass ALADIN-defiziente Nebennierenzellen eine verminderte Stressantwort auf oxidativen Stress besitzen; dies führt schlussendlich zu einer veränderten Steroidogenese. Das beschriebene ALADIN knock-down Modell in NCI-H295R1-Zellen ist ein wichtiges in vitro Werkzeug, um die Pathogenese der Nebennierenveränderungen im Triple-A-Syndrom zu erforschen. Neue Interaktionspartner von ALADIN wurden mit Hilfe von Co-Immunpräzipitation gefolgt von Proteom-Analysen durch Massenspektrometrie in einem GFP-ALADIN Überexpressionsmodell in NCI-H295R charakterisiert. Die Ergebnisse wurden durch Experimente auf endogenem Niveau in NCI-H295R-Wildtypzellen verifiziert. Mit diesen Daten wird in dieser Arbeit erstmals eine Interaktion zwischen ALADIN und dem Flavoprotein Cytochrom P450 Oxidoreduktase und Progesterone Receptor Membrane Compartment 2 nachgewiesen. Diese Ergebnisse wurden mit Co-Lokalisierungsanalysen durch Immunfluoreszenzfärbung von ALADIN und Cytochrome P450 Oxidoreduktase ergänzt. Außerdem gibt die Arbeit Hinweise darauf, dass ALADIN als Nukleoporin an dem nuklearen Export mitochondrialer Vorläuferproteine beteiligt ist. Die Regulation der Steroidogenese in der Nebennierenrinde ist komplex und es existieren zahlreiche Hinweise darauf, dass oxidativer Stress aufgrund der Ansammlung reaktiver Sauerstoffradikale und. dass die Mitochondrien involviert sind. Außerdem ist ein funktionelles Zusammenspiel verschiedener Organellen, darunter Nukleus, ER und Mitochondrien, von großer Bedeutung. Das Ziel dieser Arbeit war die Identifizierung der Funktion von ALADIN in der zellulären oxidativen Stressantwort und die möglichen Konsequenzen für die Steroidogenese in der Nebennierenrinden in Triple-A-Syndrom-Patienten.
25

Role of ALADIN for Oxidative Stress Response and Microsomal Steroidogenesis in Human Adrenocortical Cells

