Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
J Intern Med ; 287(1): 2-18, 2020 01.
Article in English | MEDLINE | ID: mdl-31858669

ABSTRACT

There is compelling evidence that the elevated plasma lipoprotein(a) [Lp(a)] levels increase the risk of atherosclerotic cardiovascular disease (ASCVD) in the general population. Like low-density lipoprotein (LDL) particles, Lp(a) particles contain cholesterol and promote atherosclerosis. In addition, Lp(a) particles contain strongly proinflammatory oxidized phospholipids and a unique apoprotein, apo(a), which promotes the growth of an arterial thrombus. At least one in 250 individuals worldwide suffer from the heterozygous form of familial hypercholesterolemia (HeFH), a condition in which LDL-cholesterol (LDL-C) is significantly elevated since birth. FH-causing mutations in the LDL receptor gene demonstrate a clear gene-dosage effect on Lp(a) plasma concentrations and elevated Lp(a) levels are present in 30-50% of patients with HeFH. The cumulative burden of two genetically determined pro-atherogenic lipoproteins, LDL and Lp(a), is a potent driver of ASCVD in HeFH patients. Statins are the cornerstone of treatment of HeFH, but they do not lower the plasma concentrations of Lp(a). Emerging therapies effectively lower Lp(a) by as much as 90% using RNA-based approaches that target the transcriptional product of the LPA gene. We are now approaching the dawn of an era, in which permanent and significant lowering of the high cholesterol burden of HeFH patients can be achieved. If outcome trials of novel Lp(a)-lowering therapies prove to be safe and cost-effective, they will provide additional risk reduction needed to effectively treat HeFH and potentially lower the CVD risk in these high-risk patients even more than currently achieved with LDL-C lowering alone.


Subject(s)
Coronary Artery Disease/prevention & control , Hyperlipoproteinemia Type II/therapy , Lipoprotein(a)/blood , Aortic Valve , Blood Component Removal , Cholesterol, LDL/blood , Coronary Artery Disease/etiology , Heart Valve Diseases/etiology , Heart Valve Diseases/prevention & control , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperlipoproteinemia Type II/blood , Hypolipidemic Agents/therapeutic use , Oligonucleotides, Antisense/therapeutic use , PCSK9 Inhibitors , Practice Guidelines as Topic , Receptors, LDL/genetics , Risk Factors , Vascular Calcification/etiology , Vascular Calcification/prevention & control
4.
Anim Reprod Sci ; 136(1-2): 1-13, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23123285

ABSTRACT

A unique non-laying strain of chickens with heritable hyperlipidemia and aortic atherosclerosis was first described in 1974. Subsequent work established that the phenotype results from a naturally occurring point mutation in the gene specifying the very low density lipoprotein (VLDL) receptor, a 95-kDa membrane protein which normally mediates the massive uptake of the main circulating hepatically-synthesized yolk precursors, VLDL and vitellogenin. As a result, hens of the mutant strain termed "restricted ovulator" (R/O) have approximately 5-fold elevations in circulating cholesterol and triglyceride concentrations compared with normal layers, and hepatic lipogenesis and cholesterogenesis are markedly attenuated due to feedback inhibition. R/O hens also exhibit hyperestrogenemia, hypoprogesteronemia, elevated circulating gonadotropins, and up-regulated pituitary progesterone receptor mRNA and isoforms. The ovaries of R/O hens are abnormal in that they lack a follicular hierarchy and contain many small preovulatory follicles of various colors, shapes, and sizes. However, since R/O hens occasionally lay eggs, it is possible that endocytic receptors other than the VLDL receptor may be able to facilitate oocyte growth and/or that yolk precursor uptake can occur via a nonspecific bulk process. A mammalian model of impaired fecundity with abnormal lipoprotein metabolism also has been described, but different mechanisms are likely responsible for its reproductive dysfunction. Nevertheless, as our understanding of the molecular physiology and biochemistry of avian oocyte growth continues to expand, in part due to studies of the R/O model, new analogies may emerge between avian and mammalian systems, which ultimately could help to answer important questions in reproductive biology.


