201
|
Abrink M, Larsson E, Gobl A, Hellman L. Expression of lactoferrin in the kidney: implications for innate immunity and iron metabolism. Kidney Int 2000; 57:2004-10. [PMID: 10792619 DOI: 10.1046/j.1523-1755.2000.00050.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Sequestering of free iron by lactoferrin (LF) is important in the defense against bacteria. In a screening for LF expression in various organs, high levels of LF mRNA were detected in human kidney. This indicated that LF is produced by the kidney and that it may participate in innate immunity of this organ. METHODS AND RESULTS Antibody staining and in situ hybridization of paraffin-embedded kidney sections showed that LF is expressed in cells lining the distal collecting ducts of the medulla. High levels of both protein and mRNA were detected in these cells. However, a clear difference in the distribution of mRNA and protein within the tissue was observed. LF mRNA was detected along a relatively large portion of the tubuli, whereas LF antigen was found mainly in the very distal regions of the same tubuli. This indicates that LF is released by large regions of the tubuli and possibly reabsorbed in the most distal parts. Using enzyme-linked immunosorbent assay, only very low LF levels were detected in urine. CONCLUSION The present study shows that LF is produced by the kidney and that both LF mRNA and protein are distributed in a highly ordered fashion. This latter finding, together with the very low levels of LF detected in urine, indicates that LF may contribute to the immune defense in the kidney by reduction of available free iron in the urine. Other possibilities are that LF may play a role in the iron metabolism by recovering free iron from urine and making it available for metabolic use, and that LF may participate in the antioxidant defense systems protecting the kidney against nonmicrobial oxidative injury, that is, ischemia, reperfusion and inflammation.
Collapse
Affiliation(s)
- M Abrink
- Department of Cell and Molecular Biology, University of Uppsala, Sweden
| | | | | | | |
Collapse
|
202
|
Chan WL, Shaw PC, Tam SC, Jacobsen C, Gliemann J, Nielsen MS. Trichosanthin interacts with and enters cells via LDL receptor family members. Biochem Biophys Res Commun 2000; 270:453-7. [PMID: 10753646 DOI: 10.1006/bbrc.2000.2441] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The type-I ribosome-inactivating protein trichosanthin displays selective cytotoxicity, suggesting specific mechanisms for entry into cells. Here we show that trichosanthin binds specifically to the endocytic receptors LRP and megalin, and that binding as well as uptake into cells is inhibited by the receptor-associated protein (RAP). The results suggest that the known abortifacient and renotoxic actions of trichosanthin are caused by LRP-mediated uptake in trophoblasts and megalin-mediated uptake in proximal tubule epithelial cells, respectively.
Collapse
Affiliation(s)
- W L Chan
- Department of Physiology, Department of Biochemistry, Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
203
|
Daugherty AL, McKee ML, FitzGerald DJ, Mrsny RJ. Epithelial application of Pseudomonas aeruginosa exotoxin A results in a selective targeting to cells in the liver, spleen and lymph node. J Control Release 2000; 65:297-302. [PMID: 10699289 DOI: 10.1016/s0168-3659(99)00251-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Pseudomonas aeruginosa exotoxin A (PE) is a 67-kDa protein expressed under the selective pressure of a low iron environment. Previous studies using non-toxic PE chimeras containing a viral surface antigen, the V3 loop of MN gp120 from human immunodeficiency virus type 1 (HIV-1), resulted in not only an effective mucosal immunization but also a striking systemic immune response following epithelial application. Presently, we have examined the possibility that such a strong dual immune response was generated by the efficient targeting of critical cells of the immune system. Mice were dosed with 10 microg of toxic PE or a non-toxic mutant of PE (ntPE) by intratracheal instillation. Examination of lung, liver and spleen tissues isolated 4, 8 and 12 h following intratracheal instillation with PE demonstrated specific cell damage in these tissues which was not observed in mice dosed with ntPE. Based upon the location and characteristics of observed responses, the cells targeted by PE appear to be involved in the antigen presentation arm of the immune response. Since ntPE chimeras with inserted peptide antigen epitopes from a wide variety of pathogens are easy to prepare and administer, these results support this approach for mucosal immunization.
Collapse
Affiliation(s)
- A L Daugherty
- Drug Delivery/Biology Group, Department of Pharmaceutical Research and Development, Genentech Inc., MS #6, 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | | | | | | |
Collapse
|
204
|
Celli S, Marto JA, Falchetto R, Shabanowitz J, Valdivia LA, Fung JJ, Hunt DF, Kelly RH. Serum protein immunogenicity: implications for liver xenografting. Electrophoresis 2000; 21:965-75. [PMID: 10768783 DOI: 10.1002/(sici)1522-2683(20000301)21:5<965::aid-elps965>3.0.co;2-d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The objectives of this study were threefold: (i) assess immunogenicity of donor plasma proteins following hepatic xenotransplantation, (ii) identify potential immunogens, and (iii) consider the implications of antibody formation against these plasma proteins in xenograft survival. We studied liver and heart xenografts in a concordant combination, hamster to rat. All grafts were examined at necropsy for evidence of rat immunoglobulin G (IgG) deposition. Cardiac xenografts were placed in recipients who had, or had not, been sensitized with hamster serum. Hepatic xenografts were placed in naive recipients to see if antibodies to hamster serum proteins could be eluted from the rejecting organ. Sera of immunized rats were examined for the presence of anti-hamster antibodies by immunoelectrophoresis and by Western blotting following sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) separation of hamster serum. Antibodies in sera of immunized rats were compared with those eluted from rejecting livers. Candidate antigens were identified by tandem mass spectrometry, sequence analysis, and reference to protein databases. Results showed that sera of immunized rats recognized a minimum of four different antigens in hamster serum by immunoelectrophoresis, and a minimum of seven by the more sensitive SDS-PAGE Western blot. IgG eluted from rejecting livers bound three of seven candidate antigens recognized by sera of the immunized animals. Sequence analysis searches revealed proteinase inhibitors in each of the three SDS-PAGE bands common to the above samples. All of these candidate proteinase inhibitor immunogens share a common catabolic fate, uptake via the lipoprotein-related protein (LRP/alpha 2-macroglobulin receptor (CD91). Sensitization to hamster serum proteins hastened cardiac xenograft rejection in 30-50% of recipients (depending on sensitization protocol). Vascular deposition of rat IgG occurred in all rejecting xenografts. Antibody binding to proteinase inhibitors could disturb their functional activity and contribute to the pathogenesis of delayed xenograft rejection.
Collapse
Affiliation(s)
- S Celli
- Department of Surgery, University of Pittsburgh Transplant Institute, PA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
205
|
Saenko EL, Yakhyaev AV, Mikhailenko I, Strickland DK, Sarafanov AG. Role of the low density lipoprotein-related protein receptor in mediation of factor VIII catabolism. J Biol Chem 1999; 274:37685-92. [PMID: 10608826 DOI: 10.1074/jbc.274.53.37685] [Citation(s) in RCA: 164] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the present study, we found that catabolism of coagulation factor VIII (fVIII) is mediated by the low density lipoprotein receptor-related protein (LPR), a liver multiligand endocytic receptor. In a solid phase assay, fVIII was shown to bind to LRP (K(d) 116 nM). The specificity was confirmed by a complete inhibition of fVIII/LRP binding by 39-kDa receptor-associated protein (RAP), an antagonist of all LRP ligands. The region of fVIII involved in its binding to LRP was localized within the A2 domain residues 484-509, based on the ability of the isolated A2 domain and the synthetic A2 domain peptide 484-509 to prevent fVIII interaction with LRP. Since vWf did not inhibit fVIII binding to LRP, we proposed that LRP receptor may internalize fVIII from its complex with vWf. Consistent with this hypothesis, mouse embryonic fibroblasts that express LRP, but not fibroblasts genetically deficient in LRP, were able to catabolize (125)I-fVIII complexed with vWf, which was not internalized by the cells. These processes could be inhibited by RAP and A2 subunit of fVIII, indicating that cellular internalization and degradation were mediated by interaction of the A2 domain of fVIII with LRP. In vivo studies of (125)I-fVIII.vWf complex clearance in mice demonstrated that RAP completely inhibited the fast phase of the biphasic (125)I-fVIII clearance that is responsible for removal of 60% of fVIII from circulation. Inhibition of the RAP-sensitive phase prolonged the half-life of (125)I-fVIII in circulation by 3.3-fold, indicating that LRP receptor plays an important role in fVIII clearance.
Collapse
Affiliation(s)
- E L Saenko
- Holland Laboratory, American Red Cross, Rockville, Maryland 20855, USA
| | | | | | | | | |
Collapse
|
206
|
Rogove AD, Siao C, Keyt B, Strickland S, Tsirka SE. Activation of microglia reveals a non-proteolytic cytokine function for tissue plasminogen activator in the central nervous system. J Cell Sci 1999; 112 ( Pt 22):4007-16. [PMID: 10547361 DOI: 10.1242/jcs.112.22.4007] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tissue plasminogen activator mediates excitotoxin-induced neurodegeneration and microglial activation in the mouse hippocampus. Here we show that tissue plasminogen activator (tPA) acts in a protease-independent manner to modulate the activation of microglia, the cells of the central nervous system with macrophage properties. Cultured microglia from tPA-deficient mice can phagocytose as efficiently as wild-type microglia. However, tPA-deficient microglia in mixed cortical cultures exhibit attenuated activation in response to lipopolysaccharide, as judged by morphological changes, increased expression of the activation marker F4/80 and the release of the pro-inflammatory cytokine tumor necrosis factor-(α). When tPA is added to tPA deficient cortical cultures prior to endotoxin stimulation, microglial activation is restored to levels comparable to that observed in wild-type cells. Proteolytically-inactive tPA can also restore activation of tPA-deficient microglia in culture and in vivo. However, this inactive enzyme does not restore susceptibility of tPA-deficient hippocampal neurons to excitotoxin-mediated cell death. These results dissociate two different functions of tPA: inactive enzyme can mediate microglial activation, whereas proteolytically-competent protein also promotes neuronal degeneration. Thus tPA is identified as a new cytokine in the central nervous system.
