• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 3
  • 1
  • Tagged with
  • 8
  • 4
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 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.
1

Biochemical Investigation of Progeroid Disease-Associated Mutations in Human ZMPSTE24

Erh-Ting Hsu (5929769) 17 January 2019 (has links)
<p>ZMPSTE24 is a unique intramembrane zinc metalloprotease that plays critical roles in the lamin A maturation pathway. Lamin A comprises a dense network underlying the inner nuclear membrane that maintains the structural integrity and proper function of the nucleus. The precursor of lamin A, prelamin A, terminates with a CAAX motif, where “C” is cysteine, “A” is typically an aliphatic amino acid, and “X” is one of several different amino acids. Like all CAAX proteins, prelamin A undergoes a series of post-translational modifications, including farnesylation of the cysteine by farnesyltransferase, endoproteolysis of the AAX residues by ZMPSTE24 or possibly a related protease RCE1, and carboxyl methylation by isoprenylcysteine carboxyl methyltransferase (ICMT). After CAAX processing, an additional cleavage event by ZMPSTE24 occurs to remove 15 residues from the C-terminus, including the farnasylated and carboxyl methylated cysteine, releasing mature lamin A into the nucleoplasm. Mutations in the gene encoding ZMPSTE24 that impair proteolytic activity cause a set of progeroid diseases, including B-type mandibuloacral dysplasia (MAD-B) and restrictive dermopathy (RD). Recently, ZMPSTE24 mutations were also detected in patients with metabolic syndrome (MS) and nonalcoholic fatty liver disease (NAFLD). Patients with these diseases have shown defective prelamin A processing, leading to the accumulation of persistently farnesylated prelamin A in the nucleus. However, how this accumulation causes disease remains unclear. We demonstrated that both ZMPSTE24 and another known CAAX protease, RCE1 are both capable of mediating the C-terminal cleavage of prelamin A. RCE1 retains the capacity to cleave the AAX residues of prelamin A in progeroid diseases induced by inactive ZMPSTE24 mutants, therefore the disease molecule will be most likely farnesylated and methylated prelamin A. These factors suggest that the ability of ZMPSTE24 to perform the upstream cleavage determines the accumulation level of uncleaved prelamin A in progeroid diseases. However, there was no available assay that could quantitatively demonstrate the <i>in vitro</i> upstream cleavage activity of ZMPSTE24. Therefore, we first developed a FRET-based assay that was able to precisely quantify the upstream cleavage activity of Ste24, the yeast homolog of ZMPSTE24. The 33-mer analog of <b>a</b>-factor, Ste24 natural substrate, has a 2-aminobenzoic acid (Abz) fluorophore at the N-terminus and a dinitophenol (Dnp) quencher located on the either side of the proposed cleavage site. After cleavage, quantification of the fluorescence from the dequenced peptide enabled us to continuously monitor the upstream cleavage activity of Ste24. We then utilized this FRET-based assay to examine the upstream cleavage activity of wild-type ZMPSTE24 and its disease mutants. We demonstrated that the <b>a</b>-factor analog FRET substrate could be recognized and cleaved at the predicted position by ZMPSTE24. Disease variants were examined using this assay and the results revealed reduced upstream cleavage activity. Moreover, blocking ubiquitylation restored catalytic activity of some ZMPSTE24 disease variants, suggesting that diminished activity of these mutants is due to protein instability. Limited trypsin digestion results also indicated that some variants may not be properly folded, as compared to wild-type. The crystal structure of ZMPSTE24 revealed that seven transmembrane helices of ZMPSTE24 surround a large intramembrane chamber. The HEXXH zinc metalloprotease motif faces inward to cap the top of the chamber inside which proteolysis is proposed to occur. The four side portals apparent in the structure may provide substrate entry and exit routes. Based on structural considerations, besides interfering with structural integrity, these disease mutations may decrease ZMPSTE24 activity by preventing substrate binding in the active site or occluding substrate entry into or exit from the enzyme chamber. Using a yeast <b>a</b>-factor sequence-based photoactive analog containing benzophenone in the farnesyl portion, which can be well processed by ZMPSTE24, we have shown that certain disease mutants may affect farnesyl binding of the C-terminal cleavage substrate. We also designed several double cysteine mutants near portal 1 in Cys-less background of Ste24. We then utilized bismaleimide sulfhydryl-to-sulfhydryl crosslinkers to block the portal opening. Some of the crosslinked double mutants showed reduced AAX cleavage activity, suggesting the portal 1 may be important for the C-terminal cleavage. Together, these data will clarify how the enzyme functions and also provide further insights into progeroid diseases.</p>
2