Jühlen, Ramona 12 November 2015 (has links)
Autosomal recessive triple A syndrome is caused by mutations in the AAAS gene encoding the protein ALADIN. The disorder manifests with the triad of adrenocorticotropin-resistant adrenal insufficiency, achalasia of the stomach cardia and impaired tear production (alacrima) in combination with progressive neurological impairment of the central, peripheral and autonomic nervous systems. ALADIN is part of the nuclear pore complex acting as a scaffold nucleoporin. In this work the role of ALADIN in the human adrenocortical tumour cell line NCI-H295R1 was investigated. These cells were engineered to either over-express or down-regulate AAAS by inducible stable transfection. Alterations in steroidogenic gene expression and functional consequences were determined. In addition, the role of ALADIN on cell viability and oxidative stress response was analysed. Using both the human adrenal NCI-H295R1-TR AAAS knock-down and over-expression models the potential impairment of the nuclear import of aprataxin, DNA ligase 1 and ferritin heavy chain 1 was investigated. For this YFP-specific vectors transiently transfected into the cell lines were employed. The findings indicate that AAAS knock-down induces a down-regulation of genes coding for type II microsomal cytochrome P450 hydroxylases CYP17A1 and CYP21A2 and their electron donor enzyme cytochrome P450 oxidoreductase, thereby decreasing biosynthesis of precursor metabolites required for glucocorticoid and androgen production. Furthermore I demonstrate that ALADIN deficiency leads to increased susceptibility to oxidative stress and alteration in redox homeostasis after paraquat treatment. Finally, I show significantly impaired nuclear import of DNA ligase 1, aprataxin and ferritin heavy chain 1 in ALADIN knock-down cells. I conclude that down-regulating ALADIN results in decreased oxidative stress response leading to alteration in steroidogenesis, highlighting the knock-down cell model as an important in vitro tool for studying the adrenal phenotype in triple A syndrome. In an approach to identify new interaction partners of ALADIN, co-immunoprecipitation followed by proteome analyses using mass spectrometry was conducted in a GFP-ALADIN over-expression model using the human adrenocortical tumor cell line NCI-H295R. These results were verified in co-immunoprecipitation assays of endogenous ALADIN using NCI-H295R wild-type cells. The results suggest a possible interaction between ALADIN and microsomal flavoprotein cytochrome P450 oxidoreductase and progesterone receptor membrane compartment 2. Co-localisation analyses of these findings were done using immunofluorescence. The data are suggestive for an involvement of ALADIN in the export of nuclear-encoded mitochondrial proteins. Regulation of adrenocortical steroidogenesis is complex and there is increasing evidence that oxidative stress due to ROS accumulation and mitochondria are significantly involved. Furthermore, there may be an important cross-talk between functional organelles comprising nucleus, ER and mitochondria which presumably involves lipid metabolism. The goal of this work was to elucidate the function of ALADIN for the cellular oxidative stress response and its possible consequences for adrenocortical steroidogenesis in triple A syndrome patients. / Mutationen im AAAS Gen verursachen die autosomal rezessive Krankheit Triple-A-Syndrom. AAAS kodiert das Nukleoporin ALADIN, welches Bestandteil des nukleären Porenkomplexes ist. Phänotypische Charakteristika des Triple-A-Syndroms sind Nebennierenrinden-Insuffizienz, Achalasie des unteren Speiseröhrenschließmuskels und eine fehlende Tränenproduktion (Alakrimie). Diese Symptome sind kombiniert mit progredienten neurologischen Störungen des zentralen, peripheren und autonomen Nervensystems. In dieser Arbeit wurde die Rolle von ALADIN in der humanen Karzinom-Zelllinie NCI-H295R1 untersucht. Diese Nebennierenrinden-Zellen wurden stabil transfiziert und mit einem induzierbaren Expressionssystem modifiziert, so dass sie AAAS entweder überexprimierten oder herunterregulierten. In NCI-H295R1-Zellen wurden Veränderungen der Genexpression von Enzymen der Steroidogenese und funktionelle Konsequenzen der Überexpression oder Herunterregulation von ALADIN gemessen. Des Weiteren wurde die Rolle von ALADIN auf die Zellviabilität und die Redox-Homöostase analysiert. ALADIN überexprimierende und herunterregulierte Zellen wurden verwendet, um die potentielle Behinderung des nukleären Imports von Proteinen zu untersuchen, welche den Zellkern gegen oxidativen Stress schützen (z.B. Aprataxin, DNA-Ligase 1 und Ferritin Heavy Chain 1). Dazu wurden YFP-spezifische Vektoren transient in diese Zellen gebracht. Mit den Ergebnissen dieser Arbeit wurde gezeigt, dass die Herunterregulation von AAAS eine Verminderung der Genexpression von CYP17A1 und CYP21A2 und deren Elektronendonor Cytochrom P450 Oxidoreduktase bewirken. Die Biosynthese der Vorläufermetabolite von Kortisol und Aldosteron ist in diesen Zellen ebenfalls vermindert. Des Weiteren zeigen die ALADIN-defizienten NCIH295R1-Zellen eine erhöhte Sensitivität gegenüber oxidativem Stress und eine veränderte Redox-Homöostase nach der Behandlung mit Paraquat. Darüber hinaus konnte in dieser Studie auch gezeigt werden, dass herunterregulierte ALADIN NCI-H295R1-Zellen einen verminderten Zellkernimport von Aprataxin, DNA-Ligase 1 und Ferritin heavy chain 1 besitzen. Aus diesen Ergebnissen kann geschlussfolgert werden, dass ALADIN-defiziente Nebennierenzellen eine verminderte Stressantwort auf oxidativen Stress besitzen; dies führt schlussendlich zu einer veränderten Steroidogenese. Das beschriebene ALADIN knock-down Modell in NCI-H295R1-Zellen ist ein wichtiges in vitro Werkzeug, um die Pathogenese der Nebennierenveränderungen im Triple-A-Syndrom zu erforschen. Neue Interaktionspartner von ALADIN wurden mit Hilfe von Co-Immunpräzipitation gefolgt von Proteom-Analysen durch Massenspektrometrie in einem GFP-ALADIN Überexpressionsmodell in NCI-H295R charakterisiert. Die Ergebnisse wurden durch Experimente auf endogenem Niveau in NCI-H295R-Wildtypzellen verifiziert. Mit diesen Daten wird in dieser Arbeit erstmals eine Interaktion zwischen ALADIN und dem Flavoprotein Cytochrom P450 Oxidoreduktase und Progesterone Receptor Membrane Compartment 2 nachgewiesen. Diese Ergebnisse wurden mit Co-Lokalisierungsanalysen durch Immunfluoreszenzfärbung von ALADIN und Cytochrome P450 Oxidoreduktase ergänzt. Außerdem gibt die Arbeit Hinweise darauf, dass ALADIN als Nukleoporin an dem nuklearen Export mitochondrialer Vorläuferproteine beteiligt ist. Die Regulation der Steroidogenese in der Nebennierenrinde ist komplex und es existieren zahlreiche Hinweise darauf, dass oxidativer Stress aufgrund der Ansammlung reaktiver Sauerstoffradikale und. dass die Mitochondrien involviert sind. Außerdem ist ein funktionelles Zusammenspiel verschiedener Organellen, darunter Nukleus, ER und Mitochondrien, von großer Bedeutung. Das Ziel dieser Arbeit war die Identifizierung der Funktion von ALADIN in der zellulären oxidativen Stressantwort und die möglichen Konsequenzen für die Steroidogenese in der Nebennierenrinden in Triple-A-Syndrom-Patienten.
26