Subject(s)
Chickens/genetics , Chickens/physiology , Infertility, Female/genetics , Oviparity/genetics , Oviparity/physiology , Animals , Female , Gene Expression Regulation/physiology , Mutation
5.
Arterioscler Thromb Vasc Biol ; 28(10): 1866-71, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18635818

ABSTRACT

OBJECTIVE: The purpose of this study was to identify rare APOA5 variants in 130 severe hypertriglyceridemic patients by sequencing, and to test their functionality, since no patient recall was possible. METHODS AND RESULTS: We studied the impact in vitro on LPL activity and receptor binding of 3 novel heterozygous variants, apoAV-E255G, -G271C, and -H321L, together with the previously reported -G185C, -Q139X, -Q148X, and a novel construct -Delta139 to 147. Using VLDL as a TG-source, compared to wild type, apoAV-G255, -L321 and -C185 showed reduced LPL activation (-25% [P=0.005], -36% [P<0.0001], and -23% [P=0.02]), respectively). ApoAV-C271, -X139, -X148, and Delta139 to 147 had little affect on LPL activity, but apoAV-X139, -X148, and -C271 showed no binding to LDL-family receptors, LR8 or LRP1. Although the G271C proband carried no LPL and APOC2 mutations, the H321L carrier was heterozygous for LPL P207L. The E255G carrier was homozygous for LPL W86G, yet only experienced severe hypertriglyceridemia when pregnant. CONCLUSIONS: The in vitro determined function of these apoAV variants only partly explains the high TG levels seen in carriers. Their occurrence in the homozygous state, coinheritance of LPL variants or common APOA5 TG-raising variant in trans, appears to be essential for their phenotypic expression.


Subject(s)
Apolipoproteins A/metabolism , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/metabolism , Mutation, Missense , Receptors, LDL/metabolism , Adult , Apolipoprotein A-V , Apolipoproteins A/genetics , DNA Mutational Analysis , Europe , Female , Heterozygote , Homozygote , Humans , Hydrolysis , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/genetics , Lipoproteins, VLDL/metabolism , Male , Models, Molecular , Phenotype , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Protein Binding , Protein Conformation , Recombinant Proteins/metabolism , Severity of Illness Index , Structure-Activity Relationship , Surface Plasmon Resonance , Triglycerides/blood
6.
Cytogenet Genome Res ; 117(1-4): 248-55, 2007.
Article in English | MEDLINE | ID: mdl-17675866

ABSTRACT

The normal development of the chicken oocyte within the ovarian follicle depends on the coordinated expression and function of several members of the low density lipoprotein receptor gene family. The human low density lipoprotein receptor is the prototype of the gene family; since its discovery and the elucidation of the medical significance of mutations in the LDLR gene, many additional family members have been discovered and characterized, and some important advances have resulted from studies in the chicken. I describe the analogies as well as the differences that exist between the molecular genetics of the mammalian and avian members of this important gene family, with emphasis on receptor-mediated oocyte growth. Recent progress in the molecular characterization of the chicken genes whose products mediate oocyte growth, follicle development, and accessory pathways is described in detail, and emerging information of preliminary nature is included. As the availability of chicken genome sequence data has enhanced the rate of progress in the field, our understanding of the physiological roles of members of this receptor family in general has already gained from studies in the avian model system.


Subject(s)
Chickens/metabolism , Oocytes/cytology , Oocytes/metabolism , Oogenesis , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Receptors, LDL/metabolism , Animals , Chickens/genetics , Extracellular Matrix/metabolism , Female , Receptors, LDL/classification , Receptors, LDL/genetics
7.
Cell Mol Life Sci ; 62(16): 1871-80, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16041564

ABSTRACT

Immunological resistance of the chick embryo is dependent upon IgG present in the yolk of the layed egg. Here we show that complement factor 3 (C3), a key component of the humoral complement system, is a yolk component of chicken eggs. C3 is transported into oocytes by LR8-mediated endocytosis. LR8 also binds and transports other major yolk components such as vitellogenin, very-low-density lipoprotein, and alpha(2)-macroglobulin. Expression studies of LR8 during chicken development and oocyte maturation, in combination with studies on the uptake of individual yolk components, suggest the following model for oocyte maturation in the chicken: all oocytes present in the ovary contain high levels of LR8 mRNA and protein long before the onset of oocyte maturation. Selected oocytes gain access to yolk precursors, and LR8 binds, internalizes, and deposits the major yolk components in the ratio of their relative abundance in the accessible pool.