Complications cardiovasculaires liées aux défauts de maturation de la lamine A : Rôle des traitements antirétroviraux et des mutations LMNA / Cardiovascular complications linked to altered lamin A maturation : role of antiretroviral treatments and LMNA mutations

Afonso, Pauline 23 September 2015 (has links)
Les patients lipodystrophiques porteurs de mutations du gène LMNA codant pour les lamines de type A, et les patients infectés par le VIH (Virus de l’Immunodéficience Humaine) traités par des antirétroviraux (ARV) sont à risque de développer une athérosclérose précoce avec calcifications et des comorbidités liées au vieillissement. Pour mieux comprendre la physiopathologie des atteintes vasculaires, j’ai étudié l’impact d’ARV ou de mutations LMNA sur des cellules musculaires lisses (CML) ou endothéliales d’artères coronaires humaines in vitro. Ce travail a révélé que certains ARV (les inhibiteurs de protéase lopinavir ou atazanavir associés au ritonavir) induisent une sénescence prématurée et des dysfonctions des cellules endothéliales, alors que d’autres ont peu ou pas de conséquences (maraviroc, dolutégravir, maraviroc/dolutégravir et darunavir/ritonavir). De plus, le lopinavir/ritonavir ou l’atazanavir/ritonavir, ou l’expression des mutations LMNA p.R482W, p.D47Y ou p.R133L induisent une sénescence prématurée, une transdifférenciation ostéogénique avec calcification, et un stress oxydant dans les CML. L’accumulation de prélamine A farnésylée (une lamine A immature) et la diminution de l’expression de ZMPSTE24, son enzyme de maturation, sont, au moins en partie, responsables de ces effets.Ainsi, les ARV étudiés ont des impacts différents et agissent par des mécanismes physiopathologiques pro-athérogéniques en partie communs avec certaines mutations LMNA associées aux lipodystrophies. Le rôle majeur de l’accumulation de prélamine A farnésylée liée à une diminution d’expression de ZMPSTE24 ouvrent de nouvelles perspectives thérapeutiques. / Patients with lipodystrophies dues to mutation in the LMNA gene encoding A-type lamins, or HIV-infected patients (Human Immunodeficiency Virus) receiving antiretroviral therapy (ARV) are prone to develop early atherosclerosis and vascular calcifications associated with comorbidities linked to premature aging. Our study focused on the impact of LMNA mutations or antiretroviral treatment in vitro on vascular smooth muscle cells (VSMC) or endothelial cells from human coronary arteries. The results obtained during my thesis showed that some ARV (the protease inhibitors lopinavir or atazanavir associated with ritonavir) induce a cellular premature senescence with associated dysfunctions in endothelial cells, whereas others have little or no consequences (maraviroc, dolutegravir, maraviroc/dolutegravir and darunavir/ritonavir). In addition, some ARV (lopinavir or atazanavir with ritonavir) or the expression of LMNA mutations p.R482W, p.D47Y or p.R133L induce premature senescence, osteogenic transdifferentiation with calcification and oxidative stress of VSMC. Our results reveal that the accumulation of farnesylated prelamin A (an immature lamin A) and the decreased expression of its processing enzyme ZMPSTE24 are, at least partly, responsible for these effects.This work shows the different effects of ARVs and highlights the existence of common pro-atherogenic pathophysiological mechanisms in HIV-infected patients receiving some protease inhibitors and in lipodystrophic patients with LMNA mutations, initiated by an accumulation of farnesylated prelamin A related to a decrease expression of ZMPSTE24. These abnormalities could give rise to new therapeutic perspectives.
3

