ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Publication Date: 2011-11-18
    Description: Abstract 2236 Lipoprotein(a) [Lp(a)] has been identified as an independent risk factor for cardiovascular diseases such as coronary heart disease. Lp(a) levels vary over 1000-fold within the human population and Lp(a) possesses both proatherogenic and prothrombotic properties due to the LDL-like moiety and apolipoprotein(a) [apo(a)] components, respectively. Apo(a) is highly homologous to plasminogen and thus can potentially interfere with plasminogen activation. Plasmin generated in the context of fibrin mediates the breakdown of blood clots, which are the causative factors in heart attacks and strokes. Plasmin generated on the surface of vascular cells plays a role in cell migration and proliferation, two of the fibroproliferative inflammatory events that underlie atherosclerosis. Previous studies have suggested that apo(a) may inhibit pericellular plasminogen activation on the basis of observations that apo(a) decreases plasminogen binding to cells. We have undertaken analysis of the mechanism by which apo(a) may interfere with pericellular plasminogen activation to allow for a more definitive description of the role of Lp(a) within the vasculature. Plasminogen activation was found to be markedly inhibited by the recombinant apo(a) variant 17K, in a dose dependent manner, on human umbilical vein endothelial cells (HUVECs), human monocytic leukemia cells (THP-1), THP-1 macrophages, and smooth muscle cells. The strong lysine binding site in kringle IV type 10, as well as kringle V appear to be required for this effect since apo(a) variants lacking these elements (17KΔAsp and 17KΔV, respectively) failed to inhibit activation. However, the role of lysine-dependent binding of apo(a) itself to the cells is not clear. Carboxypeptidase treatment of cells did not decrease apo(a) binding, and apo(a) does not compete directly for plasminogen binding to the cells. Rather, apo(a) and plasminogen may bind to the cells as a complex. We next attempted to identify the cell-surface receptor(s) that mediate plasminogen activation on the cell surface as well as its inhibition by apo(a). Urokinase-type plasminogen activator receptor (uPAR) has been previously shown to bind to urokinase-type plasminogen activator (uPA), vitronectin, and β3 integrins. uPAR is involved in the remodeling of the extracellular matrix (ECM) through regulation of plasminogen activation. We found evidence that uPAR is a potential receptor for both plasminogen and apo(a). Knockdown of uPAR in HUVECs results in decreased binding of plasminogen, 17K and, to a lesser extent, 17KΔAsp and 17KΔV. Similar experiments in SMCs revealed no changes in binding. A decrease in tPA-mediated plasminogen activation following uPAR knockdown occurred in HUVECs, and addition of 17K did not result in any further decrease. Overexpression of uPAR in THP-1 macrophages leads to greater than a two fold increase in 17K and plasminogen binding. Plasminogen activation increases over two-fold as a result of overexpression of uPAR, while 17K blunts the effect of uPAR overexpression. These results indicate that uPAR plays a crucial role in both plasminogen and apo(a) binding to the cell surface of specific cells and inhibition by apo(a) of plasminogen activation. Macrophage-1-antigen (Mac-1) receptor consists of CD11b (αM) and CD18 (β2) integrin and has been previously shown to recognize uPA and control migration and adhesion. Furthermore, αVβ3 has been previously shown to bind to vitronectin and the uPA-uPAR complex which promotes cell adhesion through binding of both vitronectin and αVβ3 integrins. We found that blocking the αM, β2, or αVβ3 receptors with monoclonal antibodies in THP-1 cells leads to a decrease in plasminogen activation, as well as a blunting of the inhibitory effects of apo(a) on plasminogen activation. These results indicate a role for Mac-1 and αVβ3 in apo(a) binding and inhibition of plasminogen activation. In conclusion, we have demonstrated, for the first time, the role of specific receptors in binding of apo(a) to vascular cell surfaces and in mediating the inhibitory effect of apo(a) on pericellular plasminogen activation. Disclosures: No relevant conflicts of interest to declare.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 2
    Publication Date: 2011-11-18