Collapse
Affiliation(s)
- A D Rogove
- Program in Pharmacology, MSTP Program, Department of Psychiatry, University Medical Center at Stony Brook, Stony Brook, NY 11794-8101, USA
| | | | | | | | | |
Collapse
|
207
|
Laithwaite JE, Benn SJ, Yamate J, FitzGerald DJ, LaMarre J. Enhanced macrophage resistance to Pseudomonas exotoxin A is correlated with decreased expression of the low-density lipoprotein receptor-related protein. Infect Immun 1999; 67:5827-33. [PMID: 10531236 PMCID: PMC96962 DOI: 10.1128/iai.67.11.5827-5833.1999] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/1999] [Accepted: 09/03/1999] [Indexed: 11/20/2022] Open
Abstract
Cellular intoxification by exotoxin A of Pseudomonas aeruginosa (PEA) begins when PEA binds to its cellular receptor, the low-density lipoprotein receptor-related protein (LRP). This receptor is particularly abundant on macrophages. We hypothesize here that inducible changes in cellular expression levels of the LRP represent an important mechanism by which macrophage susceptibility to PEA is regulated by the host. We have examined the effect of lipopolysaccharide (LPS) on LRP expression and PEA sensitivity in the macrophage-like cell line HS-P. Using a [(3)H]leucine incorporation assay to measure inhibition of protein synthesis, we have demonstrated that HS-P macrophages are highly sensitive to PEA and that PEA toxicity is decreased by the LRP antagonist receptor-associated protein. LPS pretreatment decreases HS-P PEA sensitivity in a time- and dose-dependent manner. The dose of toxin required to inhibit protein synthesis by 50% increased from 11.3 +/- 1.2 ng/ml in untreated cells to 25.7 +/- 2.0 ng/ml in cells treated with LPS. In pulse experiments, involving brief exposure to saturating concentrations of PEA, [(3)H]leucine incorporation was more than threefold higher in cells pretreated with LPS than in untreated macrophages. These changes in HS-P PEA sensitivity following LPS treatment were consistently associated with a fivefold decrease in HS-P LRP mRNA expression as measured by Northern blot analysis and a three-and-a-half-fold decrease in HS-P LRP-specific ligand internalization as determined by activated alpha(2)-macroglobulin internalization studies. These data demonstrate for the first time that modulation of LRP levels by extracellular signaling molecules can alter cellular PEA sensitivity.
Collapse
Affiliation(s)
- J E Laithwaite
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
208
|
Sharma S, Podder SK, Karande AA. Comparative studies on kinetics of inhibition of protein synthesis in intact cells by ricin and a conjugate of ricin B-chain with momordin. Mol Cell Biochem 1999; 200:133-41. [PMID: 10569193 DOI: 10.1023/a:1007043218769] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ribosome inactivating proteins from plants have been widely used for the preparation of immunotoxins and hormonotoxins, which have potential application in the therapy of diseases such as cancer. However, these hybrid toxins have been found to be less cytotoxic than native ribosome inactivating proteins. Therefore, it is important to understand the factors that control the intrinsic toxicity of RIPs and the hybrid toxins prepared using them. Here, a hybrid toxin has been prepared by coupling ricin B-chain to momordin and the cytotoxicity of this hybrid toxin has been compared to that observed in case of native ricin. In the two cell types used here, thymocytes and macrophages, the conjugate was found to be about 40 fold less toxic than native ricin. Kinetics of inhibition of protein synthesis showed that prior to onset of inhibition the conjugate exhibits a longer lag phase than native ricin. The rates of inhibition of protein synthesis by the conjugate were also found to be slower than ricin. Analysis of the results suggests that in addition to cell surface binding, the B-chain of ricin facilitates another step in the transmembrane translocation of ricin A-chain to the cytosol.
Collapse
Affiliation(s)
- S Sharma
- Department of Biochemistry, Indian Institute of Science, Bangalore
| | | | | |
Collapse
|
209
|
Lenting PJ, Neels JG, van den Berg BM, Clijsters PP, Meijerman DW, Pannekoek H, van Mourik JA, Mertens K, van Zonneveld AJ. The light chain of factor VIII comprises a binding site for low density lipoprotein receptor-related protein. J Biol Chem 1999; 274:23734-9. [PMID: 10446132 DOI: 10.1074/jbc.274.34.23734] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the present study, the interaction between the endocytic receptor low density lipoprotein receptor-related protein (LRP) and coagulation factor VIII (FVIII) was investigated. Using purified components, FVIII was found to bind to LRP in a reversible and dose-dependent manner (K(d) approximately 60 nM). The interaction appeared to be specific because the LRP antagonist receptor-associated protein readily inhibited binding of FVIII to LRP (IC(50) approximately 1 nM). In addition, a 12-fold molar excess of the physiological carrier of FVIII, i.e. von Willebrand factor (vWF), reduced the binding of FVIII to LRP by over 90%. Cellular degradation of (125)I-labeled FVIII by LRP-expressing cells ( approximately 8 fmol/10(5) cells after a 4.5-h incubation) was reduced by approximately 70% in the presence of receptor-associated protein. LRP-directed antibodies inhibited degradation to a similar extent, indicating that LRP indeed contributes to binding and transport of FVIII to the intracellular degradation pathway. Degradation of FVIII was completely inhibited by vWF. Because vWF binding by FVIII involves its light chain, LRP binding to this subunit was studied. In ligand blotting experiments, binding of FVIII light chain to LRP could be visualized. More detailed analysis revealed that FVIII light chain interacts with LRP with moderate affinity (k(on) approximately 5 x 10(4) M(-1) s(-1); k(off) approximately 2.5 x 10(-3) s(-1); K(d) approximately 50 nM). Furthermore, experiments using recombinant FVIII C2 domain showed that this domain contributes to the interaction with LRP. In contrast, no association of FVIII heavy chain to LRP could be detected under the same experimental conditions. Collectively, our data demonstrate that in vitro LRP is able to bind FVIII at the cell surface and to mediate its transport to the intracellular degradation pathway. FVIII-LRP interaction involves the FVIII light chain, and FVIII-vWF complex formation plays a regulatory role in LRP binding. Our findings may explain the beneficial effect of vWF on the in vivo survival of FVIII.
Collapse
Affiliation(s)
- P J Lenting
- Department of Plasma Protein Technology, CLB, 1066 CX Amsterdam The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Abstract
The low-density lipoprotein (LDL) receptor (LDL-R) family consists of cell-surface receptors that recognize extracellular ligands and internalize them for degradation by lysosomes. The LDL-R is the prototype of this family, which also contains very-low-density lipoprotein receptors (VLDL-R), apolipoprotein E receptor 2, LRP, and megalin. The family members contain four major structural modules: the cysteine-rich complement-type repeats, epidermal growth factor precursor-like repeats, a transmembrane domain, and a cytoplasmic domain. Each structural module serves distinct and important functions. These receptors bind several structurally dissimilar ligands. It is proposed that instead of a primary sequence, positive electrostatic potential in different ligands constitutes a receptor binding domain. This family of receptors plays crucial roles in various physiologic functions. LDL-R plays an important role in cholesterol homeostasis. Mutations cause familial hypercholesterolemia and premature coronary artery disease. LDL-R-related protein plays an important role in the clearance of plasma-activated alpha 2-macroglobulin and apolipoprotein E-enriched lipoproteins. It is essential for fetal development and has been associated with Alzheimer's disease. Megalin is the major receptor in absorptive epithelial cells of the proximal tubules and an antigenic determinant for Heymann nephritis in rats. Mutations in a chicken homolog of VLDL-R cause female sterility and premature atherosclerosis. This receptor is not expressed in liver tissue; however, transgenic expression of VLDL-R in liver corrects hypercholesterolemia in experiment animals, which suggests that it can be a candidate for gene therapy for various hyperlipidemias. The functional importance of individual receptors may lie in their differential tissue expression. The regulation of expression of these receptors occurs at the transcriptional level. Expression of the LDL-R is regulated by intracellular sterol levels involving novel membrane-bound transcription factors. Other members of the family are not regulated by sterols. All the members are, however, regulated by hormones and growth factors, but the mechanisms of regulation by hormones have not been elucidated. Studies of these receptors have provided important insights into receptor structure-function and mechanisms of ligand removal and catabolism. It is anticipated that increased knowledge about the LDL-R family members will open new avenues for the treatment of many disorders.
Collapse
Affiliation(s)
- M M Hussain
- Department of Biochemistry, MCP Hahnemann University, Philadelphia, Pennsylvania 19129, USA.
| | | | | |
Collapse
|
211
|
Increased Clearance Explains Lower Plasma Levels of Tissue-Type Plasminogen Activator by Estradiol: Evidence for Potently Enhanced Mannose Receptor Expression in Mice. Blood 1999. [DOI: 10.1182/blood.v94.4.1330.416k19_1330_1336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several clinical studies have demonstrated an inverse relationship between circulating levels of estrogen and tissue-type plasminogen activator (t-PA). The present study was designed to test the hypothesis that estrogens lower plasma levels of t-PA by increasing its clearance from the bloodstream. 17-Ethinyl estradiol (EE) treatment resulted in a significant increase in the clearance rate of recombinant human t-PA in mice (0.46 mL/min in treated mice v 0.32 mL/min in controls; P < .01). The clearance of endogenous, bradykinin-released t-PA in rats was also significantly increased after EE treatment (area under the curve [AUC], 24.9 ng/mL · min in treated animals v 31.9 ng/mL · min in controls; P < .05). Two distinct t-PA clearance systems exist in vivo: the low-density lipoprotein receptor-related protein (LRP) on liver parenchymal cells and the mannose receptor on mainly liver endothelial cells. Inhibition of LRP by intravenous injection of receptor-associated protein (RAP) as a recombinant fusion protein with Salmonella japonicum glutathione S-transferase (GST) significantly retarded t-PA clearance in control mice (from 0.41 to 0.25 mL/min; n = 5, P < .001) and EE-treated mice (from 0.66 to 0.35 mL/min; n = 5, P < .005), but did not eliminate the difference in clearance capacity between the 2 experimental groups. Similar results were obtained in mice in which LRP was inhibited via overexpression of the RAP gene in liver by adenoviral gene transduction. In contrast, administration of mannan, a mannose receptor antagonist, resulted in identical clearances (0.22 mL/min in controls and 0.24 mL/min in EE-treated mice). Northern blot analysis showed a 6-fold increase in mannose receptor mRNA expression in the nonparenchymal liver cells of EE-treated mice, whereas the parenchymal LRP mRNA levels remained unchanged. These findings were confirmed at the protein level by ligand blotting and Western blotting analysis. Our results demonstrate that EE treatment results in increased plasma clearance rate of t-PA via induction of the mannose receptor and could explain for the inverse relationship between estrogen status and plasma t-PA concentrations as observed in humans.
Collapse
|
212
|
Increased Clearance Explains Lower Plasma Levels of Tissue-Type Plasminogen Activator by Estradiol: Evidence for Potently Enhanced Mannose Receptor Expression in Mice. Blood 1999. [DOI: 10.1182/blood.v94.4.1330] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractSeveral clinical studies have demonstrated an inverse relationship between circulating levels of estrogen and tissue-type plasminogen activator (t-PA). The present study was designed to test the hypothesis that estrogens lower plasma levels of t-PA by increasing its clearance from the bloodstream. 17-Ethinyl estradiol (EE) treatment resulted in a significant increase in the clearance rate of recombinant human t-PA in mice (0.46 mL/min in treated mice v 0.32 mL/min in controls; P < .01). The clearance of endogenous, bradykinin-released t-PA in rats was also significantly increased after EE treatment (area under the curve [AUC], 24.9 ng/mL · min in treated animals v 31.9 ng/mL · min in controls; P < .05). Two distinct t-PA clearance systems exist in vivo: the low-density lipoprotein receptor-related protein (LRP) on liver parenchymal cells and the mannose receptor on mainly liver endothelial cells. Inhibition of LRP by intravenous injection of receptor-associated protein (RAP) as a recombinant fusion protein with Salmonella japonicum glutathione S-transferase (GST) significantly retarded t-PA clearance in control mice (from 0.41 to 0.25 mL/min; n = 5, P < .001) and EE-treated mice (from 0.66 to 0.35 mL/min; n = 5, P < .005), but did not eliminate the difference in clearance capacity between the 2 experimental groups. Similar results were obtained in mice in which LRP was inhibited via overexpression of the RAP gene in liver by adenoviral gene transduction. In contrast, administration of mannan, a mannose receptor antagonist, resulted in identical clearances (0.22 mL/min in controls and 0.24 mL/min in EE-treated mice). Northern blot analysis showed a 6-fold increase in mannose receptor mRNA expression in the nonparenchymal liver cells of EE-treated mice, whereas the parenchymal LRP mRNA levels remained unchanged. These findings were confirmed at the protein level by ligand blotting and Western blotting analysis. Our results demonstrate that EE treatment results in increased plasma clearance rate of t-PA via induction of the mannose receptor and could explain for the inverse relationship between estrogen status and plasma t-PA concentrations as observed in humans.