Subject(s)
Avian Proteins/metabolism , Complement C3/metabolism , Endocytosis , Lipoproteins/metabolism , Oocytes/metabolism , Animals , Avian Proteins/genetics , Cell Line , Coated Vesicles/metabolism , Egg Yolk/metabolism , Female , Humans , In Vitro Techniques , Lipoproteins/genetics , Lipoproteins, VLDL/metabolism , Oocytes/growth & development , RNA, Messenger/metabolism , Vitellogenins/metabolism , alpha-Macroglobulins/metabolism
8.
Cell Mol Life Sci ; 60(5): 892-903, 2003 May.
Article in English | MEDLINE | ID: mdl-12827279

ABSTRACT

For many years, the low-density lipoprotein (LDL) receptor and the LDL receptor-related protein (LRP) have been considered to be prototypes of cargo receptors which deliver, via endocytosis, macromolecules into cells. However, the recent identification of additional members of this gene family and examination of their biology has revealed that at least some of these proteins are also signaling receptors. Very low density lipoprotein receptor and ApoER2 transmit the extracellular reelin signal into migrating neurons, and thus are key components of the reelin pathway which governs neuronal layering of the forebrain during embryonic brain development. LRP5 and LRP6 are integral components of the Wnt signaling pathway which is central to many processes of metazoan development, cell proliferation, and tumor formation. Adaptor proteins interacting with the cytosolic domains of these receptors might orchestrate their ability to deliver their cargo or a signal.


Subject(s)
Endocytosis/physiology , Receptors, LDL/physiology , Signal Transduction/physiology , Animals , Egg Proteins/physiology , Humans , Hyperlipoproteinemia Type II/metabolism , LDL-Receptor Related Proteins , Lipoproteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Receptors, Cell Surface/physiology , Receptors, LDL/chemistry , Receptors, Lipoprotein/physiology , Reelin Protein
9.
Arterioscler Thromb Vasc Biol ; 21(9): 1501-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11557679

ABSTRACT

Since the molecular identification of the low density lipoprotein receptor (LDLR), an ever increasing number of related proteins have been discovered. These receptors belonging to the LDLR family are thought to play key roles in lipoprotein metabolism in a variety of tissues, including the arterial wall. We have discovered that the expression of a 250-kDa mosaic LDLR-related protein, which we termed LR11 for the presence of 11 LDLR ligand-binding repeats, is markedly induced in smooth muscle cells in the hyperplastic intima of animal models used for the study of atherosclerosis. Here, we demonstrate that the human LR11, when overexpressed in hamster cells, binds and internalizes 39-kDa receptor-associated protein (RAP), an in vitro ligand for all receptors belonging to the LDLR family. Furthermore, LR11 binds the apolipoprotein E (apoE)-rich lipoproteins, beta-very low density lipoproteins (VLDLs), with a high affinity similar to that of other members, such as the LDLR and VLDL receptor. RAP and beta-VLDL compete with each other; however, other serum lipoproteins are not able to inhibit their binding. LR11 shows specific binding of apoE-enriched HDL prepared from human cerebrospinal fluid as well as of beta-VLDL, suggesting that the apoE content of lipoproteins is most likely important for mediating the high-affinity binding to the receptor. LR11-overexpressing cells are able to internalize and degrade the bound beta-VLDL; these cells also show increased accumulation of cholesteryl esters when incubated with beta-VLDL. Incubation for 48 hours with beta-VLDL of LR11-overexpressing cells, but not of control cells, promotes the appearance of numerous intracellular lipid droplets. Taken together, LR11, a mosaic LDLR family member whose expression in smooth muscle cells is markedly induced in atheroma, has all the properties of a receptor for the endocytosis of lipoproteins, particularly for the incorporation of apoE-rich lipoproteins.


