Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Am J Obstet Gynecol ; 212(5): 647.e1-11, 2015 May.
Article in English | MEDLINE | ID: mdl-25731694

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the effects of insulin on human placental transcriptome and biological processes in first-trimester pregnancy. STUDY DESIGN: Maternal plasma and placenta villous tissue were obtained at the time of voluntary termination of pregnancy (7-12 weeks) from 17 lean (body mass index, 20.9±1.5 kg/m2) and 18 obese (body mass index, 33.5±2.6 kg/m2) women. Trophoblast cells were immediately isolated for in vitro treatment with insulin or vehicle. Patterns of global gene expression were analyzed using genome microarray profiling after hybridization to Human Gene 1.1 ST and real time reverse transcription-polymerase chain reaction. RESULTS: The global trophoblast transcriptome was qualitatively separated in insulin-treated vs untreated trophoblasts of lean women. The number of insulin-sensitive genes detected in the trophoblasts of lean women was 2875 (P<.001). Maternal obesity reduced the number of insulin-sensitive genes recovered by 30-fold. Insulin significantly impaired several gene networks regulating cell cycle and cholesterol homeostasis but did not modify pathways related to glucose transport. Obesity associated with high insulin and insulin resistance, but not maternal hyperinsulinemia alone, impaired the global gene profiling of early gestation placenta, highlighting mitochondrial dysfunction and decreased energy metabolism. CONCLUSION: We report for the first time that human trophoblast cells are highly sensitive to insulin regulation in early gestation. Maternal obesity associated with insulin resistance programs the placental transcriptome toward refractoriness to insulin with potential adverse consequences for placental structure and function.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Obesity/genetics , Placenta/drug effects , Pregnancy Complications/genetics , RNA, Messenger/drug effects , Transcriptome/drug effects , Trophoblasts/drug effects , Abortion, Induced , Adolescent , Adult , Case-Control Studies , Cells, Cultured , Female , Gene Expression Profiling , Humans , Obesity/metabolism , Placenta/metabolism , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Trimester, First , RNA, Messenger/metabolism , Transcriptome/genetics , Trophoblasts/metabolism , Young Adult
2.
J Clin Endocrinol Metab ; 99(9): 3427-34, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24796925

ABSTRACT

CONTEXT: Adiponectin (adpN) production is down-regulated in several situations associated with insulin resistance. The hypoadiponectinemia, which develops in late pregnancy, suggests a role of adpN in pregnancy-induced insulin resistance. OBJECTIVE: In obese pregnancy there is a decreased systemic adpN, which results from down-regulation of gene expression in adipose tissue. SETTING AND DESIGN: One hundred and thirty-three women with uncomplicated pregnancies and a wide range in pre-gravid body mass index (18-62 kg/m(2)) were recruited at term for a scheduled cesarean delivery. Maternal blood, placenta, and sc abdominal adipose tissue were obtained in the fasting state. DNA methylation was analyzed by MBD-based genome-wide methylation sequencing and methyl-specific PCR of placenta and maternal adipose tissue. mRNA and protein expression were characterized by real-time RT-PCR and immunodetection. Plasma adpN, leptin, and insulin were assayed by ELISA. RESULTS: Maternal adipose tissue was the prominent site of adpN gene expression with no detectable mRNA or protein in placenta. In obese women, adipose tissue adpN mRNA was significantly decreased (P < .01) whereas DNA methylation was significantly increased (P < .001) compared with lean women. The decreased adipose tissue expression resulted in normal-weight women having significantly greater plasma adpN compared with the severely obese (12.8 ± 4.3 ng/mL vs 8.6 ± 3.1, P < .001). Plasma adpN was negatively correlated with maternal body mass index (r = -0.28, P < .001) and homeostasis model assessment indices of insulin sensitivity (r = -0.32, P < .001) but not with gestational weight gain. CONCLUSIONS: Maternal adipose tissue is the primary source of circulating adpN during pregnancy. Further, based on our results, the placenta does not synthesize adiponectin at term. Obesity in pregnancy is associated with negative regulation of adpN adipose expression with increase in adpN DNA methylation associated with lower mRNA concentrations and hypoadiponectinemia. Maternal hypoadiponectinemia may have functional consequences in down-regulating biological signals transmitted by adpN receptors in various tissues, including the placenta.