Collapse
|
213
|
Hussaini IM, Brown MD, Weaver AM, Carpenter J, Karns LR, Vandenberg SR, Gonias SL. Stable antisense RNA expression neutralizes the activity of low-density lipoprotein receptor-related protein and promotes urokinase accumulation in the medium of an astrocytic tumor cell line. ANTISENSE & NUCLEIC ACID DRUG DEVELOPMENT 1999; 9:183-90. [PMID: 10355824 DOI: 10.1089/oli.1.1999.9.183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Low-density lipoprotein receptor-related protein (LRP) binds and internalizes multiple ligands that are structurally and functionally diverse. However, the effects of LRP on cellular phenotype remain unclear. To study LRP in human astrocytic tumor cells, we designed LRP antisense RNA expression constructs in which the antisense cDNA fragment was expressed under the control of the cytomegalovirus (CMV) promoter. U-1242 MG astrocytic tumor cells were transfected with the antisense constructs and cloned from single cells to yield multiple cell lines with decreased LRP expression. Further studies were performed with two cell lines in which LRP antigen was completely eliminated (L(alpha)42) or substantially decreased (Lalpha47), as determined by Western blot analysis. Untransfected U-1242 MG cells and cells that were stably transfected with empty vector (pBK-CMV) bound activated alpha2-macroglobulin (alpha2M) in a specific and saturable manner. The Bmax was about 5000 receptors/cell. Lalpha42 cells did not bind alpha2M, and binding was decreased by >60% in Lalpha47 cells. Lalpha42 and Lalpha47 cells also demonstrated reduced susceptibility to the cytotoxin, Pseudomonas exotoxin A, and accumulated greatly increased levels of urokinase-type plasminogen activator (uPA) in conditioned medium. The accumulation of uPA demonstrates a major role for LRP in the catabolism of this protein in astrocytic tumor cells. The LRP-deficient cell lines, developed using antisense technology, represent a new model system for studying LRP function in astrocytes.
Collapse
Affiliation(s)
- I M Hussaini
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville 22908, USA
| | | | | | | | | | | | | |
Collapse
|
214
|
Clatworthy AE, Stockinger W, Christie RH, Schneider WJ, Nimpf J, Hyman BT, Rebeck GW. Expression and alternate splicing of apolipoprotein E receptor 2 in brain. Neuroscience 1999; 90:903-11. [PMID: 10218790 DOI: 10.1016/s0306-4522(98)00489-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Apolipoprotein E isoforms affect the risk of developing Alzheimer's disease. Apolipoprotein E-associated risk may be related to its binding to and clearance by cell surface receptors, including members of the low-density lipoprotein receptor family. We examined the brain expression of the most recently identified member of this receptor family, apolipoprotein E receptor 2, in human brain and placenta. We analysed apolipoprotein E receptor 2 messenger RNA by reverse transcription-polymerase chain reaction and apolipoprotein E receptor 2 protein by immunohistochemistry. Four exons of the apolipoprotein E receptor 2 message were alternately spliced in both fetal and adult brain tissue. Exon 5, encoding three of the seven ligand binding repeats, was absent in the apolipoprotein E receptor 2 messenger RNA examined. Apolipoprotein E receptor 2 messages lacking exon 8, encoding an epidermal growth factor precursor repeat, exon 15, encoding the O-glycosylation region, or exon 18, encoding a cytoplasmic domain, were also present as minor splice variants in the brain and placenta. No differences were observed in the pattern of apolipoprotein E receptor 2 splicing between control and Alzheimer brains. Immunohistochemistry of mouse brain showed that apolipoprotein E receptor 2 was expressed in neurons throughout the brain, with strong expression in pyramidal neurons of the hippocampus, granule cells of the dentate gyrus, cortical neurons and Purkinje cells of the cerebellum. Thus, apolipoprotein E receptor 2 is the fourth apolipoprotein E receptor identified on neuronal cells.
Collapse
Affiliation(s)
- A E Clatworthy
- Alzheimer Research Unit, Massachusetts General Hospital, Boston 02129, USA
| | | | | | | | | | | | | |
Collapse
|
215
|
Paka L, Kako Y, Obunike JC, Pillarisetti S. Apolipoprotein E containing high density lipoprotein stimulates endothelial production of heparan sulfate rich in biologically active heparin-like domains. A potential mechanism for the anti-atherogenic actions of vascular apolipoprotein e. J Biol Chem 1999; 274:4816-23. [PMID: 9988721 DOI: 10.1074/jbc.274.8.4816] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Reduced heparin and heparan sulfate (HS) proteoglycans (PG) have been observed in both inflammation and atherosclerosis. Methods to increase endogenous heparin and heparan sulfate are not known. We found that incubation of endothelial cells with 500-1,000 micrograms/ml high density lipoprotein (HDL) increased 35SO4 incorporation into PG by 1.5-2.5-fold. A major portion of this increase was in HS and was the result of increased synthesis. Total PG core proteins were not altered by HDL; however, the ratio of 35SO4 to [3H]glucosamine was increased by HDL, suggesting increased sulfation of glycosaminoglycans. In addition, HDL increased the amount of highly sulfated heparin-like HS in the subendothelial matrix. HS from HDL-treated cells bound 40 +/- 5% more 125I-antithrombin III (requires 3-O sulfated HS) and 49 +/- 3% fewer monocytes. Moreover, the HS isolated from HDL-treated cells inhibited smooth muscle cell proliferation (by 83 +/- 5%) better than control HS (56 +/- 6%) and heparin (42 +/- 6%). HDL isolated from apolipoprotein E (apoE)-null mice did not stimulate HS production unless apoE was added. ApoE also stimulated HS production in the absence of HDL. ApoE did not increase 35SO4 incorporation in macrophages and fibroblasts, suggesting that this is an endothelial cell-specific process. Receptor-associated protein inhibited apoE-mediated stimulation of HS only at higher (20 micrograms/ml) doses, suggesting the involvement of a receptor-associated protein-sensitive pathway in mediating apoE actions. In summary, our data identify a novel mechanism by which apoE and apoE-containing HDL can be anti-atherogenic. Identification of specific apoE peptides that stimulate endothelial heparin/HS production may have important therapeutic applications.
Collapse
Affiliation(s)
- L Paka
- Division of Preventive Medicine and Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | |
Collapse
|
216
|
Xu PT, Gilbert JR, Qiu HL, Ervin J, Rothrock-Christian TR, Hulette C, Schmechel DE. Specific regional transcription of apolipoprotein E in human brain neurons. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:601-11. [PMID: 10027417 PMCID: PMC1850012 DOI: 10.1016/s0002-9440(10)65305-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/26/1998] [Indexed: 02/03/2023]
Abstract
In central nervous system injury and disease, apolipoprotein E (APOE, gene; apoE, protein) might be involved in neuronal injury and death indirectly through extracellular effects and/or more directly through intracellular effects on neuronal metabolism. Although intracellular effects could clearly be mediated by neuronal uptake of extracellular apoE, recent experiments in injury models in normal rodents and in mice transgenic for the human APOE gene suggest the additional possibility of intraneuronal synthesis. To examine whether APOE might be synthesized by human neurons, we performed in situ hybridization on paraffin-embedded and frozen brain sections from three nondemented controls and five Alzheimer's disease (AD) patients using digoxigenin-labeled antisense and sense cRNA probes to human APOE. Using the antisense APOE probes, we found the expected strong hybridization signal in glial cells as well as a generally fainter signal in selected neurons in cerebral cortex and hippocampus. In hippocampus, many APOE mRNA-containing neurons were observed in sectors CA1 to CA4 and the granule cell layer of the dentate gyrus. In these regions, APOE mRNA containing neurons could be observed adjacent to nonhybridizing neurons of the same cell class. APOE mRNA transcription in neurons is regionally specific. In cerebellar cortex, APOE mRNA was seen only in Bergmann glial cells and scattered astrocytes but not in Purkinje cells or granule cell neurons. ApoE immunocytochemical localization in semi-adjacent sections supported the selectivity of APOE transcription. These results demonstrate the expected result that APOE mRNA is transcribed and expressed in glial cells in human brain. The important new finding is that APOE mRNA is also transcribed and expressed in many neurons in frontal cortex and human hippocampus but not in neurons of cerebellar cortex from the same brains. This regionally specific human APOE gene expression suggests that synthesis of apoE might play a role in regional vulnerability of neurons in AD. These results also provide a direct anatomical context for hypotheses proposing a role for apoE isoforms on neuronal cytoskeletal stability and metabolism.
Collapse
Affiliation(s)
- P T Xu
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | |
Collapse
|
217
|
Yamamoto M, Ikeda K, Ohshima K, Tsugu H, Kimura H, Tomonaga M. Expression and cellular localization of low-density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor in human glioblastoma in vivo. Brain Tumor Pathol 1999; 15:23-30. [PMID: 9879460 DOI: 10.1007/bf02482097] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Low-density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor (LRP) has been proposed to mediate the cellular uptake and clearance of inactivated protease-inhibitor complexes in regulating proteinase activity at the cell surface, which is necessary for cellular migration and invasive processes. In this study, we investigated the presence of both LRP and urokinase-type plasminogen activator receptor (uPAR) in glioblastoma by reverse transcriptase-polymerase chain reaction (RT-PCR), and the cellular localization of LRP in glioblastoma tissues by immunohistochemical analysis. LRP mRNA was frequently expressed in glioblastomas and anaplastic astrocytomas compared with low-grade astrocytomas by RT-PCR analysis, and was well correlated with uPAR expression. The immunohistochemistry of LRP on sequential frozen sections showed that neoplastic glial cells and endothelial cells of glioblastomas exhibited intense LRP immunoreactivity, whereas LRP was almost undetectable in low-grade astrocytomas or in normal glial cells and endothelial cells of normal brain tissue. Glioblastomas from 11 patients in which the expression of LRP mRNA was observed by PCR displayed strong to moderate LRP immunoreactivity, with predominantly diffuse cytoplasmic and cell-surface localization. In normal brain tissues, LRP immunoreactivity was identified in the pyramidal neurons of the cerebral cortex. These results indicate that LRP is present both in the cellular cytoplasm and on the cell surface of glioblastomas with an increased expression of uPAR. Altered LRP expression might contribute to the stimulation of cell-surface proteolytic activity that in turn facilitates the invasiveness of glioblastoma in vivo.