Subject(s)
Apolipoproteins E/metabolism , Lipoproteins, VLDL/metabolism , Receptors, LDL/physiology , Animals , Arteriosclerosis/metabolism , CHO Cells , Carrier Proteins/metabolism , Cholesterol/metabolism , Cricetinae , Endocytosis , Glycoproteins/metabolism , Immunohistochemistry , LDL-Receptor Related Protein-Associated Protein , Lipid Metabolism , Lipoproteins, HDL/metabolism , Mutation , Receptors, LDL/genetics , Receptors, LDL/immunology , Transfection
10.
J Biol Chem ; 276(25): 22160-9, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11294845

ABSTRACT

LR7/8B and ApoER2 are recently discovered members of the low density lipoprotein (LDL) receptor family. Although structurally different, these two proteins are derived from homologous genes in chicken and man by alternative splicing and contain 7 or 8 LDL receptor ligand-binding repeats. Here we present the cDNA for ApoER2 cloned from mouse brain and describe splice variants in the ligand binding domain of this protein, which are distinct from those present in man and chicken. The cloned cDNA is coding for a receptor with only five LDL receptor ligand-binding repeats, i.e. comprising repeats 1-3, 7, and 8. Reverse transcriptase-polymerase chain reaction analysis of mRNA from murine brain revealed the existence of two additional transcripts. One is lacking repeat 8, and in the other repeat 8 is substituted for by a 13-amino acid insertion with a consensus site for furin cleavage arising from an additional small exon present in the murine gene. None of the transcripts in the mouse, however, contain repeats 4-6. In murine placenta only the form containing repeats 1-3 and 7 and the furin cleavage site is detectable. Analysis of the corresponding region of the murine gene showed the existence of 6 exons coding for a total of 8 ligand binding repeats, with one exon encoding repeats 4-6. Exon trapping experiments demonstrated that this exon is constitutively spliced out in all murine transcripts. Thus, the murine ApoER2 gene codes for receptor variants harboring either 4 or 5 binding repeats only. Recombinant expression of the 5-repeat and 4-repeat variants showed that repeats 1-3, 7, and 8 are sufficient for binding of beta-very low density lipoprotein and reelin, but not for recognition of alpha(2)-macroglobulin, which binds to the avian homologue of ApoER2 harboring 8 ligand binding repeats.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Receptors, Lipoprotein/genetics , alpha-Macroglobulins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cloning, Molecular , DNA, Complementary , Exons , Introns , LDL-Receptor Related Proteins , Ligands , Mice , Molecular Sequence Data , Nerve Tissue Proteins , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/metabolism , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases
11.
Biochim Biophys Acta ; 1529(1-3): 287-98, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11111096

ABSTRACT

The discovery of an ever growing number of low density lipoprotein (LDL) receptor gene family members has triggered research into many different directions. Here we first summarize the results of classical studies on the role of the LDL receptor in cholesterol transport, the structure/function relationships delineated with the help of LDL receptor mutations in familial hypercholesterolemia, and the elegant way in which cells regulate cholesterol at the transcriptional level. The second part deals with a multifunctional, structurally very close relative, the very low density lipoprotein (VLDL) receptor. While it is involved in lipoprotein transport in certain tissues and species, detailed studies on its function have generated new knowledge about the growing spectrum of ligands and about exciting and unexpected aspects of receptor biology. In particular, these investigations have elucidated the roles of LDL receptor gene family members in ligand-mediated signal transduction. In the third part of this review article, we provide first insight into the roles of the VLDL receptor and of another small relative, the so-called apolipoprotein E receptor-2, in such signaling processes. These findings suggest that to date, only the tip of an iceberg has been uncovered.