Subject(s)
Adiponectin/genetics , DNA Methylation/physiology , Obesity/genetics , Pregnancy Complications/genetics , Adiponectin/deficiency , Adiponectin/metabolism , Adipose Tissue/metabolism , Adult , Body Mass Index , Down-Regulation/genetics , Female , Gene Expression Regulation/physiology , Humans , Infant, Newborn , Insulin/metabolism , Insulin Resistance/physiology , Leptin/metabolism , Obesity/metabolism , Placenta/metabolism , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Trimester, Third/genetics , Pregnancy Trimester, Third/metabolism , RNA, Messenger/metabolism , Young Adult
3.
Obesity (Silver Spring) ; 20(11): 2213-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22836687

ABSTRACT

Remodeling of adipose tissue is required to support the expansion of adipose mass. In obesity, an increased death of adipocytes contributes to the accelerated cellular turnover. We have shown that obesity in pregnancy is associated with metabolic and immune alterations in the adipose tissue. In this study, we characterized the mechanisms responsible for increased death of adipose cells of pregnant obese women and its functional consequences. We postulated that a higher turnover of dead cells in white adipose tissue of obese women would translate into release of cell-free DNA (cfDNA) into their systemic circulation. Increase in adipose mass of obese compared to lean women results from a lesser number of hypertrophic adipocytes and an accumulation of macrophages in the stromal vascular fraction (SVF). The adipocytes of obese displayed enhanced necrosis with a loss of perilipin staining at the plasma membrane. Apoptosis was prominent in SVF cells with an increased expression of caspase 9 and caspase 3 and a higher rate of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) positive CD68 macrophages in obese vs. lean. Whereas circulating fetal cfDNA concentrations were not changed, there was a twofold increase in circulating glyceraldehyde-3-phosphate dehydrogenase (GAPDH) cfDNA and adipose tissue GAPDH mRNA in obese women. The maternal systemic GAPDH cfDNA was positively correlated with BMI and gestational weight gain. These data suggest that the active remodeling of adipose tissue of obese pregnant women results in an increased release of cfDNA of maternal origin into the circulation.


Subject(s)
Adipocytes , Adipose Tissue, White/physiopathology , Inflammation/physiopathology , Obesity/physiopathology , Adipocytes/metabolism , Adipocytes/pathology , Adult , Apoptosis , Cell Death , Cell Differentiation , Cell-Free System , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Necrosis , Pregnancy
4.
Am J Physiol Endocrinol Metab ; 303(7): E832-40, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22811467

ABSTRACT

Changes in adipose tissue metabolism are central to adaptation of whole body energy homeostasis to pregnancy. To gain insight into the molecular mechanisms supporting tissue remodeling, we have characterized the longitudinal changes of the adipose transcriptome in human pregnancy. Healthy nonobese women recruited pregravid were followed in early (8-12 wk) and in late (36-38 wk) pregnancy. Adipose tissue biopsies were obtained in the fasting state from the gluteal depot. The adipose transcriptome was examined via whole genome DNA microarray. Expression of immune-related genes and extracellular matrix components was measured using real-time RT-PCR. Adipose mass, adipocyte size, and cell number increased in late pregnancy compared with pregravid measurements (P < 0.001) but remained unchanged in early pregnancy. The adipose transcriptome evolved during pregnancy with 10-15% of genes being differently expressed compared with pregravid. Functional gene cluster analysis revealed that the early molecular changes affected immune responses, angiogenesis, matrix remodeling, and lipid biosynthesis. Increased expression of macrophage markers (CD68, CD14, and the mannose-6 phosphate receptor) emphasized the recruitment of the immune network in both early and late pregnancy. The TLR4/NF-κB signaling pathway was enhanced specifically in relation to inflammatory adipokines and chemokines genes. We conclude that early recruitment of metabolic and immune molecular networks precedes the appearance of pregnancy-related physiological changes in adipose tissue. This biphasic pattern suggests that physiological inflammation is an early step preceding the development of insulin resistance, which peaks in late pregnancy.