Collapse
Affiliation(s)
- M Yamamoto
- Department of Neurosurgery, Fukuoka University School of Medicine, Japan.
| | | | | | | | | | | |
Collapse
|
218
|
Beshgetoor D, Lönnerdal B. Identification of an alpha2-macroglobulin receptor in human mammary epithelial cells. J Nutr 1999; 129:152-7. [PMID: 9915892 DOI: 10.1093/jn/129.1.152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several cases of zinc (Zn) deficiency in human infants caused by abnormally low concentrations of Zn in breast milk were recently reported, the underlying mechanism of which is not known. Alpha2-macroglobulin (alpha2-M), a major Zn-binding ligand in serum, presents a potential vehicle for mammary Zn uptake. This study was conducted to determine if an alpha2-M receptor is present in human mammary epithelial cells, where it may be involved in the endocytosis of alpha2-M into the mammary gland. Normal human mammary epithelial cells were grown to confluency in serum-free medium. For all binding and uptake studies, alpha2-M, preactivated with methylamine and labeled with 125I, was added to cells for varied lengths of time to determine saturation over time and at varied concentrations to determine saturation over increasing concentration of ligand. Nonspecific and competitive binding were measured by addition of a 100-fold molar excess of unlabeled alpha2-M and serum albumin or lactoferrin, respectively. Binding at 4 degreesC was specific for alpha2-M and approached saturation kinetics at 56 nmol/L. Scatchard plot analysis of the binding data demonstrated more than one binding site: a high affinity, saturable binding site and a low affinity, nonsaturable binding site. Uptake of alpha2-M at 37 degreesC was rapid and continuous over increasing concentrations of alpha2-M, and internalized alpha2-M was rapidly degraded. Results from this study present evidence for receptor-mediated uptake of alpha2-M in human mammary epithelial cells, which in turn, provides a potential mechanism for Zn acquisition by the cell.
Collapse
Affiliation(s)
- D Beshgetoor
- Department of Nutrition, University of California, Davis, CA 95616, U.S. Fax: (530) 752-8966 Tel: (530) 752-8347 E-mail:
| | | |
Collapse
|
219
|
Hussaini IM, Brown MD, Karns LR, Carpenter J, Redpath GT, Gonias SL, Vandenberg SR. Epidermal growth factor differentially regulates low density lipoprotein receptor-related protein gene expression in neoplastic and fetal human astrocytes. Glia 1999. [DOI: 10.1002/(sici)1098-1136(19990101)25:1<71::aid-glia7>3.0.co;2-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
220
|
Zhang JC, Sakthivel R, Kniss D, Graham CH, Strickland DK, McCrae KR. The low density lipoprotein receptor-related protein/alpha2-macroglobulin receptor regulates cell surface plasminogen activator activity on human trophoblast cells. J Biol Chem 1998; 273:32273-80. [PMID: 9822706 DOI: 10.1074/jbc.273.48.32273] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein receptor-related protein/alpha2-macroglobulin receptor (LRP/alpha2MR) mediates the internalization of numerous ligands, including prourokinase (pro-UK) and complexes between two-chain urokinase (tc-u-PA) and plasminogen activator inhibitor type-1 (PAI-1). It has been suggested that through its ability to internalize these ligands, LRP/alpha2MR may regulate the expression of plasminogen activator activity on cell surfaces; this hypothesis, however, has not been experimentally confirmed. To address this issue, we assessed the ability of LRP/alpha2MR to regulate plasminogen activator activity on human trophoblast cells, which express both LRP/alpha2MR and the urokinase receptor (uPAR). Trophoblasts internalized and degraded exogenous 125I-pro-UK (primarily following its conversion to tc-u-PA and incorporation into tc-u-PA.PAI complexes) in an LRP/alpha2MR-dependent manner, which was inhibited by the LRP/alpha2MR receptor-associated protein. Receptor-associated protein also caused a approximately 50% reduction in cell surface plasminogen activator activity and delayed the regeneration of unoccupied uPAR by cells on which uPAR were initially saturated with pro-UK. Identical effects were caused by anti-LRP/alpha2MR antibodies. These results demonstrate that LRP/alpha2MR promotes the expression of cell surface plasminogen activator activity on trophoblasts by facilitating the clearance of tc-u-PA.PAI complexes and regeneration of unoccupied cell surface uPAR.
Collapse
Affiliation(s)
- J C Zhang
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | |
Collapse
|
221
|
Williams KR, Saunders AM, Roses AD, Armati PJ. Uptake and internalization of exogenous apolipoprotein E3 by cultured human central nervous system neurons. Neurobiol Dis 1998; 5:271-9. [PMID: 9848097 DOI: 10.1006/nbdi.1998.0198] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Apolipoprotein E (apoE) has been confirmed as a risk factor for late-onset Alzheimer's disease (AD) and is associated with neurofibrillary tangles and senile plaques, the microscopic pathological characteristics of AD. There has been no direct evidence that human central nervous system neurons can take up and internalize exogenous apoE, which may be important in order for apoE to be involved in the development of the disease. This paper demonstrates by immunohistochemistry and confocal microscopy that cultured human brain neurons can take up and internalize exogenous recombinant human apoE3. We confirm that neurons express the low-density lipoprotein receptor-related protein (LRP) but do not express the low-density lipoprotein receptor. We also demonstrate that the LRP mediates the neuronal uptake of apoE.
Collapse
Affiliation(s)
- K R Williams
- Neuroscience Unit, School of Biological Sciences, University of Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
222
|
Popović M, Caballero-Bleda M, Puelles L, Popović N. Importance of immunological and inflammatory processes in the pathogenesis and therapy of Alzheimer's disease. Int J Neurosci 1998; 95:203-36. [PMID: 9777440 DOI: 10.3109/00207459809003341] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The contribution of autoimmune processes or inflammatory components in the etiology and pathogenesis of Alzheimer's disease (AD) has been suspected for many years. The presence of antigen-presenting, HLA-DR-positive and other immunoregulatory cells, components of complement, inflammatory cytokines and acute phase reactants have been established in tissue of AD neuropathology. Although these data do not confirm the immune response as a primary cause of AD, they indicate involvement of immune processes at least as a secondary or tertiary reaction to the preexisting pathogen and point out its driving-force role in AD pathogenesis. These processes may contribute to systemic immune response. Thus, experimental and clinical studies indicate impairments in both humoral and cellular immunity in an animal model of AD as well as in AD patients. On the other hand, anti-inflammatory drugs applied for the treatment of some chronic inflammatory diseases have been shown to reduce risk of AD in these patients. Therefore, it seems that anti-inflammatory drugs and other substances which can control the activity of immunocompetent cells and the level of endogenous immune response can be valuable in the treatment of AD patients.
Collapse
Affiliation(s)
- M Popović
- Departamento de Ciencias Morfológicas y Psicobiología, Facultad de Medicina, Universidad de Murcia, Espinardo, Spain
| | | | | | | |
Collapse
|
223
|
Birkenmeier G, Usbeck E, Schäfer A, Otto A, Glander HJ. Prostate-specific antigen triggers transformation of seminal alpha2-macroglobulin (alpha2-M) and its binding to alpha2-macroglobulin receptor/low-density lipoprotein receptor-related protein (alpha2-M-R/LRP) on human spermatozoa. Prostate 1998; 36:219-25. [PMID: 9719021 DOI: 10.1002/(sici)1097-0045(19980901)36:4<219::aid-pros2>3.0.co;2-a] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND The aim of this study was to identify the proteolytic activity which triggers the transformation of human alpha2-macroglobulin (alpha2-M) in seminal fluid and its binding to its receptor. METHODS Measurement of the concentrations of total and transformed alpha2-M in seminal fluid was accomplished by ELISA. Zymography of seminal plasma was performed in SDS-polyacrylamide gels containing casein as proteolytic substrate. Rate electrophoresis, SDS-PAGE, and Western blotting were applied to study the complex formation of prostate-specific antigen (PSA) with alpha-M. Ligand-binding analysis of sperm cells was performed using [125I] labeled proteins. Detection of receptor on sperm cells was achieved by immunofluorescence. RESULTS The mean concentration of total alpha2-M in a random collection of seminal plasma was 4.6 microg/ml. On average, between 33-98% of the inhibitor was found to be transformed. Zymography of seminal plasma revealed a proteolytic activity which is associated with a 33-kDa protein identified as PSA. Its proteolytic activity could be inhibited by 0.2-M. Both purified PSA and seminal plasma were capable of transforming native alpha2-M. Binding of PSA to alpha2-M triggers the exposition of receptor binding sites in the inhibitor molecule, which causes binding of the complex to alpha2-M-R/LRP identified on spermatozoa. CONCLUSIONS PSA, the main proteinase in seminal fluid, is responsible for the transformation of alpha2-M and for its binding to alpha2-M-R/LRP present on spermatozoa. The binding of alpha2-M-PSA complexes to the spermatozoa receptor may exert an impact on normal sperm-cell functions.
Collapse
Affiliation(s)
- G Birkenmeier
- Institute of Biochemistry and Department of Andrology, University of Leipzig, Germany.
| | | | | | | | | |
Collapse
|
224
|
Kanaki T, Bujo H, Hirayama S, Tanaka K, Yamazaki H, Seimiya K, Morisaki N, Schneider WJ, Saito Y. Developmental regulation of LR11 expression in murine brain. DNA Cell Biol 1998; 17:647-57. [PMID: 9726247 DOI: 10.1089/dna.1998.17.647] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Receptors belonging to the low density lipoprotein receptor (LDLR) superfamily play important biological roles in addition to mediating lipoprotein metabolism. The recent discovery of a novel mosaic LDLR family member by us (Yamazaki H., Bujo, H., Kusunoki, J., Seimiya, K., Kanaki, T., Morisaki, N., Schneider, W.J., and Saito, Y. (1996) J. Biol. Chem. 271, 24761-24768) and others, which we termed LR11, offers the opportunity to gain new insights into receptor multifunctionality. The predominant expression of LR11 in brain and the presence of elements found in neural adhesion molecules suggested a function(s) in the central nervous system (CNS). In order to gain information about this complex receptor in an accessible system, we have molecularly characterized the murine LR11 and report on its detailed localization and developmental expression pattern. The primary sequence of the murine protein further establishes that LRlls are among the closest relatives within the LDLR family and that brain is the predominant site of expression. In situ hybridization showed that neuronal bodies such as Purkinje cells in the cerebellum and other neurons in the hippocampal formations and the cerebral cortex are particularly rich in LR11 transcripts. The developmental pattern of LR11 expression in brain, which peaks at 2 weeks, is in contrast to those of two other LDLR family members, the very low density lipoprotein receptor and the LDLR. During early development, murine LR11 expression levels are highly dependent on neural cell types. These findings are compatible with function(s) of LR11 in neural organization and, possibly, pathogenesis of degenerative brain diseases. In addition, detailed knowledge of LR11 biology will help to elucidate the roles of other mosaic proteins that share with LR11 elements whose function is not yet known.