Subject(s)
Cholesterol/metabolism , Receptors, LDL/metabolism , Receptors, Lipoprotein/metabolism , Animals , Apolipoprotein E2 , Apolipoproteins E/metabolism , Biological Transport , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Lipoproteins, VLDL/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mutation , Nerve Tissue Proteins , RNA Splicing , Receptors, LDL/genetics , Receptors, Lipoprotein/genetics , Reelin Protein , Serine Endopeptidases , Signal Transduction , Structure-Activity Relationship
12.
J Biol Chem ; 275(37): 28866-72, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10979984

ABSTRACT

The extracellular matrix surrounding vertebrate oocytes is called the zona pellucida in mammals and perivitelline membrane (pvm) in birds. We have analyzed this structure in chicken follicles and laid eggs and have identified a 95-kDa component of the pvm, which, by protein sequencing, shows homology to mammalian zona pellucida proteins. Surprisingly, we could not detect this protein in ovarian granulosa cells or oocytes but instead found high levels in the liver of the laying hen. In contrast, it is absent in rooster liver but can be efficiently induced by estrogen treatment of the animal. An immunoscreen of a liver lambda-ZAP library yielded a cDNA coding for a protein of 934 amino acids. It displayed significant homology to members of the ZP1/ZPB family from other species, notably to mouse and rat ZP1, and was therefore designated chkZP1. It is clearly different from a protein designated chkZPB that had been deposited in the data base previously. Alignment of the known members of the ZP1/ZPB family demonstrated the existence of at least three subgroups, with representatives of both the ZP1 and the ZPB sequence homology group occurring in vertebrates. Northern blot analysis of liver extracts revealed the presence of a single 3. 2-kilobase mRNA coding for chkZP1, distinct from the chkZPB transcript detectable in follicles. Immunohistochemical analysis of follicle sections demonstrates that chkZP1 can be found in the blood vessels of the theca cell layer as well as in the pvm surrounding the oocyte. Thus, in the chicken, at least one of the major pvm components is synthesized in the liver and is transported via the bloodstream to the follicle.


Subject(s)
Egg Proteins/chemistry , Liver/metabolism , Membrane Glycoproteins/chemistry , Receptors, Cell Surface , Amino Acid Sequence , Animals , Chickens , Cloning, Molecular , Egg Proteins/analysis , Egg Proteins/biosynthesis , Female , Immunohistochemistry , Membrane Glycoproteins/analysis , Membrane Glycoproteins/biosynthesis , Molecular Sequence Data , Molecular Weight , Rabbits , Zona Pellucida Glycoproteins
13.
Biochem Biophys Res Commun ; 275(2): 365-73, 2000 Aug 28.
Article in English | MEDLINE | ID: mdl-10964672

ABSTRACT

An involvement of the low density lipoprotein receptor (LDLR) gene family in both intracellular signal pathways for neural organization and metabolic pathways for lipoprotein homeostasis is now well established. The discovery of LR11, a mosaic LDLR family member offers the opportunity to gain new insights into receptor multifunctionality. Here, we studied the proliferation-dependent expression of LR11 mRNA and protein using two cultured cell lines, IMR32 neuroblastoma and PC12 pheochromocytoma. Within 24 h, the LR11 protein rose 1.9-fold in proliferating IMR32 cells, and increased further to 5.3-fold at 72 h. This conformed with a transcript level increase of 4.7-fold at 72 h in the proliferating cells. On the other hand, under differentiation conditions, a 2.9-fold increase was observed within 24 h, but at 72 h thereafter the protein levels decreased to 60% of control. The transcript also increased to 1. 8-fold within 24 h, and then decreased to 1.1-fold at 72 h. In order to assess the transcriptional activities of the LR11 gene, we identified the 5'-flanking region of the murine LR11 gene. Transfection of IMR32 and PC12 cells with plasmids containing the whole or deleted fragments of 5'-flanking region showed that element(s) responsible for the above described different transcriptional activities are located in the upstream sequence between -861 and -396. Thus, the transcription of LR11 in these two cell systems is regulated differently during proliferation and differentiation, suggesting that the multifunctionality of LR11, as well as other LDLR family members, for rapid cell growth in malignant cells and neural outgrowth in cultured neurons, respectively. The possible involvement of LR11 in cellular proliferation and differentiation sheds new light on its functions in neurons, malignant, and vascular cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Transport Proteins , Neuroblastoma/metabolism , Receptors, LDL/metabolism , Animals , Base Sequence , COS Cells , Cell Differentiation , Cell Division , DNA Primers , DNA, Neoplasm , Molecular Sequence Data , Neuroblastoma/pathology , PC12 Cells , Promoter Regions, Genetic , Rats , Receptors, LDL/genetics , Tumor Cells, Cultured
14.
J Biol Chem ; 275(45): 35320-7, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10948196