    Description: Abstract 1189 Thrombin-activatable fibrinolysis inhibitor (TAFI) is a basic carboxypeptidase zymogen that plays important roles in modulation of fibrinolysis and inflammation. Activated TAFI (TAFIa) removes carboxyl-terminal lysine and/or arginine residues from substrates such as partially-degraded fibrin, cell-surface plasminogen receptors, bradykinin, the anaphylatoxins C3a and C5a, and thrombin-cleaved osteopontin. The plasma pool of TAFI arises from expression of its gene (CPB2) in the liver. However, CPB2 is expressed in other locations including platelets (arising from expression in megakaryocytes), monocytes, and macrophages. An additional source of CPB2 expression has been shown to be the hippocampus; this TAFI variant was reported to be expressed from a CPB2 mRNA in which (i) exon 7 had been skipped resulting in an in-frame loss of 37 codons and (ii) alternative splicing had occurred in exon 11 resulting in a frameshift that deletes the final 42 codons and introduces a novel 16-amino acid carboxyl-terminus. Most recently, skipping of exon 7 has been reported in HepG2 (human hepatocellular carcinoma) cells, a phenomenon that appears to play a role in balancing selection at the CPB2 locus in the human population. As much as 12.5% of the CPB2 transcript in HepG2 cells was reported to lack exon 7. Accordingly, we have characterized, using RT-PCR, molecular cloning, and quantitative RT-PCR, the splicing patterns of CPB2 mRNA in a variety of cell types. We examined RNA isolated from human liver, HepG2 cells, the megakaryocytoid cell line Dami, platelets, the monocytoid cell line THP-1, and human cerebral cortex and cerebellum. We found evidence for alternative splicing/exon skipping in all cell types tested. All cells contained CPB2 mRNA lacking exon 7. Only platelets, cortex, and cerebellum CPB2 mRNA featured alternatively spliced exon 11, and all cDNA clones identified that contained exon 11 alternative splicing also lacked exon 7. Quantitative analysis of the proportion of total CPB2 transcripts that lack exon 7 showed that HepG2 cells had almost 10% exon 7-less transcripts but all other cell types tested had far lower proportions, ranging from 1% (Dami cells, peripheral blood mononuclear cells and cerebellum) to less than 0.1% (liver, THP-1 cells, platelets). Studies of CPB2 expressed in the hippocampus suggested that the variant lacking exon 7 and featuring alternative splicing in exon 11 encodes a protein that is localized in the endoplasmic reticulum of neural cells and that possesses endopeptidase activity against amyloid precursor protein. To test the functional properties of the TAFI proteins encoded by the TAFI variants, we transfected baby hamster kidney cells with expression plasmids encoding variants lacking exon 7, alternatively spliced exon 11, or both variations. Interestingly, unlike wild-type recombinant TAFI in these cells, the variant proteins could not be secreted, despite the presence of an intact signal peptide in each. Western blot analyses of transfected cell lysates revealed immunoreactive bands between 40 and 45 kDa, consistent with hypoglycosylated TAFI; lysates of cells expressing wild-type TAFI contained a 45 kDa species and a 60 kDa mature preproprotein. We therefore propose that the variant proteins are aberrantly folded and thus do not exit the ER. Notably, none of the variant proteins could be activated by thrombin-thrombomodulin and they did not show activity in a specific functional assay for TAFIa. Deletion of exon 7-encoded residues removes two surface α-helices and a single internal β-strand from the TAFI structure. Alternative splicing in exon 11 deletes a critical catalytic residue (Glu363). It is therefore not surprising that the variants are aberrantly folded, are not secretable, and lack TAFIa activity. It is also difficult to envisage how such a protein could acquire endopeptidase activity. We therefore speculate that variant TAFI resulting from exon skipping and alternative splicing may act as a chaperone for the presumptive peptidase that recognizes amyloid precursor protein. Moreover, full-length TAFI is expressed in the brain and may regulate brain-expressed tPA and plasminogen to influence neural function. Finally, it is possible that, under certain circumstances, the extent of exon skipping/alternative splicing is sufficient to impact the secretion of functional TAFI from liver or other cell types. Disclosures: No relevant conflicts of interest to declare.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 3
    Publication Date: 2008-11-16
    Description: Thrombin-activable fibrinolysis inhibitor (TAFI) is a carboxypeptidase B-like proenzyme that, once activated by thrombin, the thrombin-thrombomodulin complex, or plasmin, attenuates fibrinolysis. Aberrant regulation of the TAFI pathway alters the balance between coagulation and fibrinolysis and may underlie severe haemostatic disorders such as thrombosis and hemophilia. Indeed, high plasma TAFI levels have been associated with several cardiovascular diseases. It is important to note that there is a large inter-individual variability in plasma TAFI levels within the population, and this variation is primarily due to non-genetic factors. Therefore, variation in TAFI gene expression is a risk factor for thrombotic disorders and may be an important means by which TAFI responds to environmental and physiological stimuli. Novel associations between plasma TAFI levels and sex hormones have triggered interest in determining the role of TAFI as a mediator of the