Collapse
Affiliation(s)
- T Kanaki
- Second Department of Internal Medicine, Chiba University School of Medicine, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Wheal HV, Chen Y, Mitchell J, Schachner M, Maerz W, Wieland H, Van Rossum D, Kirsch J. Molecular mechanisms that underlie structural and functional changes at the postsynaptic membrane during synaptic plasticity. Prog Neurobiol 1998; 55:611-40. [PMID: 9670221 DOI: 10.1016/s0301-0082(98)00026-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The synaptic plasticity that is addressed in this review follows neurodegeneration in the brain and thus has both structural as well as functional components. The model of neurodegeneration that has been selected is the kainic acid lesioned hippocampus. Degeneration of the CA3 pyramidal cells results in a loss of the Schaffer collateral afferents innervating the CA1 pyramidal cells. This is followed by a period of structural plasticity where new synapses are formed. These are associated with changes in the numbers and shapes of spines as well as changes in the morphometry of the dendrites. It is suggested that this synaptogenesis is responsible for an increase in the ratio of NMDA to AMPA receptors mediating excitatory synaptic transmission at these synapses. Changes in the temporal and spatial properties of these synapses resulted in an altered balance between LTP and LTD. These properties together with a reduction in the inhibitory drive increased the excitability of the surviving CA1 pyramidal cells which in turn triggered epileptiform bursting activity. In this review we discuss the insights that may be gained from studies of the underlying molecular machinery. Developments in one of the collections of the cogs in this machinery has been summarized through recent studies characterizing the roles of neural recognition molecules in synaptic plasticity in the adult nervous systems of vertebrates and invertebrates. Such investigations of neural cell adhesion molecules, cadherins and amyloid precursor protein have shown the involvement of these molecules on the morphogenetic level of synaptic changes, on the one hand, and signal transduction effects, on the other. Further complex cogs are found in the forms of the low-density lipoprotein receptor (LDL-R) family of genes and their ligands play pivotal roles in the brain development and in regulating the growth and remodelling of neurones. Evidence is discussed for their role in the maintenance of cognitive function as well as Alzheimer's. The molecular mechanisms responsible for the clustering and maintenance of transmitter receptors at postsynaptic sites are the final cogs in the machinery that we have reviewed. Postsynaptic densities (PSD) from excitatory synapses have yielded many cytoskeletal proteins including actin, spectrin, tubulin, microtubule-associated proteins and calcium/calmodulin-dependent protein kinase II. Isolated PSDs have also been shown to be enriched in AMPA, kainate and NMDA receptors. However, recently, a new family of proteins, the MAGUKs (for membrane-associated guanylate kinase) has emerged. The role of these proteins in clustering different NMDA receptor subunits is discussed. The MAGUK proteins are also thought to play a role in synaptic plasticity mediated by nitric oxide (NO). Both NMDA and non-NMDA receptors are highly clustered at excitatory postsynaptic sites in cortical and hippocampal neurones but have revealed differences in their choice of molecular components. Both GABAA and glycine (Gly) receptors mediate synaptic inhibition in the brain and spinal cord. Whilst little is known about how GABAA receptors are localized in the postsynaptic membrane, considerable progress has been made towards the elucidation of the molecular mechanisms underlying the formation of Gly receptors. It has been shown that the peripheral membrane protein gephyrin plays a pivotal role in the formation of Gly receptor clusters most likely by anchoring the receptor to the subsynaptic cytoskeleton. Evidence for the distribution as well as function of gephyrin and Gly receptors is discussed. Postsynaptic membrane specializations are complex molecular machinery subserving a multitude of functions in the proper communication between neurones. Despite the fact that only a few key players have been identified it will be a fascinating to watch the story as to how they contribute to structural and functional plasticity unfold.
Collapse
Affiliation(s)
- H V Wheal
- Neuroscience Research Group, School of Biological Sciences, University of Southampton, U.K..
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Neels J, Horn I, van den Berg B, Pannekoek H, van Zonneveld AJ. Ligand-receptor interactions of the low density lipoprotein receptor-related protein, a multi-ligand endocytic receptor. ACTA ACUST UNITED AC 1998. [DOI: 10.1016/s0268-9499(98)80016-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
227
|
Bu G, Sun Y, Schwartz AL, Holtzman DM. Nerve growth factor induces rapid increases in functional cell surface low density lipoprotein receptor-related protein. J Biol Chem 1998; 273:13359-65. [PMID: 9582384 DOI: 10.1074/jbc.273.21.13359] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein receptor-related protein (LRP) is a large endocytic receptor that binds multiple ligands and is highly expressed in neurons. Several LRP ligands, including apolipoprotein E/lipoproteins and amyloid precursor protein, have been shown to participate either in Alzheimer's disease pathogenesis or pathology. However, factors that regulate LRP expression in neurons are unknown. In the current study, we analyzed the effects of nerve growth factor (NGF) treatment on LRP expression, distribution, and function within neurons in two neuronal cell lines. Our results show that NGF induces a rapid increase of cell surface LRP expression in a central nervous system-derived neuronal cell line, GT1-1 Trk, which was seen within 10 min and reached a maximum at about 1 h of NGF treatment. This increase of cell surface LRP expression is concomitant with an increase in the endocytic activity of LRP as measured via ligand uptake and degradation assays. We also found that the cytoplasmic tail of LRP is phosphorylated and that NGF rapidly increases the amount of phosphorylation. Furthermore, we detected a significant increase of LRP expression at the messenger RNA level following 24 h of NGF treatment. Both rapid and long term induction of LRP expression were also detected in peripheral nervous system-derived PC12 cells following NGF treatment. Taken together, our results demonstrate that NGF regulates LRP expression in neuronal cells.
Collapse
Affiliation(s)
- G Bu
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
228
|
Glial fibrillary acidic protein-apolipoprotein E (apoE) transgenic mice: astrocyte-specific expression and differing biological effects of astrocyte-secreted apoE3 and apoE4 lipoproteins. J Neurosci 1998. [PMID: 9547235 DOI: 10.1523/jneurosci.18-09-03261.1998] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The epsilon4 allele of apolipoprotein E (apoE) is associated with increased risk for Alzheimer's disease (AD) and poor outcome after brain injury. In the CNS, apoE is expressed by glia, predominantly astrocytes. To define the potential biological functions of different human apoE isoforms produced within the brain, transgenic mice were generated in which human apoE3 and apoE4 expression is under control of the astrocyte-specific glial fibrillary acidic protein (GFAP) promoter. These animals were then bred back to apoE knock-out mice. Human apoE protein is found within astrocytes and the neuropil throughout development and into the adult period, as assessed by immunocytochemistry and immunoblot analysis in several GFAP-apoE3 and E4 lines. Cultured astrocytes from these mice secrete apoE3 and apoE4 in lipoproteins that are high-density lipoprotein-like in size. When primary hippocampal neurons are grown in the presence of astrocyte monolayers derived from these transgenic mice, there is significantly greater neurite outgrowth from neurons grown in the presence of apoE3-secreting astrocytes compared with apoE4-secreting or apoE knock-out astrocytes. These effects are not dependent on direct astrocyte-neuron contact and appear to require the low-density lipoprotein receptor-related protein. These data suggest that astrocyte-secreted, apoE3-containing lipoproteins have different biological effects than apoE4-containing lipoproteins. In addition to providing information regarding the role of astrocyte-secreted apoE lipoproteins in the normal brain, these animals will also be useful in models of both AD and CNS injury.
Collapse
|
229
|
Ramdin L, Perks B, Sheron N, Shute JK. Regulation of interleukin-8 binding and function by heparin and alpha2-macroglobulin. Clin Exp Allergy 1998; 28:616-24. [PMID: 9645599 DOI: 10.1046/j.1365-2222.1998.00283.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increased expression of interleukin-8 (IL-8), a potent neutrophil chemoattractant, is associated with a number of inflammatory diseases. Interleukin-8 binds to the glycosaminoglycan (GAG) heparin and the protease inhibitor alpha2-macroglobulin, molecules which regulate the function of a number of cytokines. Heparan sulphate was previously shown to enhance neutrophil chemotactic responses to IL-8. OBJECTIVE The purpose of this study was to investigate the effect of heparin, heparan sulphate and alpha2-macroglobulin on IL-8 binding to neutrophils and subsequent functional effects in vitro. METHODS The binding of 125I-IL-8 to normal neutrophils at 4 degrees C was studied and the IL-8 induced neutrophil chemotactic response was investigated using micro-Boyden chambers. Complexation of IL-8 with alpha2-macroglobulin was confirmed using gel filtration chromatography. RESULTS Heparin, but not heparan sulphate, inhibited the binding of 125I-IL-8 to neutrophils (IC50=26 microg/mL) and IL-8 induced neutrophil chemotactic responses (IC50=4 microg/mL). The specific inhibitory effect of heparin was apparently due to an interaction with IL-8 which was charge-dependent, since dextran sulphate had a greater inhibitory effect on chemotactic responses (IC50=2 microg/mL) and FITC-heparin did not bind to neutrophils. The heparin-induced inhibition of IL-8 binding and chemotactic responses was reversed in a dose-dependent manner in the presence of alpha2-macroglobulin. The binding of 125I-IL-8 to neutrophils in the presence of alpha2-macroglobulin appears to be, in part, through the specific IL-8 receptor. CONCLUSION These results point to an anti-inflammatory role for heparin and a novel, potentially, pro-inflammatory role for alpha2-macroglobulin which together indicate the importance of cytokine-binding macromolecules in determining net cytokine function.
Collapse
Affiliation(s)
- L Ramdin
- University Medicine, Southampton General Hospital, UK
| | | | | | | |
Collapse
|
230
|
Abstract
Atherosclerosis is characterized by the presence of lipid-loaded cells which are derived from macrophages and smooth muscle cells. Several lipoprotein receptors may be involved in cellular lipid uptake. These receptors include: scavenger receptor(s); LDL receptor-related protein/alpha2-macroglobulin receptor (LRP); LDL receptor; and VLDL receptor. With the exception of the LDL receptor, all of these receptors are expressed in atherosclerotic lesions. While scavenger receptors are mostly expressed in macrophages, the LRP and VLDL receptor may play an important role in mediating lipid uptake in smooth muscle cells. It is evident that no single receptor pathway is solely responsible for the increased lipid uptake in lesion cells but several redundant mechanisms may contribute to the uptake and degradation of lipoproteins in atherosclerotic lesions.