ABSTRACT

Development of the follicle in egg-laying species such as the chicken is regulated by systemic factors as well as by the highly orchestrated interplay of differentially expressed genes within this organ. Differential mRNA display analysis of defined phases of follicle development resulted in the characterization of coagulation factor XIIIA. It is expressed and produced by cells of the theca externa in a highly regulated manner during distinct growth phases of the follicle. Transcripts for factor XIIIA are already detectable at the beginning of follicle development and peak at the end of phase 2. Protein levels, however, still increase during phase 3, peak shortly after ovulation, and persist until the postovulatory tissue is completely resorbed. Factor XIIIA is secreted as a monomer into the extracellular matrix of the theca externa and is not associated with factor XIIIB as is the case in plasma. Our data suggest that, due to its transglutaminase activity, factor XIIIA stabilizes the follicular wall by cross-linking matrix components. Thus, coagulation factor XIIIA might play a key role in coping with the massive mechanical stress exerted by the large amount of yolk accumulating during the rapid growth phase of the oocyte.


Subject(s)
Ovarian Follicle/metabolism , Theca Cells/metabolism , Transglutaminases/biosynthesis , Amino Acid Sequence , Animals , Basement Membrane/metabolism , Blotting, Northern , Blotting, Western , Calcium Chloride/pharmacology , Cell Line , Chickens , DNA, Complementary/metabolism , Edetic Acid/pharmacology , Female , Gene Expression Profiling , Glutathione Transferase/metabolism , Granulosa Cells/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Ovulation/physiology , Precipitin Tests , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Time Factors , Transfection , Transglutaminases/metabolism
15.
Cytogenet Cell Genet ; 89(1-2): 67-78, 2000.
Article in English | MEDLINE | ID: mdl-10894941

ABSTRACT

Sex-determination mechanisms in birds and mammals evolved independently for more than 300 million years. Unlike mammals, sex determination in birds operates through a ZZ/ZW sex chromosome system, in which the female is the heterogametic sex. However, the molecular mechanism remains to be elucidated. Comparative gene mapping revealed that several genes on human chromosome 9 (HSA 9) have homologs on the chicken Z chromosome (GGA Z), indicating the common ancestry of large parts of GGA Z and HSA 9. Based on chromosome homology maps, we isolated a Z-linked chicken ortholog of DMRT1, which has been implicated in XY sex reversal in humans. Its location on the avian Z and within the sex-reversal region on HSA 9p suggests that DMRT1 represents an ancestral dosage-sensitive gene for vertebrate sex-determination. Z dosage may be crucial for male sexual differentiation/determination in birds.


Subject(s)
Chickens/genetics , Chromosomes, Human, Pair 9/genetics , Conserved Sequence/genetics , Sex Chromosomes/genetics , Sex Determination Processes , Transcription Factors/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Dosage Compensation, Genetic , Evolution, Molecular , Female , Gene Dosage , Genes/genetics , Genetic Linkage/genetics , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Molecular Sequence Data , Physical Chromosome Mapping , Sequence Alignment , Sex Characteristics , Transcription Factors/chemistry
16.
J Biol Chem ; 275(33): 25625-32, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10827199