Subject(s)
Adaptation, Physiological , Adipose Tissue/physiology , Inflammation/physiopathology , Pregnancy Trimester, First/physiology , Pregnancy Trimester, Third/physiology , Adipokines/genetics , Adipokines/immunology , Adipokines/metabolism , Adipose Tissue/immunology , Antigens, CD/biosynthesis , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/immunology , Chemokines/genetics , Chemokines/immunology , Chemokines/metabolism , Female , Humans , Inflammation/genetics , Inflammation/immunology , Lipid Metabolism/genetics , Lipid Metabolism/immunology , Lipid Metabolism/physiology , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/immunology , Pregnancy , Pregnancy Trimester, First/genetics , Pregnancy Trimester, First/immunology , Pregnancy Trimester, Third/genetics , Pregnancy Trimester, Third/immunology , Receptor, IGF Type 2/biosynthesis , Receptor, IGF Type 2/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/physiology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transcriptome/genetics , Transcriptome/immunology , Transcriptome/physiology
6.
Am J Obstet Gynecol ; 205(3): 265.e1-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22071058

ABSTRACT

OBJECTIVE: The purpose of this study was to gain insight into the pathways that are associated with inflammation at the maternal-fetal interface. This study examined the molecular characteristics of monocytes that were derived from the maternal circulation and the placenta of obese women. STUDY DESIGN: Mononuclear cells were isolated from placenta, venous maternal, and umbilical cord blood at term delivery; activated monocytes were separated with CD14 immunoselection. The genotype and expression pattern of the monocytes were analyzed by microarray and real-time reverse transcriptase-polymerase chain reaction. RESULTS: The transcriptome of the maternal blood and placental CD14 monocytes exhibited 73% homology, with 10% (1800 common genes) differentially expressed. Genes for immune sensing and regulation, matrix remodeling, and lipid metabolism were enhanced 2-2006 fold in placenta, compared with maternal monocytes. The CD14 placental monocytes exhibited a maternal genotype (9% DYS14 expression) as opposed to the fetal genotype (90% DYS14 expression) of the trophoblast cells. CONCLUSION: CD14 monocytes from the maternal blood and the placenta share strong phenotypic and genotypic similarities with an enhanced inflammatory pattern in the placenta. The functional traits of the CD14 blood and placental monocytes suggest that they both contribute to propagation of inflammation at the maternal-fetal interface.


Subject(s)
Fetal Blood/metabolism , Fetus/metabolism , Inflammation/metabolism , Monocytes/metabolism , Placenta/metabolism , Adult , Female , Fetal Blood/immunology , Fetus/immunology , Gene Expression , Humans , Inflammation/immunology , Lipopolysaccharide Receptors/metabolism , Monocytes/immunology , Obesity/immunology , Obesity/metabolism , Placenta/immunology , Pregnancy , Transcriptome
7.
Obesity (Silver Spring) ; 19(3): 476-82, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20930711

ABSTRACT

Obese pregnant women develop severe insulin resistance and enhanced systemic and placental inflammation, suggesting associated modifications of endocrine and immune functions. Activation of innate immunity by endotoxins/lipopolysaccharides (LPS) has been proposed as a mechanism for enhancing metabolic alterations in disorders with insulin resistance. The aim of this study was to characterize the immune responses developed by the adipose tissue (AT) and their potential links to maternal endotoxemia in pregnancy with obesity. Blood and subcutaneous abdominal AT were obtained from 120 lean and obese women (term pregnancy) recruited at delivery. Gene expression was assessed in AT and stromal vascular cells isolated from a subset of 24 subjects from the same cohort. Doubling of plasma endotoxin concentrations indicated subclinical endotoxemia in obese compared with lean women. This was associated with significant increase in systemic C-reactive protein and interleukin-6 (IL-6) but not tumor necrosis factor-α (TNF-α) concentrations. AT inflammation was characterized by accumulation of CD68(+) macrophages with a threefold increased gene expression of the macrophage markers CD68, EMR1, and CD14. Gene expression for cytokines IL-6, TNF-α, IL-8, and monocyte chemotactic protein-1 (MCP1) and for LPS-sensing CD14, toll-like receptor 4 (TLR4), translocating chain-associated membrane protein 2 was 2.5-5-fold higher in stromal cells of obese compared to lean. LPS-treated cultured stromal cells of obese women expressed a 5-16-fold stimulation of the same cytokines upregulated in vivo. Our data demonstrate that subclinical endotoxemia is associated with systemic and AT inflammation in obese pregnant women. Recognition of bacterial pathogens may contribute to the combined dysfunction of innate immunity and the metabolic systems in AT.