A-type Lamins in Cell Cycle Regulation

Parman-Ryans, Jaime L 01 May 2017 (has links)
Proteins of the nuclear lamina provide structural support to the nuclear envelope and participate in a variety of cellular functions, such as chromatin organization and transcriptional regulation. One of these proteins, Lamin A (72kDa), is synthesized as a 74 kDa precursor protein, Prelamin A, which undergoes an unusual maturation pathway that requires two farnesylation-dependent endoproteolytic cleavages. The second cleavage is unique to lamin A in higher vertebrates and is specifically carried out by the endoprotease zmpste24. Although most previous studies have focused mainly on the function of mature Lamin A, recent evidence from our laboratory shows important biological functions for Prelamin A as well. Prelamin A concentration in proliferating cells is very low or undetectable. Conversely, during quiescence induced by mitogen withdrawal or contact inhibition, Prelamin A levels increase dramatically. These variations are directly regulated by changes in expression and enzymatic activity of zmpste24. The central hypothesis of this dissertation is that full-length farnesylated and carboxymethylated prelamin A (FC-PreA) antagonizes both proliferation and apoptosis, therefore playing a role in cellular quiescence/senescence. To accomplish this goal, we studied the transcriptional regulation of zmpste24 and the interaction of FC-preA with proteins that participate in cell cycle control. 1) We identified and characterized a functional site for the E2F1 transcription factor (involved in the control of cell cycle) in the proximal 5’ UTR region of zmpste24. 2) By using proximity-labeling and co-immunoprecipitation-mass spectrometry techniques, we identified a set of proteins that interact preferentially with L467R-Prelamin A (uncleavable mutant) but not with mature Lamin A. Many of these proteins function to regulate progression through cell cycle.
4

Prelamin A Endoproteolytic Processing in Vitro by Recombinant zmpste24

Corrigan, Douglas P., Kuszczak, Danuta, Rusinol, Antonio E., Thewke, Douglas P., Hrycyna, Christine A., Michaelis, Susan, Sinensky, Michael S. 01 April 2005 (has links)
The nuclear lamins form a karyoskeleton providing structural rigidity to the nucleus. One member of the lamin family, lamin A, is first synthesized as a 74 kDa precursor, prelamin A. After the endopeptidase and methylation reactions which occur after farnesylation of the CAAX-box cysteine, there is a second endoproteolysis that occurs 15 amino acids upstream from the C-terminal farnesylated cysteine residue. Studies with knockout mice have implicated the enzyme Zmpste24 (Face-1) as a suitable candidate to perform one or both of these proteolytic reactions. Evidence has been presented elsewhere establishing that Zmpste24 possesses a zinc-dependent CAAX endopeptidase activity. In the present study, we confirm this CAAX endopeptidase activity with recombinant, membrane-reconstituted Zmpste24 and show that it can accept a prelamin A farnesylated tetrapeptide as substrate. To monitor the second upstream endoproteolytic cleavage of prelamin A, we expressed a 33 kDa prelamin A C-terminal tail in insect cells. We demonstrate that this purified substrate possesses a C-terminal farnesylated and carboxyl-methylated cysteine and, therefore, constitutes a valid substrate for assaying the second endoproteolytic step in lamin A maturation. With this substrate, we demonstrate that insect cell membranes bearing recombinant Zmpste24 can also catalyse the second upstream endoproteolytic cleavage.
5

Role of Zmpste24 in Prelamin A Maturation.

Corrigan, Douglas Paul 16 August 2005 (has links) (PDF)
The nuclear lamins form a karyoskeleton providing structural rigidity to the nucleus. One member of the lamin family, lamin A, is first synthesized as a 74 kDa precursor, prelamin A. Following the endopeptidase and methylation reactions which occur after farnesylation of the CAAX-box cysteine, there is a second endoproteolysis that occurs 15 amino acids upstream from the C-terminal farnesylated cysteine residue. Studies with knockout mice have implicated the enzyme Zmpste24 as a candidate to carry out one or both of these proteolytic reactions. In this body of work, the CAAX endopeptidase activity of recombinant, membrane reconstituted, Zmpste24 is demonstrated using a prelamin A farnesylated tetrapeptide as substrate. To monitor the second upstream endoproteolytic cleavage a 33 kDa prelamin A carboxyl terminal tail of prelamin A was expressed in insect cells. This purified substrate possesses a fully processed CAAX box, and, therefore, constitutes a valid substrate for assaying the second endoproteolytic step in lamin A maturation. In vitro reactions with this substrate and insect cell membranes bearing recombinant Zmpste24 demonstrate that Zmpste24 may possess the ability to directly catalyze the second endoproteolytic cleavage. Previous studies on nuclear envelope fractions have ascribed this second activity to a chymotrypsin like protease. However, Zmpste24 contains the canonical HEXXH domain, a common characteristic of zinc metalloproteinases. Experiments on Zmpste24 in this work demonstrate that inactivating the HEXXH domain by site directed mutagenesis results in a loss of the first endoproteolysis reaction, while the second endoproteolytic activity is retained. Supporting these data is the observation that a truncated mutant of Zmpste24 (residues: Met1 - Pro230) that does not contain the HEXXH motif, loses the first endoproteolytic activity while retaining the second. Furthermore, this second activity is not sensitive to the metalloproteinase inhibitors EDTA and 1,10-orthophenanthroline, but is sensitive to the chymotrypsin inhibitor TPCK and its fluorescent analogue, FFCK. The fact that Zmpste24 can be affinity labeled with FFCK suggests that this second activity is directly caused by a second, yet unidentified, active site with a chymotrypsin-like catalytic mechanism.
6