The Human Rev Interacting Protein (hRIP) is Required for Rev Function and HIV-1 Replication: a Dissertation

Sánchez-Velar, Nuria 07 January 2005 (has links)
Retroviruses have evolved sophisticated mechanisms to ensure timely export of incompletely spliced viral messenger ribonucleic acids (mRNAs) for gene expression and for viral packaging. For example, the Human Immunodeficiency Virus type 1 (HIV-1) encodes the Rev regulatory protein, a sequence-specific RNA-binding protein that is responsible for the cytoplasmic accumulation of intron-containing viral mRNAs. The HIV-1 Rev protein contains an amino terminal (N-terminal) Arginine-Rich Motif (ARM) RNA-binding domain (RBD) and a carboxy terminal (C-terminal) leucine-rich activation domain which functions as a Nuclear Export Signal (NES). The Rev ARM interacts in a sequence-specific manner with a cis-acting viral RNA stem-loop structure, the Rev Responsive Element (RRE), located in all incompletely spliced viral mRNAs. This initial interaction is followed by the recruitment of additional Rev molecules to form a RiboNucleoProtein (RNP) complex involving the RRE and Rev molecules. The cytoplasmic accumulation of the Rev:RRE RNP complex is dependent on the interaction of Rev with key cellular cofactors. Rev activation domain mutants exhibit a trans-dominant negative phenotype, suggesting that this domain of Rev interacts with cellular proteins required for Rev function. Biochemical and genetic studies have identified several cellular proteins that bind to the activation domain of Rev and are therefore candidate cofactors for Rev function. Amongst these is the human Rev Interacting Protein [hRIP, 79], which is also known as the Rev/Rex activation domain-binding protein [Rab, 18]. hRIP was identified in a yeast two-hybrid assay with the HIV-1 Rev and its functionally equivalent Human T-cell Leukemia Virus type-1 (HTLV-1) Rex protein as baits. The interaction between hRIP and HIV-1 Rev is dependent on a functional Rev NES, as predicted for a bona fide Rev cellular cofactor, and the Nucleoporin-like (Nup-like) repeats in the C-terminus of hRIP (18, 79]. Additional genetic studies indicated that the interaction between hRIP and Rev is indirect and is most likely mediated by the cellular export receptor CRM1 (Chromosomal Region Maintenance 1) [1, 153]. A role for hRIP in Rev function or HIV-1 replication has remained elusive. The goal of this dissertation was to determine whether hRIP is required for Rev function and HIV-1 replication. We used two approaches, a dominant-negative mutant and RNA interference (RNAi), to ablate hRIP activity and analyzed Rev function and HIV-1 replication using standard assays. The results of this dissertation demonstrate that hRIP is required for Rev function and HIV-1 replication. We show that Rev function is inhibited upon ablation of hRIP activity by either a trans-dominant negative mutant or RNAL Furthermore, we find that depletion of endogenous hRIP by RNAi results in the loss of viral replication in human cell lines and primary human macrophages. Unexpectedly, in the absence of functional hRIP, RRE-containing viral RNAs accumulate in the nuclear periphery where hRIP is localized. Comparable ablation of hRIP activity did not affect the intracellular localization or trafficking of a variety of proteins or cellular poly (A+ mRNA, suggesting that the inhibition of Rev-directed RNA export is specific. In conclusion, the results of this dissertation demonstrate that hRIP is involved in the movement of Rev-directed RNAs from the nuclear periphery to the cytoplasm. Therefore, hRIP is required for Rev function and HIV-1 replication. The hRIP protein is not essential for the maintenance of cell viability and thus might represent a novel target for the development of antiviral agents for HIV-1.
27