Subject(s)
Adipose Tissue/immunology , Endotoxemia/immunology , Inflammation Mediators/metabolism , Inflammation/immunology , Obesity/immunology , Pregnancy Complications/immunology , Adipose Tissue/metabolism , Adult , Antigens, CD/metabolism , C-Reactive Protein/metabolism , Chemokine CCL2/metabolism , Cytokines/metabolism , Endotoxemia/etiology , Endotoxemia/metabolism , Female , Humans , Immunity, Innate , Inflammation/etiology , Inflammation/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Obesity/complications , Obesity/metabolism , Pregnancy , Pregnancy Complications/metabolism , Toll-Like Receptor 4/metabolism , Young Adult
8.
Am J Obstet Gynecol ; 201(2): 209.e1-209.e10, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19560108

ABSTRACT

OBJECTIVE: Changes in metabolic homeostasis in pregnant diabetic women are potential determinants of increased adiposity of the fetus. The aim of this study was to characterize diabetes mellitus-induced changes in genes for fetoplacental energy metabolism in relation to fetal adiposity. STUDY DESIGN: Placentas of women with type 1 diabetes mellitus, gestational diabetes mellitus (GDM), or no complications were analyzed by microarray profiling. The pattern of gene expression was assessed in primary placental cell cultures. RESULTS: Diabetes mellitus was associated with 49 alterations in gene expression at key steps in placental energy metabolism, with 67% of the alterations related to lipid pathways and 9% of the alterations related to glucose pathways. Preferential activation of lipid genes was observed in pregnancy with GDM. Type 1 diabetes mellitus induced fewer lipid modifications but an enhancement of glycosylation and acylation pathways. Oleate enhanced expression of genes for fatty acid esterification and the formation of lipid droplets 3 times as much as glucose in cultured placental cells. CONCLUSION: These results point to fatty acids as preferential lipogenic substrates for placental cells and suggest that genes for fetoplacental lipid metabolism are enhanced selectively in GDM. The recruited genes may be instrumental in increasing transplacental lipid fluxes and the delivery of lipid substrates for fetal use.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Gene Expression Profiling , Lipid Metabolism/genetics , Adipose Tissue/physiology , Adult , Blood Glucose/genetics , Blood Glucose/metabolism , Cells, Cultured , Energy Metabolism/genetics , Female , Fetus/physiology , Gene Expression Regulation, Developmental/physiology , Homeostasis/genetics , Humans , Obesity/genetics , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Placenta/cytology , Placenta/physiology , Pregnancy
9.
Obesity (Silver Spring) ; 17(6): 1144-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19197253

ABSTRACT

Circulating adiponectin reflects the degree of energy homeostasis and insulin sensitivity of adult individuals. Low abundance of the high molecular weight (HMW) multimers, the most active forms mediating the insulin-sensitizing effects of adiponectin, is indicative of impaired metabolic status. The increase in fetal adiponectin HMW compared with adults is a distinctive features of human neonates. To further understand the functional properties of adiponectin during fetal life, we have evaluated the associations of adiponectin with insulin sensitivity, body composition, and gender. Umbilical cord adiponectin, adiponectin complexes, and metabolic parameters were measured at term by elective cesarean delivery. The associations between adiponectin, measures of body composition, and insulin sensitivity were evaluated in relation to fetal gender in 121 singleton neonates. Higher total adiponectin concentrations in female compared with male fetuses (34.3+/-9.5 vs. 24.9+/-8.6, P<0.001) were associated with a 3.2-fold greater abundance in circulating HMW complexes (0.20+/-0.03 vs. 0.08+/-0.03, P<0.001, n=9). Adiponectin was positively correlated with neonatal fat mass (r=0.27, P<0.04) and percent body fat in female fetuses (r=0.28, P<0.03) and with lean mass in males (r=0.28, P<0.03). There was no significant correlation between cord adiponectin and fasting insulin concentrations or fetal insulin sensitivity as estimated by homeostasis model assessment of insulin resistance (HOMA-IR). The gender dimorphism for plasma adiponectin concentration and complex distribution first appears in utero. In sharp contrast to the inverse correlation found in adults, the positive relationship between adiponectin and body fat is a specific feature of the fetus.