cardioprotective effects of estrogens and progestagens, or as a mediator of the increased thrombotic risk that accompanies use of oral contraceptives or hormone replacement therapy. Plasma TAFI concentrations rise with age in women but not in men, and are elevated in post-menopausal women compared to pre-menopausal women. In addition, plasma TAFI levels have been shown to be decreased by selective estrogen receptor modulators such as HMR 3339 and raloxifene, estradiol plus trimegestone, transdermal estradiol, and oral estradiol plus gestodene. On the other hand, some studies have reported minimal to no change in plasma TAFI levels occurring with the use of oral contraceptive, Raloxifene, or Tamoxifen. Paradoxically, it has been shown that both plasma TAFI levels and clot lysis time rise during pregnancy and then promptly return to basal levels after delivery. These studies illustrate the controversies surrounding the role of sex steroids in modulating plasma TAFI levels. In the present study, we have attempted to directly measure the effect of sex steroids on hepatic TAFI gene expression, and to uncover the molecular mechanisms underlying these regulatory events. HepG2 (human hepatocellular carcinoma) cells were cultured in the presence or absence of progesterone and b-estradiol and TAFI mRNA abundance was measured using real-time RT-PCR. We found that both of these hormones significantly decrease endogenous TAFI mRNA abundance in a dose-dependant manner. To assess the ability of these hormones to influence transcription of the gene encoding TAFI, we treated HepG2 cells that had been transiently transfected with luciferase reporter plasmids containing the 5′-flanking region of the TAFI gene. Interestingly, the change in promoter activity closely paralleled changes in mRNA abundance, suggesting that the effect of the hormones is mediated at the level of transcription. Furthermore, changes in TAFI mRNA abundance following treatments with estrogen were not associated with a decrease in TAFI mRNA stability when compared to the untreated control. TAFI protein levels were also decreased in a dose-dependent manner as assessed by western blot analysis. Inspection of the sequence of the TAFI 5′-flanking region does not show any consensus estrogen responsive elements, although we cannot exclude a role for more complex transcriptional system such as an estrogen response unit. The effect of estrogen could also be performed indirectly through the modulation of other transcription factors such SP-1 or members of the basal transcriptional machinery. We also investigated whether progesterone decreases TAFI gene expression via the binding of the progesterone receptor to the established glucocorticoid responsive element (GRE) within the TAFI promoter. Our results showed that progesterone generates the same decrease in promoter activity even when the GRE site was mutated, indicating that progesterone may act through a different site. In conclusion, our studies are beginning to reveal the molecular basis for the apparent relationship between female sex steroids and plasma concentrations of TAFI: specifically, a direct downregulatory effect on transcription of the gene encoding TAFI.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 4
    Publication Date: 2008-11-16
    Description: Thrombin-activable fibrinolysis inhibitor (TAFI) is a carboxypeptidase zymogen defining a pathway that functions as a molecular link between coagulation and fibrinolysis. Activation by thrombin, the thrombin-thrombomodulin complex, or plasmin, the resultant enzyme (TAFIa) affects the balance between these two cascades by attenuating positive feedback in the fibrinolytic cascade, thereby inhibiting fibrin clot lysis. Plasma TAFI antigen levels vary significantly between individuals, which has implicated TAFI as a risk factor for thrombotic diseases. TAFIa can also inactivate pro-inflammatory peptides such as the anaphylatoxins and bradykinin, suggesting a role for the TAFI pathway as a link between coagulation and inflammation. TAFI expression in cultured hepatic cells is decreased by interleukins −1 and −6, and plasma TAFI levels in human are decreased in experimental endotoxemia. Although the liver is the main source of plasma TAFI, TAFI has also been identified in platelets, and TAFI mRNA has been detected in the Dami (megakaryoblastic) cell line (but not the MEG-01 cell line). TAFI mRNA has also been detected in adipocytes of patients with type 2 diabetes; however, TAFI mRNA expression in human umbilical vein endothelial cells is still a point of controversy. It has been hypothesized that platelet TAFI arises from TAFI gene expression in megakaryocytes (MK). Using RT-PCR and real-time RT-PCR, we not only confirmed the presence of TAFI mRNA in Dami cells, but also found that TAFI mRNA abundance was increased throughout Dami cell differentiation along the megakaryocytes/platelet lineage (up to 8 fold increase after 48 hours) stimulated by