ABSTRACT

Correct positioning of neurons during embryonic development of the brain depends, among other processes, on the proper transmission of the reelin signal into the migrating cells via the interplay of its receptors with cytoplasmic signal transducers. Cellular components of this signaling pathway characterized to date are cell surface receptors for reelin like apolipoprotein E receptor 2 (ApoER2), very low density lipoprotein receptor (VLDLR), and cadherin-related neuronal receptors, and intracellular components like Disabled-1 and the nonreceptor tyrosine kinase Fyn, which bind to the intracellular domains of the ApoER2 and VLDL receptor or of cadherin-related neuronal receptors, respectively. Here we show that ApoER2, but not VLDLR, also binds the family of JNK-interacting proteins (JIPs), which act as molecular scaffolds for the JNK-signaling pathway. The ApoER2 binding domain on JIP-2 does not overlap with the binding sites for MLK3, MKK7, and JNK. These results suggest that ApoER2 is able to assemble a multiprotein complex containing Disabled-1 and JIPs, together with their binding partners, to the cell surface of neurons. This complex might participate in ApoER2-specific reelin signaling and thus would explain the different phenotype of mice lacking the ApoER2 from that of VLDLR-deficient mice.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Proline/metabolism , Receptors, Lipoprotein/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Blotting, Northern , Brain/metabolism , Cell Differentiation , Cells, Cultured , Cytoplasm/chemistry , DNA, Complementary/metabolism , Epididymis/metabolism , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , LDL-Receptor Related Proteins , Male , Mice , Microscopy, Fluorescence , Models, Biological , Molecular Sequence Data , Nerve Tissue Proteins , Neurons/cytology , Protein Binding , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Serine Endopeptidases , Signal Transduction , Stem Cells/metabolism , Tissue Distribution , Two-Hybrid System Techniques
17.
J Biol Chem ; 275(22): 16837-44, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10747878

ABSTRACT

Yolk is the major source of nutrients for the developing chicken embryo, but molecular details of the delivery mechanisms are largely unknown. During oogenesis in the chicken, the main yolk components vitellogenin and very low density lipoprotein (VLDL) are taken up into the oocytes via a member of the low density lipoprotein receptor gene family termed LR8 (Bujo, H., Hermann, M., Kaderli, M. O., Jacobsen, L., Sugawara, S., Nimpf, J., Yamamoto, T., and Schneider, W. J. (1994) EMBO J. 13, 5165-5175). This endocytosis is accompanied by partial degradation of the yolk precursor protein moieties; however, fragmentation does not abolish binding of VLDL to LR8. The receptor exists in two isoforms that differ by a so-called O-linked sugar domain; the shorter form (LR8-) is the major form in oocytes, and the longer protein (LR8+) predominates in somatic cells. Here we show that both LR8 isoforms are expressed at ratios that vary with embryonic age in the extraembryonic yolk sac, which mobilizes yolk for utilization by the embryo, and in the allantois, the embryo's catabolic sink. Stored yolk VLDL interacts with LR8 localized on the surface of the yolk sac endodermal endothelial cells (EEC), is internalized, and degraded, as demonstrated by the catabolism of fluorescently labeled VLDL in cultured EEC. Addition to the incubation medium of the 39-kDa receptor-associated protein, which inhibits all known LR8/ligand interactions, blocks the uptake of VLDL by EEC. The levels of endogenous receptor-associated protein correspond to those of LR8+ but not LR8-, suggesting that it may play a role in the modulation of surface presentation of LR8+. Importantly, EEC express significant levels of microsomal triglyceride transfer protein and protein disulfide isomerase, key components required for lipoprotein synthesis. Because the apolipoprotein pattern of VLDL isolated from the yolk sac-efferent omphalomesenteric vein is very different from that of yolk VLDL, these data strongly suggest that embryo plasma VLDL is resynthesized in the EEC. LR8 is a key mediator of a two-step pathway, which affects the uptake of VLDL from the yolk sac and the subsequent delivery of its components to the growing embryo.