Collapse
Affiliation(s)
- T P Hiltunen
- Department of Medical Biochemistry, Tampere University Medical School and University Hospital, Finland
| | | |
Collapse
|
231
|
DeMattos RB, Curtiss LK, Williams DL. A minimally lipidated form of cell-derived apolipoprotein E exhibits isoform-specific stimulation of neurite outgrowth in the absence of exogenous lipids or lipoproteins. J Biol Chem 1998; 273:4206-12. [PMID: 9461617 DOI: 10.1074/jbc.273.7.4206] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Within the central nervous system, apolipoprotein E (apoE) synthesis is increased in response to nerve injury, a finding that may reflect a role for apoE in neuronal remodeling. Recent studies show that apoE3 promotes and apoE4 inhibits neurite outgrowth in cultured neuronal cells. Interestingly, these isoform-specific effects are observed only when apoE is presented to cells in the presence of an exogenous lipid source such as rabbit beta-very low density lipoprotein (beta-VLDL), making it difficult to discern the biologically active form of apoE or to understand the role of the lipid source. In the present study we tested whether a cell-derived lipidated form of apoE can alter neurite outgrowth in the absence of beta-VLDL by constructing Neuro-2a cell lines expressing high levels of apoE. Our results showed that endogenous apoE3 stimulated neurite outgrowth, whereas the endogenous apoE4 isoform was neutral. Furthermore, beta-VLDL antagonized the stimulatory effects of the endogenous apoE3. Characterization of the secreted apoE3 indicated that the neurite outgrowth-stimulating activity could be recovered from culture medium with an anti-apoE immunoaffinity column and was present in a poorly lipidated particle with a density between 1.19 and 1.26 g/ml. These results indicated that the biological activity of apoE3 in stimulating neurite outgrowth was inherent in the cell-derived apoE particle and was not dependent on either (a) an interaction of apoE3 with an artificial lipid source or (b) independent actions of apoE3 and beta-VLDL.
Collapse
Affiliation(s)
- R B DeMattos
- Department of Pharmacological Sciences, University Medical Center, State University of New York, Stony Brook, New York 11794, USA
| | | | | |
Collapse
|
232
|
Isoform-specific effect of apolipoprotein E on cell survival and beta-amyloid-induced toxicity in rat hippocampal pyramidal neuronal cultures. J Neurosci 1998. [PMID: 9412500 DOI: 10.1523/jneurosci.18-01-00195.1998] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although the genetic link between the epsilon4 allele of apolipoprotein E (apoE) and Alzheimer's disease is well established, the isoform-specific activity of apoE underlying this correlation remains unclear. To determine whether apoE influences the neurotoxic actions of beta-amyloid (Abeta), we examined the effect of native preparations of apoE3 and E4 on Abeta-induced toxicity in primary cultures of rat hippocampal pyramidal neurons. The source of apoE was conditioned medium from HEK-293 cells stably transfected with human apoE3 or E4 cDNA. ApoE4 (10 microg/ml) alone was toxic to the cultures, whereas apoE3 had no effect. ApoE3 treatment prevented the toxicity induced by 10 microM Abeta(1-40) or Abeta(25-35). The apoE3 protective effect appears to be specific to Abeta-induced toxicity, because apoE3 did not protect against the cytotoxicity produced by NMDA or staurosporine, nor did apoE3 affect the increase in intracellular calcium induced by either NMDA or KCl. ApoE3 had no effect on the toxicity produced by Abeta in the presence of receptor-associated protein, an inhibitor of apoE receptors, particularly the LDL-receptor-related protein. Interaction with apoE receptors may not mediate the toxic actions of apoE4, because receptor-associated protein did not affect apoE4-induced neurotoxicity. Consistent with our previous biochemical experiments, analysis of the culture medium revealed that SDS-stable apoE3:Abeta complex is present in greater abundance than apoE4:Abeta complex. Thus, the protection from Abeta-induced neurotoxicity afforded by apoE3 treatment may result from clearance of the peptide by apoE3:Abeta complex formation and uptake by apoE receptors.
Collapse
|
233
|
Otto A, Bär J, Birkenmeier G. Prostate-specific antigen forms complexes with human alpha 2-macroglobulin and binds to the alpha 2-macroglobulin receptor/LDL receptor-related protein. J Urol 1998; 159:297-303. [PMID: 9400498 DOI: 10.1016/s0022-5347(01)64085-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE To investigate the binding of the prostate-specific antigen (PSA) to human alpha 2-macroglobulin (alpha 2-M) and to alpha 1-antichymotrypsin (ACT). MATERIALS AND METHODS Binding analysis was evaluated by electrophoresis, Western-blotting, enzyme-linked immunosorption assay (ELISA) and size exclusion chromatography. Quantification of PSA and of different forms of alpha 2-M was performed using commercial test kits. The cleavage site of PSA in alpha 2-M was analyzed by SDS-PAGE and microsequencing. RESULTS Binding of PSA to alpha 2-M is initiated by the cleavage of the peptide bond between amino acids Tyr 686 and Glu 687 of the bait region indicating a chymotrypsin-like activity of the PSA. The PSA's proteolytic cleavage triggers the transformation of alpha 2-M as detected by conformation-specific monoclonal antibodies. Kinetic analysis revealed faster binding of PSA to alpha 2-M than to ACT. The PSA bound to alpha 2-M is caged by the inhibitor and thus escapes detection by antibodies. This results in an incorrect calculation of the level of PSA when released from prostate into the blood. Complexes of PSA-alpha 2-M and PSA-ACT were found to bind to the alpha 2-macroglobulin receptor/LDL receptor-related protein (alpha 2-M-R/LRP) which may be the clearance receptor for PSA. CONCLUSIONS Quantifying free PSA and PSA-ACT complexes, as routinely done in managing prostate-associated diseases, does not represent the total secretion capacity of the prostate. The proteinase inhibitor alpha 2-M has to be considered as a main contributor to PSA complex formation in the blood.
Collapse
Affiliation(s)
- A Otto
- Institute of Biochemistry, University of Leipzig, Germany
| | | | | |
Collapse
|
234
|
Grimsley PG, Quinn KA, Chesterman CN, Owensby DA. Low density lipoprotein receptor-related protein (LRP) expression varies among Hep G2 cell lines. Thromb Res 1997; 88:485-98. [PMID: 9610960 DOI: 10.1016/s0049-3848(97)00287-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The multiligand receptor, low density lipoprotein receptor-related protein (LRP), is implicated in processes such as atherosclerosis and fibrinolysis through its mediation of the catabolism of lipoproteins, proteases, and protease inhibitor complexes. The hepatoma cell line Hep G2 expresses LRP and has been used widely to investigate the catabolism of LRP ligands including tissue-type plasminogen activator (tPA). However, the mechanism and degree by which tPA interacts with Hep G2 has been reported with some inconsistencies which may reflect variation in their level of LRP expression. To address this possibility we characterized, antigenically and functionally, LRP expression in high and low passage Hep G2 cells both from the parental line (ATCC sourced) and a cloned subline, a16. The LRP contribution to 125I-tPA binding varied from 65% for high passage a16 cells, to 20% for low passage parent cells as quantified by inhibition in the presence of 39-kD receptor associated protein (RAP) which prevents binding of all known LRP ligands. The same trend in LRP expression among Hep G2 sublines was further evident in their ability to degrade 125I-tPA and survive Pseudomonas exotoxin A challenge. These results imply wide variability in basal LRP expression among Hep G2 lines dependent on cell lineage and long-term culture conditions.
Collapse
Affiliation(s)
- P G Grimsley
- Centre for Thrombosis and Vascular Research, University of New South Wales, Sydney, Australia.
| | | | | | | |
Collapse
|
235
|
Storm D, Herz J, Trinder P, Loos M. C1 inhibitor-C1s complexes are internalized and degraded by the low density lipoprotein receptor-related protein. J Biol Chem 1997; 272:31043-50. [PMID: 9388254 DOI: 10.1074/jbc.272.49.31043] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Like other serpin-enzyme complexes (SECs), proteinase-complexed C1 inhibitor (C1-INH) is rapidly cleared from the circulation and thought to be a neutrophil chemoattractant, suggesting that complex formation causes structural rearrangements exposing a domain which is recognized by specific cell surface receptors. However, the cellular receptor(s) responsible for the catabolism and potential mediation of chemotaxis by C1-INH-protease complexes remained obscure. To determine whether the SEC receptor mediates the binding and potential chemotaxis of C1-INH.Cs, we performed binding assays with HepG2 cells, neutrophils, and monocytes, and the results show that C1-INH.Cs neither bind to these cells nor cause a chemotactic response of neutrophils and monocytes. Furthermore, C1-INH.Cs, the COOH-terminal C1 inhibitor peptide, or the tetrameric C1-INH.Cs.Cr. C1-INH complex were found to be significantly less effective in competing with the SEC receptor ligand 125I-peptide 105Y for the binding to HepG2 cells than unlabeled 105Y, indicating that the SEC receptor does not sufficiently recognize C1-INH-protease complexes. The asialoglycoprotein receptor was also ruled out to be responsible for the removal of the heavily glycosylated C1-INH.Cs complex, since asialoorosomucoid did not compete for the clearance of C1-INH. 125I-Cs and asialoglycoprotein receptor knockout mice showed no alterations in the C1-INH.125I-Cs clearance rate. We found that C1-INH.125I-Cs complexes were efficiently degraded by normal murine fibroblasts expressing the low density lipoprotein receptor-related protein (LRP) and cellular degradation was significantly reduced by chloroquine and the receptor-associated protein, which is a potent inhibitor of the binding of all known ligands to LRP. Moreover, receptor-associated protein inhibited the in vivo clearance of C1-INH.125I-Cs and murine fibroblasts genetically deficient for LRP did not degrade C1-INH.125I-Cs. Our results demonstrate that C1-INH. Cs complexes do not stimulate neutrophil or monocytic chemotaxis but are removed by LRP, further underscoring its role as a serpin-enzyme complex clearance receptor.
Collapse
Affiliation(s)
- D Storm
- Institute of Medical Microbiology and Hygiene, Johannes Gutenberg University, 55101 Mainz, Germany
| | | | | | | |
Collapse
|
236
|
Fabrizi C, Businaro R, Persichini T, Fumagalli L, Lauro GM. The expression of the LDL receptor-related protein (LRP) correlates with the differentiation of human neuroblastoma cells. Brain Res 1997; 776:154-61. [PMID: 9439808 DOI: 10.1016/s0006-8993(97)01035-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The low density lipoprotein receptor-related protein (LRP) has been localized in human brain at the level of neurons, astrocytes and along capillary membranes. It is a multifunctional receptor responsible for binding and internalization of lipoproteins enriched with apoliprotein E, lipoprotein lipase, protease-alpha 2 macroglobulin complexes and plasminogen activator-inhibitor complexes. LRP expression is observed in cells involved in Alzheimer's disease, neoplastic transformation and tissue repair. Moreover, its synthesis is modulated during brain development. In this study we used the SK-N-AS human neuroblastoma cell line as a model system to study LRP expression during cellular differentiation induced by phorbol esters, retinoic acid and interferon gamma. Since LRP plays a major role in the regulation of lipid metabolism, the decreased levels of LRP measured by immunofluorescence, western blot and PCR on differentiated neuroblastoma cells may be the consequence of the lower requirements of cholesterol and lipids of differentiated cells in relation to their reduced mitotic index.