Exploring Nuclear Pore Complexes: Unraveling Structural and Functional Insights through Super-Resolution Microscopy

Junod, Samuel, 0000-0002-4288-0240 12 1900 (has links)
The nuclear pore complex (NPC) is a pivotal subcellular structure governing nucleocytoplasmic transport through a selectively permeable barrier. Comprising approximately 30 distinct proteins, it includes FG-Nups with phenylalanine-glycine (FG) motifs and non-FG Nups forming the pore's scaffold. The selectively permeable barrier formed by FG-Nups enables the passive diffusion of small molecules and facilitates the transport of larger ones recognized by nuclear transport receptors (NTRs). Their roles are critical in regulating mRNA and pre-ribosome nuclear export and the nuclear import of transcription factors, underscoring their significance in cellular processes. However, studying NPCs remains challenging due to their structural complexity, heterogeneity, dynamic interactions, and inaccessibility within live cells. In this dissertation, three core questions were investigated to elucidate the structure and function of the NPC. First, the nuclear export dynamics of pre-ribosomal subunits revealed significantly higher transport efficiency compared to other large cargos. Through inhibition of nuclear transport receptor (NTR), CRM1, by small-molecule inhibitor, leptomycin B, we found a dose-dependent inhibition of CRM1s played a crucial role in pre-ribosome export efficiency. We confirmed these results through a series of controlled environments with both import and export NTRs. Our results suggest that cooperative NTR mechanisms may enhance the nucleocytoplasmic transport of not only pre-ribosomal subunits but other protein complexes as well. Second, we investigated the dynamic properties of the NPC’s selectivity barrier by altering the concentration of O-linked β-N-acetylglucosamine (O-GlcNAc) sites on nuclear pore proteins. Using small-molecule inhibitors of O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA) to decrease or increase NPC O-GlcNAcylation, respectively, we found a significant change in the overall 3D spatial density of NPC O-GlcNAc sites. Then, by applying the same OGT- and OGA-inhibited conditions, we found that NPC O-GlcNAcylation significantly impacted the nuclear export of mRNA, suggesting that NPC O-GlcNAcylation regulates mRNA’s passage through the NPC’s selective permeability barrier. Third, we examined the nuclear transport mechanism for intrinsically disordered proteins (IDPs). Our findings revealed that IDPs, unlike large folded proteins, can passively diffuse through NPCs independent of size, and their diffusion behaviors are differentiated by the content ratio of charged (Ch) and hydrophobic (Hy) amino acids. Thus, we proposed a Ch/Hy-ratio mechanism for IDP nucleocytoplasmic transport. In summary, comprehending the dynamic behavior of the NPC selectivity barrier and its involvement in mediating large transiting complexes and IDPs has provided valuable insights into the fundamental nucleocytoplasmic transport mechanism, emphasizing the NPC's crucial role in cellular health and function. / Biology
28

The function of Nup358 in nucleocytoplasmic transport / Die Funktion von Nup358 im nukleocytoplasmatischen Transport

Wälde, Sarah 23 August 2010 (has links)
No description available.
29

Analysis of CRM1- and Nup214- dependent nuclear export of proteins / Analyse des CRM1- und Nup214- abhängigen Kernexportes von Proteinen

Roloff, Stephanie 21 May 2012 (has links)
No description available.
30

Implication of the Nup133 subunit of nuclear pores in cell division and differentiation : partners and mechanisms. / Implication de la nucléoporine Nup133 dans la division et la différentiation cellulaire : partenaires et mécanismes.