Involvement of Xeroderma Pigmentosum Group A (XPA) in Progeria Arising From Defective Maturation of Prelamin A

Liu, Yiyong, Wang, Youjie, Rusinol, Antonio E., Sinensky, Michael S., Liu, Ji, Shell, Steven M., Zou, Yue 01 February 2008 (has links)
Cellular accumulation of DNA damage has been widely implicated in cellular senescence, aging, and premature aging. In Hutchinson-Gilford progeria syndrome (HGPS) and restrictive dermopathy (RD), premature aging is linked to accumulation of DNA double-strand breaks (DSBs), which results in genome instability. However, how DSBs accumulate in cells despite the presence of intact DNA repair proteins remains unknown. Here we report that the recruitment of DSB repair factors Rad50 and Rad51 to the DSB sites, as marked by γ-H2AX, was impaired in human HGPS and Zmpste24-deficient cells. Consistently, the progeria-associated DSBs appeared to be unrepairable although DSBs induced by camptothecin were efficiently removed in the progeroid cells. We also found that these progeroid cells exhibited nuclear foci of xeroderma pigmentosum group A (XPA), a unique nucleotide excision repair protein. Strikingly, these XPA foci colocalized with the DSB sites in the progeroid cells. This XPA-DSB association was further confirmed and found to be mediated by DNA, using a modified chromatin immunoprecipitation assay and coimmunoprecipitation. RNA interference (RNAi) knockdown of XPA in HGPS cells partially restored DSB repair as evidenced by Western blot analysis, immunofluorescence and comet assays. We propose that the uncharacteristic localization of XPA to or near DSBs inhibits DSB repair, thereby contributing to the premature aging phenotypes observed in progeria arising from genetic defects in prelamin A maturation.
7