Collapse
MESH Headings
- Blotting, Western
- Cell Differentiation/drug effects
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic
- Humans
- Interferon-gamma/pharmacology
- Low Density Lipoprotein Receptor-Related Protein-1
- Neuroblastoma
- Phorbol Esters/pharmacology
- RNA, Messenger/metabolism
- Receptors, Immunologic/analysis
- Receptors, Immunologic/genetics
- Receptors, LDL/analysis
- Receptors, LDL/genetics
- Tretinoin/pharmacology
- Tumor Cells, Cultured/chemistry
- Tumor Cells, Cultured/cytology
- Tumor Cells, Cultured/drug effects
Collapse
Affiliation(s)
- C Fabrizi
- Department of Biology, Università di Roma Tre, Italy
| | | | | | | | | |
Collapse
|
237
|
Li Y, Wood N, Yellowlees D, Donnelly P. Expression of alpha 2 macroglobulin receptor/low density lipoprotein receptor-related protein is cell culture density-dependent in human breast cancer cell line BT-20. Biochem Biophys Res Commun 1997; 240:122-7. [PMID: 9367895 DOI: 10.1006/bbrc.1997.7619] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
alpha 2Macroglobulin receptor/low density lipoprotein receptor-related protein (alpha 2MR/LRP) is a multifunctional cell plasma membrane receptor that binds and facilitates the endocytosis of a number of ligands involved in protease regulation and lipoprotein metabolism. In the invasive breast cancer cell line BT-20 we show that the expression of alpha 2MR/LRP can be strongly affected by cell culture density. By comparing measurements of mRNA, total cellular alpha 2MR/LRP antigen, and cell surface alpha 2MR/LRP expression we have demonstrated a marked cell density-dependent regulation of this receptor expression. Increasing the cell density results in a 3.4-fold increase in cell surface alpha 2MR/LRP expression. This corresponds to a marked increase in alpha 2MR/LRP mRNA in Northern blots of total RNA from cells cultured at high density and to consistent increases in alpha 2MR/LRP heavy chain in Western blots of cell lysates from high density cultures. These studies together demonstrate the significant up-regulation of alpha 2MR/LRP expression in BT-20 by increased cell density.
Collapse
Affiliation(s)
- Y Li
- Department of Surgery, North Queensland Clinical School, University of Queensland, Townsville, Australia
| | | | | | | |
Collapse
|
238
|
Neurotoxicity of the 22 kDa thrombin-cleavage fragment of apolipoprotein E and related synthetic peptides is receptor-mediated. J Neurosci 1997. [PMID: 9221767 DOI: 10.1523/jneurosci.17-15-05678.1997] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Potent neurotoxicity is associated with both apolipoprotein E (apoE)-related synthetic peptides and the 22 kDa N-terminal thrombin-cleavage fragment of apoE. Furthermore, the E4 isoform of the 22 kDa fragment is significantly more toxic than the same fragment derived from the E3 isoform, suggesting the possibility of a direct role of apoE-associated neurotoxicity in the pathophysiology of Alzheimer's disease. In the present study, the potential role of cell surface receptors in mediating neurotoxicity was assessed by using a variety of agents that should block the heparin-binding and receptor-binding activity of apoE. Effective inhibitors of neurotoxicity of both the apoE peptides and the apoE fragment include heparin, heparan sulfate, sodium chlorate and heparinase, the low-density lipoprotein (LDL) receptor-related protein receptor-associated protein, and a polyclonal anti-LDL receptor-related protein antibody. These results suggest that the neurotoxicity of the 22 kDa thrombin cleavage fragment of apoE and related peptides is receptor-mediated, and that the most likely candidate receptor is a heparan sulfate proteoglycan-LDL receptor-related protein complex.
Collapse
|
239
|
An In Vitro Model for Toxin-Mediated Vascular Leak Syndrome: Ricin Toxin A Chain Increases the Permeability of Human Endothelial Cell Monolayers. Blood 1997. [DOI: 10.1182/blood.v90.6.2323.2323_2323_2334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vascular leak syndrome (VLS) is the dose-limiting toxicity observed in clinical trials of immunotoxins containing ricin toxin A chain (RTA). RTA itself is thought to cause VLS by damaging vascular endothelial cells, but the exact mechanism remains unclear. This is partially due to the paucity of appropriate models. To study VLS, we developed an in vitro model in which human umbilical vein-derived endothelial cells were first grown to confluence on microporous supports and then cultured under low pressure in the presence or absence of RTA. Endothelial cell barrier function was assessed by measuring the volume of fluid that passed through each monolayer per unit time. We found that RTA significantly increased monolayer permeability at times and concentrations consistent with the onset of VLS in patients treated with RTA-based immunotoxins. Scanning electron microscopy showed that intercellular gaps formed in endothelial monolayers exposed to RTA. Intercellular gap formation followed endothelial cell death caused by the enzymatic activity of RTA. We conclude that RTA is directly toxic to endothelial cells in vitro and speculate that this contributes to VLS in vivo.
Collapse
|
240
|
An In Vitro Model for Toxin-Mediated Vascular Leak Syndrome: Ricin Toxin A Chain Increases the Permeability of Human Endothelial Cell Monolayers. Blood 1997. [DOI: 10.1182/blood.v90.6.2323] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractVascular leak syndrome (VLS) is the dose-limiting toxicity observed in clinical trials of immunotoxins containing ricin toxin A chain (RTA). RTA itself is thought to cause VLS by damaging vascular endothelial cells, but the exact mechanism remains unclear. This is partially due to the paucity of appropriate models. To study VLS, we developed an in vitro model in which human umbilical vein-derived endothelial cells were first grown to confluence on microporous supports and then cultured under low pressure in the presence or absence of RTA. Endothelial cell barrier function was assessed by measuring the volume of fluid that passed through each monolayer per unit time. We found that RTA significantly increased monolayer permeability at times and concentrations consistent with the onset of VLS in patients treated with RTA-based immunotoxins. Scanning electron microscopy showed that intercellular gaps formed in endothelial monolayers exposed to RTA. Intercellular gap formation followed endothelial cell death caused by the enzymatic activity of RTA. We conclude that RTA is directly toxic to endothelial cells in vitro and speculate that this contributes to VLS in vivo.
Collapse
|
241
|
Janciauskiene S, al Rayyes O, Floren CH, Eriksson S. Low density lipoprotein catabolism is enhanced by the cleaved form of alpha-1-antitrypsin. Scand J Clin Lab Invest 1997; 57:325-35. [PMID: 9249880 DOI: 10.3109/00365519709099406] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The frequent occurrence of hypocholesterolaemia following inflammatory processes is well known but unexplained. Elevated plasma levels of serine proteinase inhibitors (serpins) and their complexes with target enzymes have been demonstrated in inflammatory, malignant and infectious diseases which are also often accompanied by low plasma cholesterol levels. Under inflammatory conditions, uncomplexed, but cleaved inactive serpins arising from slow deacylation of the serpin-proteinase complex may be present in the circulation. To determine the influence of native and cleaved forms of serpins, such as alpha-1-antitrypsin (AAT), on lipoprotein metabolism, we investigated the effect of these forms on low density lipoprotein (LDL) catabolism in human HepG2 cell line. We have found that the cleaved form of AAT in concentrations from 125 to 2000 nmol l-1 stimulates LDL binding to the HepG2 cells, by up to 49% with a subsequent increase in LDL uptake and degradation of up to 79 and 65% respectively. Native AAT was also found to increase LDL binding and internalization by 20-25%, independently of the amount of AAT added, an effect most probably due to the cleaved form of AAT produced by local proteolysis of native AAT incubated in the cell culture. Moreover we have shown that the cleaved form of AAT interacts with LDL in vitro, and that such an interaction abolishes AAT ability to stimulate LDL binding and internalization. This study for the first time describes the ability of the cleaved form of AAT to stimulate LDL binding and internalization in HepG2 cell culture, and provides evidence that hypocholesterolaemia occurring during inflammatory processes may be mediated by cleaved forms of serpins.
Collapse
Affiliation(s)
- S Janciauskiene
- Department of Medicine, Lund University Hospital, Malmö, Sweden
| | | | | | | |
Collapse
|
242
|
Noorman F, Rijken D. Regulation of tissue-type plasminogen activator concentrations by clearance via the mannose receptor and other receptors. ACTA ACUST UNITED AC 1997. [DOI: 10.1016/s0268-9499(97)80048-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
243
|
Tiggelman AM, Linthorst C, Boers W, Brand HS, Chamuleau RA. Transforming growth factor-beta-induced collagen synthesis by human liver myofibroblasts is inhibited by alpha2-macroglobulin. J Hepatol 1997; 26:1220-8. [PMID: 9210607 DOI: 10.1016/s0168-8278(97)80455-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Transforming growth factor-beta (TGFbeta) plays a central role in the stimulation of matrix production during liver fibrosis. The action of TGFbeta in different systems has been shown to be influenced by alpha2-macroglobulin (alpha2M), a serum protein with strong protease-scavenging and cytokine-binding properties. AIMS In the present study, alpha2M derived from normal human plasma has been tested for its ability to modulate the TGFbeta-induced collagen production by human liver fat-storing cells (FSC), which had transformed into alpha-smooth muscle actin-expressing myofibroblasts in culture. METHODS Alpha2M has been tested after activation with methylamine (alpha2M-Me), an in vitro equivalent of protease activated alpha2M. The binding of 125I-TGFbeta1 to activated forms of alpha2M was demonstrated by rate electrophoresis. Collagen synthesis was examined in human liver myofibroblast cultures obtained from three different human livers by incorporation of 3H-proline into TCA-precipitable, specific collagenase degradable proteins. Uptake of alpha2M was studied by means of immunofluorescence. RESULTS TGFbeta (1 ng/ml) significantly stimulated collagen synthesis of controls in the absence of TGFbeta. Alpha2M-Me reduced this TGFbeta-induced collagen synthesis dose-dependently, reaching significant inhibition from 10 microg/ml alpha2M-Me onward. Upon addition of 100 microg/ml alpha2M-Me the effect of TGFbeta was reduced by 60% to 128+/-31% (mean+/-SD) of control values in the absence of TGFbeta. Human liver myofibroblasts endocytosed alpha2M-Me added to the cultures as detected by immunofluorescence. Accordingly, reduction of TGFbeta-activity by alpha2M-Me may be explained by receptor-mediated clearance of alpha2M-TGFbeta complexes by the cells. CONCLUSIONS TGFbeta-induced collagen formation by human liver myofibroblasts obtained from three different livers is reduced in vitro by activated alpha2M. From these results, we hypothesize that alpha2M may have an antifibrogenic effect in vivo by interference with TGFbeta-induced matrix synthesis during liver fibrosis.