Berto, Alessandro 11 December 2017 (has links)
Les complexes des pores nucléaires (NPCs) sont des assemblages protéiques ancrés dans l’enveloppe nucléaire (EN) qui permettent et régulent les échanges entre le cytoplasme et le noyau. Au-delà de leur fonction de transport, plusieurs sous unité des NPCs (les nucléoporines, Nups) jouent un rôle important dans d’autres processus cellulaire tels que la division cellulaire et la différenciation. Le complexe-Y, composé de 9 Nups distincte, dont Nup133, représente une sous unité structurale des NPCs. Cependant, une fraction de ce complexe est localisée aux kinétochores (KTs) durant la mitose, où il est requis pour la ségrégation des chromosomes. Cette localisation aux KTs dépend du complexe Ndc80 mais également de Cenp-F. Par ailleurs, l’interaction Nup133/Cenp-F s'effectue aussi au niveau de l'EN en prophase, ce qui permet le recrutement de la dynéine, étape requise pour l’ancrage des centrosomes à l’EN dans les cellules HeLa et pour la migration du noyau vers le centrosome dans les progéniteurs neuronaux de cerveau de rat. Des études développementales chez la souris ont précédemment identifié le mutant merm qui meurt en milieu de gestation (E10.5). Bien que la mutation merm entraine l’absence de Nup133, la prolifération des cellules souches embryonnaires (mESCs) dérivées de blastocystes merm (Nup133-/-) n’est pas altérée. Cependant l’absence de Nup133 altère la différenciation des mESCs, notamment en neurones post-mitotique. Ce projet de thèse visait à comprendre les mécanismes moléculaires par lesquels Nup133 contribue à la division et la différenciation cellulaire. Afin de caractériser le phénotype des mESCs Nup133-/-, nous avons utilisé deux protocoles de différenciation in vitro, vers une voie neuroectodermale ou mésoendodermale. Cette étude a montré que le nombre de cellules est fortement diminué chez le mutant Nup133-/- comparé aux cellules contrôles lors de la différenciation des mESCs. Cependant les mESCs Nup133-/- qui survivent montrent, comme les mESCs contrôles, une diminution de l’expression des marqueurs de pluripotence et une augmentation des marqueurs de différentiation. L’analyse par cytométrie de flux n’a pas révélé d'altération majeure dans la progression du cycle cellulaire mais à mis en évidence une augmentation de la mort cellulaire lors de la différenciation des mESCs Nup133-/-. Afin de déterminer les domaines de Nup133 requis pour la différenciation des mESCs, nous avons développé une stratégie de sauvetage en établissant des lignées mESCS Nup133-/- qui expriment de manière stable GFP-Nup133, différentes délétions de Nup133 qui n’altèrent pas sa localisation au NPCs (GFP-Nup133DN, DMid, et DC) ou la GFP seule comme contrôle. Des études fonctionnelles ont indiqué que le domaine N-ter (NTD) de Nup133 est requis pour la différenciation des mESCs.Le seul partenaire identifié de Nup133-NTD étant Cenp-F, j’ai décidé de déterminer si l’interaction Nup133/Cenp-F jouait un rôle dans la différenciation des mESCs. En collaboration avec l’équipe de R. Guerois, nous avons simulé in silico l’interaction de Nup133-NTD avec un peptide de Cenp-F que nous avions identifié dans des cribles en double hybride. Cette modélisaton nous a permis de concevoir des mutants affectant la surface d’interaction Nup133/Cenp-F. Nous avons montré que ces mutations empêchent la localisation de Cenp-F à l’EN sans altérer sa présence aux KTs. J’ai également utilisé la stratégie de sauvetage décrite plus haut, pour étudier un mutant de Nup133 qui empêche son interaction avec Cenp-F. Cette étude a montré que l’interaction Nup133/Cenp-F n'est pas requise pour la différenciation in vitro des mESCs. L’étude de Cenp-F a été complétée par la caractérisation d’une mutation de Cenp_F qui affecte sa localisation aux KTs. Nous avons montré que cette mutation altère l’interaction entre Cenp-F et Bub1 mais sans affecter celle avec Nup133. Cette étude a ainsi permis d'identifier Bub1 comme un partenaire direct de Cenp-F requis pour son