Subject(s)
Adiposity , Diabetes, Gestational/blood , Fetal Blood/metabolism , Fetus/metabolism , Insulin Resistance , Adiponectin/blood , Adult , Cesarean Section , Diabetes, Gestational/physiopathology , Female , Fetus/physiopathology , Gestational Age , Humans , Male , Molecular Weight , Pregnancy , Sex Factors , Young Adult
10.
Obesity (Silver Spring) ; 16(8): 1831-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18535540

ABSTRACT

BACKGROUND: Hyperprolactinemia is associated with obesity. Furthermore, in human adipose tissue cultured in vitro, prolactin (PRL) inhibited lipoprotein lipase (LPL) activity via functional PRL receptors. OBJECTIVE: To study PRL and insulin ultradian rhythm and subcutaneous adipose tissue LPL mRNA and protein expressions in severely obese women before and after malabsorptive bariatric surgery. METHODS AND PROCEDURES: Seven severely obese, fertile women were studied twice, once before and the second time 1 year after bilio-pancreatic diversion (BPD), when the weight was stable for at least 3 months. Metabolizable energy intake and 24-h energy expenditure (EE) were measured. Fourier and PULSEFIT analyses were applied to 24-h hormonal time-series to study daily fluctuations and hormonal clearance. Insulin sensitivity was assessed by euglycemic-hyperinsulinemic clamp. Quantitative-competitive reverse transcriptase-PCR and western blot analysis were used to measure LPL gene expression. RESULTS: Spontaneous 24-h PRL secretion was significantly reduced after BPD (mean-daily release, 128.4 +/- 28.1 microg/l vs. 67.2 +/- 9.2 microg/l distribution volume (Vd/l.24 h), P = 0.02); insulin secretion also was significantly reduced (499.9 +/- 204.0 microg/Vd/l.24 h vs. 85.6 +/- 21.0 microg/Vd/l.24 h, P = 0.0001). Metabolizable energy/kg(FFM) did not change significantly after BPD. Twenty-four-hour EE, but not 24-h EE/FFM, was significantly decreased after BPD (P < 0.05). Insulin sensitivity significantly (P < 0.0001) increased after BPD from 21.41 +/- 1.92 to 68.62 +/- 5.03 micromol/kg(FFM)/min. LPL mRNA concentration (from 42.63 +/- 4.21% to 19.00 +/- 2.74% of cyclophilin mRNA, P = 0.001) as well as LPL protein level (from 8.94 +/- 2.73 to 3.16 +/- 1.05 as ratios of protein of interest vs. housekeeping protein, P = 0.038) significantly decreased after BPD. The major determinant of PRL secretion was insulin secretion, whereas the best predictors of LPL expression were insulin and PRL secretion rates. DISCUSSION: The restriction of lipid metabolizable energy rather than weight loss seems to be responsible for both reduction in PRL circulating levels and normalization of its secretion rhythm after bariatric surgery. Furthermore, the reduced adipose tissue LPL expression, being significantly correlated with the decrease in insulin and PRL, suggests a role of hyperinsulinemia and hyperprolactinemia in inducing and sustaining obesity.


Subject(s)
Adipose Tissue/enzymology , Circadian Rhythm/physiology , Insulin/metabolism , Lipoprotein Lipase/metabolism , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Prolactin/metabolism , Adipose Tissue/pathology , Adult , Bariatric Surgery , Biopsy , Energy Metabolism/physiology , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follow-Up Studies , Fourier Analysis , Humans , Insulin/blood , Insulin Resistance/physiology , Insulin Secretion , Lipoprotein Lipase/genetics , Prolactin/blood , RNA, Messenger/metabolism , Sex Hormone-Binding Globulin/metabolism
11.
Mol Biol Cell ; 18(9): 3302-12, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17567949

ABSTRACT

The principal infectious and pathogenic agent in all prion disorders is a beta-sheet-rich isoform of the cellular prion protein (PrP(C)) termed PrP-scrapie (PrP(Sc)). Once initiated, PrP(Sc) is self-replicating and toxic to neuronal cells, but the underlying mechanisms remain unclear. In this report, we demonstrate that PrP(C) binds iron and transforms to a PrP(Sc)-like form (*PrP(Sc)) when human neuroblastoma cells are exposed to an inorganic source of redox iron. The *PrP(Sc) thus generated is itself redox active, and it induces the transformation of additional PrP(C), simulating *PrP(Sc) propagation in the absence of brain-derived PrP(Sc). Moreover, limited depletion of iron from prion disease-affected human and mouse brain homogenates and scrapie-infected mouse neuroblastoma cells results in 4- to 10-fold reduction in proteinase K (PK)-resistant PrP(Sc), implicating redox iron in the generation, propagation, and stability of PK-resistant PrP(Sc). Furthermore, we demonstrate increased redox-active ferrous iron levels in prion disease-affected brains, suggesting that accumulation of PrP(Sc) is modulated by the combined effect of imbalance in brain iron homeostasis and the redox-active nature of PrP(Sc). These data provide information on the mechanism of replication and toxicity by PrP(Sc), and they evoke predictable and therapeutically amenable ways of modulating PrP(Sc) load.