Collapse
Affiliation(s)
- A M Tiggelman
- Academic Medical Center, University of Amsterdam, J van Gool Laboratory for Experimental Internal Medicine, The Netherlands
| | | | | | | | | |
Collapse
|
244
|
Mikhailenko I, Krylov D, Argraves KM, Roberts DD, Liau G, Strickland DK. Cellular internalization and degradation of thrombospondin-1 is mediated by the amino-terminal heparin binding domain (HBD). High affinity interaction of dimeric HBD with the low density lipoprotein receptor-related protein. J Biol Chem 1997; 272:6784-91. [PMID: 9045712 DOI: 10.1074/jbc.272.10.6784] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Thrombospondin-1 (TSP-1) is a large modular trimeric protein that has been proposed to play a diverse role in biological processes. Newly synthesized TSP-1 either is incorporated into the matrix or binds to the cell surface where it is rapidly internalized and degraded. TSP-1 catabolism is mediated by the low density lipoprotein receptor-related protein (LRP), a large endocytic receptor that is a member of the low density lipoprotein receptor family. Using adenovirus-mediated gene transfer experiments, we demonstrate that the very low density lipoprotein receptor can also bind and internalize TSP-1. An objective of the current investigation was to identify the portion of TSP-1 that binds to these endocytic receptors. The current studies found that the amino-terminal heparin binding domain (HBD, residues 1-214) of mouse TSP-1, when prepared as a fusion protein with glutathione S-transferase (GST), bound to purified LRP with an apparent KD ranging from 10 to 25 nM. Recombinant HBD (rHBD) purified following proteolytic cleavage of GST-HBD, also bound to purified LRP, but with an apparent KD of 830 nM. The difference in affinity was attributed to the fact that GST-HBD exists in solution as a dimer, whereas rHBD is a monomer. Like TSP-1, 125I-labeled GST-HBD or 125I-labeled rHBD were internalized and degraded by wild type fibroblasts that express LRP, but not by fibroblasts that are genetically deficient in LRP. The catabolism of both 125I-labeled GST-HBD and rHBD in wild type fibroblast was blocked by the 39-kDa receptor-associated protein, an inhibitor of LRP function. GST-HBD and rHBD both completely blocked catabolism of 125I-labeled TSP-1 in a dose-dependent manner, as did antibodies prepared against the HBD. Taken together, these data provide compelling evidence that the amino-terminal domain of TSP-1 binds to LRP and thus the recognition determinants on TSP-1 for both LRP and for cell surface proteoglycans reside within the same TSP-1 domain. Further, high affinity binding of TSP-1 to LRP likely results from the trimeric structure of TSP-1.
Collapse
Affiliation(s)
- I Mikhailenko
- Holland Laboratory, American Red Cross, Rockville, Maryland 20855, USA
| | | | | | | | | | | |
Collapse
|
245
|
Brown MD, Banker GA, Hussaini IM, Gonias SL, VandenBerg SR. Low density lipoprotein receptor-related protein is expressed early and becomes restricted to a somatodendritic domain during neuronal differentiation in culture. Brain Res 1997; 747:313-7. [PMID: 9046007 DOI: 10.1016/s0006-8993(96)01321-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Low density lipoprotein receptor-related protein (LRP) is a multi-functional receptor which mediates the endocytotic uptake of several ligands implicated in neuronal pathophysiology. In this study, LRP expression and localization, in cultured hippocampal neurons from 18-day-old rats, were examined by immunofluorescence microscopy. LRP was restricted to the cell bodies and dendrites of mature neurons, where it was uniformly distributed on both dendritic shafts and spines. Immunoreactive protein was detected within the first 24 h of culture and acquired a polarized distribution by the end of the first week. Expression of LRP mRNA by the cultured neurons was demonstrated by Northern blot analysis. Binding studies with the LRP ligand, activated alpha2-macroglobulin, confirmed that LRP was present and functional on the hippocampal neuron cell surface. These studies demonstrate that neuronal LRP undergoes selective compartmentation during neuronal maturation and suggest that LRP-mediated endocytosis is largely restricted to the somatodendritic compartment.
Collapse
Affiliation(s)
- M D Brown
- Department of Pathology (Neuropathology), University of Virginia Health Sciences Center, Charlottesville 22908, USA
| | | | | | | | | |
Collapse
|
246
|
Businaro R, Fabrizi C, Persichini T, Starace G, Ennas MG, Fumagalli L, Lauro GM. Modulation of the alpha 2 macroglobulin receptor/low density lipoprotein receptor related protein by interferon-gamma in human astroglial cells. J Neuroimmunol 1997; 72:75-81. [PMID: 9003247 DOI: 10.1016/s0165-5728(96)00157-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Alpha 2 macroglobulin receptor/low density lipoprotein receptor-related protein (alpha 2 Mr/LRP) is a multi-functional cell surface receptor that has been implicated in important processes, such as atherogenesis, cellular migration, immune response and degenerative diseases. Its expression increases in human brain during Alzheimer's disease, tissue injury and neoplastic transformation. In the present paper we studied the regulation of alpha 2 Mr expression by interferon-gamma (IFN gamma) in human astrocytoma cell lines and in fetal astrocytes. Western blots demonstrated an increase of the alpha 2 Mr expression after 24 h of IFN gamma treatment. This effect paralleled the up-regulation of alpha 2 Mr mRNA, as detected by PCR. By prolonging incubation with IFN gamma, we observed a decrement of alpha 2 Mr in IFN gamma treated cells, both by western blot and cytometric analysis. Since in the same cells IFN gamma also up-regulates alpha 2 macroglobulin, this effect may be due to an augmented degradation of the receptor during its recycling.
Collapse
MESH Headings
- Antibodies, Monoclonal
- Antineoplastic Agents/pharmacology
- Astrocytoma
- Blotting, Western
- Densitometry
- Gene Expression/drug effects
- Glioblastoma
- Humans
- Interferon-gamma/pharmacology
- Low Density Lipoprotein Receptor-Related Protein-1
- Molecular Sequence Data
- Polymerase Chain Reaction/methods
- RNA, Messenger/metabolism
- RNA-Directed DNA Polymerase
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/immunology
- Receptors, LDL/metabolism
- Tumor Cells, Cultured/chemistry
- Tumor Cells, Cultured/drug effects
Collapse
Affiliation(s)
- R Businaro
- Dipartimento di Scienze Cardiovascolari e Respiratorie, Università La Sapienza, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
247
|
Affiliation(s)
- G J Snipes
- Department of Neuropathology, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
248
|
Birkenmeier G, Grosche J, Reichenbach A. Immunocytochemical demonstration of alpha 2-M-R/LRP on Müller (glial) cells isolated from rabbit and human retina. Neuroreport 1996; 8:149-51. [PMID: 9051770 DOI: 10.1097/00001756-199612200-00030] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The alpha 2-macroglobulin receptor/low-density lipoprotein receptor-related protein (alpha 2-M-R/LRP) is a multifunctional receptor which has been implicated in lipoprotein metabolism, clearance of proteinase-proteinase inhibitor complexes and regulation of growth factor/cytokine metabolism. This receptor is abundantly present in numerous tissues and organs such as liver, lung, placenta and brain. In brain it is expressed in neurones but not in normal macroglia. Using immunocytochemistry and monoclonal antibodies against the large extracellular receptor subunit we have detected alpha 2-M-R/LRP on enzymatically isolated retinal Müller (glial) cells. This receptor may be involved in vital functions of Müller cells.
Collapse
Affiliation(s)
- G Birkenmeier
- Institute of Biochemistry, University of Leipzig, Germany
| | | | | |
Collapse
|
249
|
Fagan AM, Bu G, Sun Y, Daugherty A, Holtzman DM. Apolipoprotein E-containing high density lipoprotein promotes neurite outgrowth and is a ligand for the low density lipoprotein receptor-related protein. J Biol Chem 1996; 271:30121-5. [PMID: 8939961 DOI: 10.1074/jbc.271.47.30121] [Citation(s) in RCA: 172] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Presence of the epsilon4 allele of apolipoprotein E (apoE) is a risk factor for Alzheimer's disease (AD), although the mechanism(s) by which it confers this risk is unknown. ApoE may play a direct role in AD neuropathology by modulating neuronal structure. We previously showed that apoE3-containing beta-very low density lipoprotein (beta-VLDL) can stimulate neurite outgrowth to a significantly greater extent than apoE4-enriched beta-VLDL in a central nervous system-derived neuronal cell line and that this effect is mediated by interaction with the low density lipoprotein receptor-related protein (LRP). To determine whether similar differences exist when apoE is associated with other lipoprotein particles, the effects of high density lipoprotein (HDL) derived from plasma and cerebrospinal fluid were defined. ApoE3-enriched HDL significantly enhanced neurite outgrowth as compared with apoE4-enriched HDL, and the majority of this stimulation was blocked in the presence of the receptor-associated protein or a neutralizing antibody to LRP. We also found that cholesterol esterification in the presence of apoE-containing plasma HDL was attenuated in fibroblasts lacking LRP. Therefore, apoE-containing HDL can serve as an LRP ligand, and apoE isoform-specific effects on neurite outgrowth are observed when HDL is the carrier particle.
Collapse
Affiliation(s)
- A M Fagan
- Department of Neurology, Center for the Study of Nervous System Injury, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
250
|
Jung SS, Nalbantoglu J, Cashman NR. Alzheimer's beta-amyloid precursor protein is expressed on the surface of immediately ex vivo brain cells: a flow cytometric study. J Neurosci Res 1996; 46:336-48. [PMID: 8933373 DOI: 10.1002/(sici)1097-4547(19961101)46:3<336::aid-jnr7>3.0.co;2-l] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Beta-amyloid precursor protein (beta APP) is ubiquitously expressed, but deposition of the beta APP proteolytic fragment A beta is virtually restricted to the brain, suggesting cell-specific processing of this molecule. Our laboratory has investigated expression of beta APP in mechanically dissociated, unfixed, immediately ex vivo cells from various mouse and rat organs by flow cytometry. Epitopes of predicted extracellular domains of beta APP recognized by the N-terminal 22C11 monoclonal antibody (mAb) and the juxtamembrane 4G8 mAb were not detectable on the surface of lymphoid cells, hepatocytes, or kidney cells. In contrast, surface 22C11 and 4G8 beta APP immunoreactivity was abundant on intact (propidium iodide-excluding) dissociated brain cells. The predicted C-terminal intracellular beta APP determinant recognized by the mAb Jonas was not detectable on the surface of intact brain cells, but was present in ethanol-permeabilized cells, consistent with a transmembrane configuration of beta APP in brain cells. Trypsinization of intact brain cells abolished cell surface immunoreactivity for 22C11, which was then reestablished by short-term culture. Augmentation of 22C11 and 4G8 surface immunoreactivity occurred when brain cells were cultured short-term in phenylarsine oxide, a general endocytosis inhibitor. By double staining protocols of brain cells with mAbs directed against beta APP ectodomain epitopes and the neuronal surface proteins Thy-1 or neural cell adhesion molecule (NCAM), we observed that all Thy-1+ and NCAM+ cells (approximately 50%) were immunoreactive for surface beta APP, but that some beta APP+ cells (approximately 20%) were negative for these neuronal markers. Our data suggest that neurons and a subpopulation of other brain cells, unlike peripheral cells, can support beta APP as a type 1 intrinsic membrane molecule with an intact ectodomain, and that beta APP surface abundance is regulated by an equilibrium between membranes vesicle insertion and endocytotic internalization. Transmembrane beta APP holoprotein may be a critical determinant of brain-predominant processing of beta APP to A beta, and may participate in a receptor/transducer function unique to brain cells.
Collapse
Affiliation(s)
- S S Jung
- Department of Microbiology and Immunology, McGill University, Montréal Neurological Institute, Canada
| | | | | |
Collapse
|