Subject(s)
Embryo, Nonmammalian/metabolism , Receptors, LDL/metabolism , Yolk Sac/metabolism , Alternative Splicing , Animals , Base Sequence , Chick Embryo , DNA Primers , Female , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/metabolism
18.
Arterioscler Thromb Vasc Biol ; 19(11): 2687-95, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10559012

ABSTRACT

Receptors belonging to the LDL receptor (LDLR) family are thought to play key roles in lipoprotein metabolism in a variety of tissues, including the arterial wall. Here, we report that the expression of a 250-kDa mosaic LDLR family member, which we called LR11 for the presence of 11 ligand-binding repeats, is markedly induced during the process of atherogenesis in 2 animal models. Analysis by reverse transcription-polymerase chain reaction and RNase protection assays revealed that LR11 transcript levels rise in rabbit aortas displaying atheromatous lesions after the rabbits have been fed a high-cholesterol diet. Immunohistochemistry demonstrated that the highest induction of LR11 occurs in intimal smooth muscle cells (SMCs), followed by medial SMCs close to the intimal border of the atheromatous lesions. Experimental intimal hyperplasia by endothelial denudation showed that LR11 mRNA levels were also increased in the arteries after balloon injury, with the transcripts localized primarily in the hyperplastic intimal layer. In agreement with the correlation of LR11 induction during increased cell proliferation, cultured SMCs showed an increase in LR11 expression in the proliferative phase. Furthermore, Northern and Western blot analyses showed that medium conditioned by the monocyte-macrophage cell line THP-1 enhanced LR11 expression in cultured SMCs. These findings suggest that upregulation of LR11 might be contributing to the pathological roles of intimal and medial SMCs during arteriosclerotic lesion development and provide the first insight into the as yet unknown functional significance of this intriguing LDLR family member.


Subject(s)
Arteriosclerosis/physiopathology , Membrane Transport Proteins , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/pathology , Receptors, LDL/genetics , Angioplasty, Balloon/adverse effects , Animals , Aorta/cytology , Arteriosclerosis/pathology , Carotid Artery, Common/pathology , Carotid Artery, Common/physiopathology , Cells, Cultured , Cholesterol, Dietary/pharmacology , Culture Media, Conditioned/pharmacology , Diet, Atherogenic , Disease Models, Animal , Endothelium, Vascular/pathology , Gene Expression/drug effects , In Situ Hybridization , Male , Mosaicism , Muscle, Smooth, Vascular/injuries , RNA, Messenger/analysis , Rabbits , Receptors, LDL/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
20.
Biol Reprod ; 60(5): 1057-68, 1999 May.
Article in English | MEDLINE | ID: mdl-10208965

ABSTRACT

The expression and localization of vitellogenin (VTG) receptor (VTGR) mRNA were identified throughout ovarian development in the rainbow trout, Oncorhynchus mykiss. Northern blot confirmed the presence of a transcript (approximately 3.9 kilobases [kb]) that was specific to the ovary. The expression of VTGR mRNA varied throughout ovarian development and was highest in previtellogenic ovaries and in ovaries at the onset of vitellogenesis containing ovarian follicles (OF) from 35 to 600 microm in diameter. In situ hybridization using 35S riboprobes showed that the transcription of the VTGR gene was initiated in OF measuring 45-50 microm in diameter, with transcripts being exclusively localized in the ooplasm. A dramatic increase in mRNA synthesis occurred during previtellogenic growth (OF from 50 to 200 microm); this was followed by a gradual decrease during the vitellogenic growth phase. VTGR mRNA was not detected in OF greater than 1000 microm in diameter (oocytes actively sequestering VTG). Immunocytolocalization of yolk proteins derived from VTG demonstrated that oocytes started to sequester VTG when they were around 300 microm in diameter, shortly after the time of maximal density of VTGR mRNA in the ooplasm. The timing of transcription of the VTGR gene, predominantly during previtellogenesis, suggests that the VTGR is recycled to the oocyte surface during the vitellogenic growth phase.


Subject(s)
Egg Proteins , Oncorhynchus mykiss/metabolism , Oogenesis/physiology , Ovarian Follicle/metabolism , RNA, Messenger/biosynthesis , Receptors, Cell Surface/biosynthesis , Animals , Autoradiography , Blotting, Northern , Female , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Microscopy, Electron , RNA Probes , Tissue Fixation
SELECTION OF CITATIONS
SEARCH DETAIL
...