Subject(s)
Brain/pathology , Endopeptidase K/metabolism , Iron/pharmacology , PrPC Proteins/metabolism , PrPC Proteins/pathogenicity , Prion Diseases/metabolism , Tissue Extracts/metabolism , Aged , Animals , Brain/drug effects , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Chlorides , Deferoxamine/pharmacology , Ferric Compounds/pharmacology , Ferritins/metabolism , Humans , Iron Deficiencies , Mice , Middle Aged , Oxidation-Reduction/drug effects , PrPSc Proteins/metabolism , Protein Conformation/drug effects
12.
Mol Cell Biol ; 26(7): 2697-715, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16537913

ABSTRACT

The C-transmembrane form of prion protein ((Ctm)PrP) has been implicated in prion disease pathogenesis, but the factors underlying its biogenesis and cyotoxic potential remain unclear. Here we show that (Ctm)PrP interferes with cytokinesis in cell lines where it is transported to the plasma membrane. These cells fail to separate following cell division, assume a variety of shapes and sizes, and contain multiple nuclei, some of which are pyknotic. Furthermore, the synthesis and transport of (Ctm)PrP to the plasma membrane are modulated through a complex interaction between cis- and trans-acting factors and the endoplasmic reticulum translocation machinery. Thus, insertion of eight amino acids before or within the N region of the N signal peptide (N-SP) of PrP results in the exclusive synthesis of (Ctm)PrP regardless of the charge conferred to the N region. Subsequent processing and transport of (Ctm)PrP are modulated by specific amino acids in the N region of the N-SP and by the cell line of expression. Although the trigger for (Ctm)PrP upregulation in naturally occurring prion disorders remains elusive, these data highlight the underlying mechanisms of (Ctm)PrP biogenesis and neurotoxicity and reinforce the idea that (Ctm)PrP may serve as the proximate cause of neuronal death in certain prion disorders.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Prions/metabolism , Prions/pathogenicity , Animals , CHO Cells , Cricetinae , Cricetulus , Cytokinesis , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/ultrastructure , Mice , Prions/chemistry , Prions/ultrastructure , Protein Processing, Post-Translational , Protein Transport , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism
13.
J Neurosci ; 24(50): 11280-90, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15601934

ABSTRACT

Foodborne transmission of bovine spongiform encephalopathy (BSE) to humans as variant Creutzfeldt-Jakob disease (CJD) has affected over 100 individuals, and probably millions of others have been exposed to BSE-contaminated food substances. Despite these obvious public health concerns, surprisingly little is known about the mechanism by which PrP-scrapie (PrP(Sc)), the most reliable surrogate marker of infection in BSE-contaminated food, crosses the human intestinal epithelial cell barrier. Here we show that digestive enzyme (DE) treatment of sporadic CJD brain homogenate generates a C-terminal fragment similar to the proteinase K-resistant PrP(Sc) core of 27-30 kDa implicated in prion disease transmission and pathogenesis. Notably, DE treatment results in a PrP(Sc)-protein complex that is avidly transcytosed in vesicular structures across an in vitro model of the human intestinal epithelial cell barrier, regardless of the amount of endogenous PrP(C) expression. Unexpectedly, PrP(Sc) is cotransported with ferritin, a prominent component of the DE-treated PrP(Sc)-protein complex. The transport of PrP(Sc)-ferritin is sensitive to low temperature, brefeldin-A, and nocodazole treatment and is inhibited by excess free ferritin, implicating a receptor- or transporter-mediated pathway. Because ferritin shares considerable homology across species, these data suggest that PrP(Sc)-associated proteins, in particular ferritin, may facilitate PrP(Sc) uptake in the intestine from distant species, leading to a carrier state in humans.


Subject(s)
Ferritins/metabolism , Intestinal Absorption , PrPSc Proteins/metabolism , Aged , Brain/metabolism , Caco-2 Cells , Creutzfeldt-Jakob Syndrome/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Multiprotein Complexes/metabolism , Pepsin A/metabolism , Peptide Fragments/metabolism , Peptide Hydrolases/metabolism , PrPC Proteins/metabolism , Protein Binding , Protein Transport , Species Specificity , Tight Junctions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...