ancrage aux KTs. / Nuclear pores complexes (NPCs) are macromolecular assemblies anchored in the nuclear envelope (NE) providing the gates that allow and regulate all exchanges between the nucleus and the cytoplasm. Beyond their function in transport, several NPC subunits, the nucleoporins (Nups), have been demonstrated to also play important roles in other cellular processes including cell division and differentiation.The Y-complex, composed of 9 distinct Nups, including Nup133, represents a major structural subunit stably bound to both the cytoplasmic and nuclear faces of the NPCs. Beyond its structural role at nuclear pores, the Y-complex localizes at kinetochores in mitosis, where it is required for chromosome segregation. This kinetochores localization relies on the Ndc80 complex, but also on Nup133/Cenp-F interaction. The Nup133/Cenp-F interaction also contributes to the recruitment of dynein to the NE, a process that is required for the correct centrosomes tethering at the NE in prophase HeLa and for the migration of the nucleus towards the centrosomes prior to mitotic entry in rat brain progenitor cells.Developmental studies previously identified the mouse merm mutant that dies in midgestation (E10.5). In collaboration with the team of E. Lacy, the team of V. Doye showed that the merm mutation leads to the absence of Nup133. Importantly this study further revealed that self-renewal is not impaired in embryonic stem cells (mESCs) derived from merm (Nup133-/-) blastocyst. However, the lack of Nup133 impairs mESC differentiation into postmitotic neurons. How Nup133 contributes to ESCs differentiation remains however unknown.This PhD project aimed at understanding the cellular mechanisms explaining Nup133 contribution to cell division and differentiation.To characterize Nup133-/- mESCs differentiation phenotype, we used two distinct in vitro differentiation protocols, towards either neuroectodermal or mesoendodermal fate. This study revealed that cell number was strongly decreased in Nup133-/- relative to WT in mESC differentiation. However, the few Nup133-/- mESCs that survived displayed, as WT mESCs, a decreased expression of pluripotency markers and acquired differentiated state based on marker expression. FACS analyses did not reveal any major alteration of cell cycle progression but showed increased cell death upon differentiation.To determine which domain of Nup133 is critical for mESC differentiation, we developed a “rescue strategy” using Nup133 alleles deleted for structurally defined domains. Therefore, we established Nup133-/- mESC lines stably expressing GFP-Nup133, GFP-Nup13-ΔN, different ΔC-ter domain (that did not impair the binding of Nup133 to the NPC) or GFP alone as control. Functional studies indicated that while full length and C-ter deleted Nup133 rescue Nup133-/- ESCs defect in differentiation, Nup133-ΔN does not.The only identified partner of Nup133-NTD is Cenp-F. In view of the role of Nup133-NTD in mESC differentiation, I decided to determine if Nup133/Cenp-F interaction contributes to mESC differentiation. In collaboration with R. Guerois' team we simulated in silico the interaction of Nup133-NTD with a short peptide of Cenp-F that we previously identified using yeast-2-hybrid (Y2H) screens. We could thereby design mutants affecting Nup133/Cenp-F contact and show that they prevent Cenp-F localization to the nuclear envelope without altering its kinetochore localization. I then used the “rescue strategy” described above to study a Nup133 mutant specifically impairing its interaction with Cenp-F. This analysis revealed that Nup133/Cenp-F interaction is dispensable for in vitro mESC differentiation. This study on Cenp-F was completed by the characterization of a mutation within an adjacent leucine zipper affecting Cenp-F targeting to kinetochores. We evidenced that this mutation impairs Cenp-F interaction with Bub1 but not with Nup133, identifying Bub1 as a direct kinetochore tether of Cenp-F.

Page generated in 0.0532 seconds