phorbol myristate acetate (PMA) treatment. The quantitative real-time RT-PCR experiments revealed that TAFI mRNA is present in differentiated Dami cells at a level that is only one-hundredth of that observed in HepG2 (hepatoma) cells. Using transfection experiments with luciferase reporter plasmids containing progressive deletions of the human TAFI 5′-flanking region, we identified the sequence between −438 and −257 (relative to the initiator methionine codon) to be responsible for the enhanced TAFI gene transcription as Dami cells differentiate into more mature MK-like cells. Moreover, using western blot analysis, we detected TAFI protein expression in the medium of differentiated Dami cells, but not untreated Dami cells. Together, these data provide further evidence supporting the idea that platelet TAFI is generated from TAFI gene expression in megakaryocytes rather than by uptake from the plasma. To study TAFI gene regulation in monocytes and macrophages, RT-PCR and realtime RT-PCR were used to detected and quantify, respectively, TAFI mRNA expression in both THP-1 and THP-1 cells that have been differentiated into macrophage-like cells (THP-1ma) by PMA treatment. TAFI mRNA abundance was similar in THP-1 cells as what was observed in differentiated Dami cells. In addition, we found a progressive decrease in TAFI mRNA abundance throughout the THP-1 differentiation with an 85% decrease after 24 hours of PMA treatment. Transfection experiments using luciferase reporter plasmids representing progressive deletions of the human TAFI 5′-flanking region identified sequences between −151 and −121 as harboring key promoter elements for the differentiation-associated decrease in TAFI gene expression as THP-1 differentiate into macrophage-like cells. However, no TAFI protein was detected in either THP-1 or THP-1ma conditioned medium using western blot analyses. Nonetheless, extra-hepatic expression of TAFI, such as platelet, monocytes and macrophages, suggests novel roles for TAFI pathway beyond regulation of fibrin clot breakdown.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 5
    Publication Date: 2002-10-01
    Description: A decrease in light transmittance before clot formation, manifesting as a biphasic waveform (BPW) pattern in coagulation assays, was previously correlated with the onset of disseminated intravascular coagulation (DIC). In this study of 1187 consecutive admissions to the intensive care unit, the degree of this change on admission predicts DIC better than D-dimer measurements. Additionally, the BPW preceded the time of DIC diagnosis by 18 hours, on average, in 56% (203 of 362) of DIC patients. The BPW is due to the rapid formation of a precipitate and coincident turbidity change on recalcification of plasma. The isolated precipitate contains very-low–density lipoprotein (VLDL) and C-reactive protein (CRP). The addition of CRP and Ca++ to normal plasma also causes the precipitation of VLDL and IDL, but not LDL or HDL. The Kd of the CRP/VLDL interaction is 340 nM, and the IC50 for Ca++ is 5.0 mM. In 15 plasmas with the BPW, CRP was highly elevated (77-398 μg/mL), and the concentration of isolated VLDL ranged from 0.082 to 1.32 mM (cholesterol). The turbidity change on recalcification correlates well with the calculated level of the CRP–VLDL complex. Clinically, the BPW better predicts for DIC than either CRP or triglyceride alone. The complex may have pathophysiological implications because CRP can be detected in the VLDL fraction from sera of patients with the BPW, and the VLDL fraction has enhanced prothrombinase surface activity. The complex has been designated lipoprotein complexed C-reactive protein.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 6
  • 7
    Publication Date: 2008-01-01
    Description: Thrombin-activable fibrinolysis inhibitor (TAFI) is a plasma zymogen that acts as a molecular link between coagulation and fibrinolysis. Numerous single nucleotide polymorphisms (SNPs) have been identified in CPB2, the gene encoding TAFI, and are located in the 5′-flanking region, in the coding sequences, and in the 3′-untranslated region (UTR) of the CPB2 mRNA transcript. Associations between CPB2 SNPs and variation in plasma TAFI antigen concentrations have been described, but the identity of SNPs that are causally linked to this variation is not known. In the current study, we investigated the effect of the SNPs in the 5′-flanking region on CPB2 promoter activity and SNPs in the 3′-UTR on CPB2 mRNA stability. Whereas the 5′-flanking region SNPs (with 2 exceptions) did not have a significant effect on promoter activity, either alone or in haplotypic combinations seen in the human population, all of the 3′-UTR SNPs substantially affected mRNA stability. We speculate that these SNPs, in part, contribute to variation in plasma TAFI concentrations via modulation of CPB2 gene expression through an effect on mRNA stability.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...