<b>ELUCIDATION OF THE INITIAL SUBSTRATE ACCESS PATHWAY IN STE24, YEAST HOMOLOG OF ZMPSTE24</b>

Eric Leonard Glasser (17605800) 11 December 2023 (has links)
<p dir="ltr">Premature aging disorders such as mandibuloacral dysplasia disorder (MAD) can be caused by improper maturation of nuclear scaffolding protein lamin A from its precursor prelamin A. ZMPSTE24 is responsible for both the earlier C-terminal CAAX cleavage and a subsequent N-terminal upstream cleavage during the posttranslational processing of prelamin A to lamin A. Although ZMPSTE24’s structure and function are well characterized, the role of the 4 apparent openings into its hollow inner chamber remains unknown. We hypothesize that one of these entrances, portal 1, is the initial substrate entry point based on its proximity to the zinc-coordinating active site. Unfortunately, ZMPSTE24 is difficult to express and purify. Fortunately, the yeast homolog, Ste24, not only shares many structural and functional similarities to ZMPSTE24 but is also much easier to express and purify in an active state. Therefore, we will use Ste24 and its substrate <b>a</b>-factor as a model system for ZMPSTE24 and its substrate prelamin A to deduce whether portal 1 acts as the primary substrate entry point. We examined portal 1’s function in primary substrate entry by observing how the incubation of portal 1 mutants engineered with cysteine residues around the portal with cysteine-reactive bismaleimide crosslinkers affects the activity of the C-terminal CAAX cleavage. If crosslinking of the cysteine residues occludes the portal, we hypothesize that activity will decrease because substrate cannot enter. The cysteine-less enzyme Ste24 (QA), which cannot react with the crosslinkers, was engineered by mutagenesis to contain 1 or 2 new cysteines at specific positions around this portal. We hypothesize that portal 1 occlusion with cysteine reactive bismaleimide crosslinkers will inactivate the enzyme by preventing substrate entry. We monitored changes in CAAX cleavage activity with a radioactive endoprotease-coupled CAAX assay.</p><p dir="ltr">In crude membranes derived from yeast expressing QA Ste24, activity was not inhibited in the presence of either BMH or BMOE crosslinker. For the single cysteine-containing mutants M210C, T267C, I307C, and V311C, each crosslinker similarly decreased activity over 50%. For the double cysteine-containing mutants M210C-I307C, T267C-I307C, and T267C-V311C, we found between 20-60% decreased activity in the presence of the crosslinker which has a length most similar to the distance between the two cysteines. These results closely reflect previous data and further suggest that CAAX activity of the enzyme may be decreased due to the occlusion of the primary entry site, portal 1.</p><p dir="ltr">With purified QA Ste24, changes in activity were less apparent. Activity for purified QA was not decreased in the presence of either crosslinker. Single cysteine-containing mutants did not show decreased activity in the presence of either crosslinker. Unlike what was observed in crude membrane preparations, the double-cysteine containing mutants exhibited minimal decrease in activity in the presence of the crosslinker that has a length most similar to the distance between the two cysteines.</p><p dir="ltr">In crude membrane preparations, the cysteine-containing QA Ste24 mutants have diminished activity in the presence of crosslinkers. This may be due to the occlusion of the primary entry point, portal 1. However, we recognize the possibility that the decrease in activity was the result of the occlusion of the exit portal site. It is imperative that further experiments confirm that exit portal occlusion is not occurring. For purified cysteine-containing QA Ste24 mutants, the negligible decrease in activity suggests either that the portal 1 is not the primary substrate entry point or that the conditions of the assay were not optimized to generate inhibition. For example, the concentration of crosslinker was not sufficient in the presence of excess lipid and the reconstitution mixture sequestered the crosslinker. Further optimization of the reaction conditions is warranted. The cysteine-containing QA Ste24 mutants must be assessed for free thiols to determine how successful the reaction with the crosslinkers is. A more developed understanding of how all four portals function in the Ste24 CAAX processing of <b>a</b>-factor will be very insightful towards the mechanism of ZMPSTE24 in lamin A CAAX processing and may catalyze new targets of study for ZMPSTE24-related diseases like MAD-B.</p>
8

Hallermann-Streiff Syndrome: No Evidence for a Link to Laminopathies

Kortüm, F., Chyrek, M., Fuchs, S., Albrecht, B., Gillessen-Kaesbach, G., Mütze, U., Seemanova, E., Tinschert, S., Wieczorek, D., Rosenberger, G., Kutsche, K. 04 August 2020 (has links)
Hallermann-Streiff syndrome (HSS) is a rare inherited disorder characterized by malformations of the cranium and facial bones, congenital cataracts, microphthalmia, skin atrophy, hypotrichosis, proportionate short stature, teeth abnormalities, and a typical facial appearance with prominent forehead, small pointed nose, and micrognathia. The genetic cause of this developmental disorder is presently unknown. Here we describe 8 new patients with a phenotype of HSS. Individuals with HSS present with clinical features overlapping with some progeroid syndromes that belong to the laminopathies, such as Hutchinson-Gilford progeria syndrome (HGPS) and mandibuloacral dysplasia (MAD). HGPS is caused by de novo point mutations in the LMNA gene, coding for the nuclear lamina proteins lamin A and C. MAD with type A and B lipodystrophy are recessive disorders resulting from mutations in LMNA and ZMPSTE24 , respectively. ZMPSTE24 in addition to ICMT encode proteins involved in posttranslational processing of lamin A. We hypothesized that HSS is an allelic disorder to HGPS and MAD. As the nuclear shape is often irregular in patients with LMNA mutations, we first analyzed the nuclear morphology in skin fibroblasts of patients with HSS, but could not identify any abnormality. Sequencing of the genes LMNA, ZMPSTE24 and ICMT in the 8 patients with HSS revealed the heterozygous missense mutation c.1930C>T (p.R644C) in LMNA in 1 female. Extreme phenotypic diversity and low penetrance have been associated with the p.R644C mutation. In ZMPSTE24 and ICMT , no pathogenic sequence change was detected in patients with HSS. Together, we found no evidence that HSS is another laminopathy.

Page generated in 0.0418 seconds