Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Mol Cell Cardiol ; 112: 114-122, 2017 11.
Article in English | MEDLINE | ID: mdl-28478047

ABSTRACT

BACKGROUND: Patients with chronic kidney disease (CKD) experience high rates of atherosclerotic cardiovascular disease and death that are not fully explained by traditional risk factors. In animal studies, defective cellular cholesterol efflux pathways which are mediated by the ATP binding cassette transporters ABCA1 and ABCG1 are associated with accelerated atherosclerosis. We hypothesized that cholesterol efflux in humans would vary in terms of cellular components, with potential implications for cardiovascular disease. METHODS: We recruited 120 CKD patients (eGFR<30mL/min/1.73m2) and 120 control subjects (eGFR ≥60mL/min/1.73m2) in order to measure cholesterol efflux using either patients' HDL and THP-1 macrophages or patients' monocytes and a flow cytometry based cholesterol efflux assay. We also measured cell-surface levels of the common ß subunit of the IL-3/GM-CSF receptor (IL-3Rß) which has been linked to defective cholesterol homeostasis and may promote monocytosis. In addition, we measured plasma inflammatory cytokines and plasma metabolite profiles. RESULTS: There was a strong positive correlation between cell-surface IL-3Rß levels and monocyte counts in CKD (P<0.001). ABCA1 mRNA was reduced in CKD vs. control monocytes (P<0.05), across various etiologies of CKD. Cholesterol efflux to apolipoprotein A1 was impaired in monocytes from CKD patients with diabetic nephropathy (P<0.05), but we found no evidence for a circulating HDL-mediated defect in cholesterol efflux in CKD. Profiling of plasma metabolites showed that medium-chain acylcarnitines were both independently associated with lower levels of cholesterol transporter mRNA in CKD monocytes at baseline (P<0.05), and with cardiovascular events in CKD patients after median 2.6years of follow-up. CONCLUSIONS: Cholesterol efflux in humans varies in terms of cellular components. We report a cellular defect in ABCA1-mediated cholesterol efflux in monocytes from CKD patients with diabetic nephropathy. Unlike several traditional risk factors for atherosclerotic cardiovascular disease, plasma metabolites inversely associated with endogenous cholesterol transporters predicted cardiovascular events in CKD patients. (Funded by the National Institute of Diabetes and Digestive and Kidney DiseasesK23DK097288 and others.).


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/epidemiology , Cholesterol/metabolism , Metabolome , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , ATP Binding Cassette Transporter 1/metabolism , Aged , Biological Transport , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Carnitine/analogs & derivatives , Carnitine/metabolism , Cell Line , Cytokine Receptor Common beta Subunit/metabolism , Diabetic Nephropathies/blood , Diabetic Nephropathies/complications , Diabetic Nephropathies/metabolism , Female , Follow-Up Studies , Humans , Male , Middle Aged , Monocytes/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Risk Factors
2.
Mol Autism ; 6: 57, 2015.
Article in English | MEDLINE | ID: mdl-26500752

ABSTRACT

BACKGROUND: Platelets have been proven to be a useful cellular model to study some neuropathologies, due to the overlapping biological features between neurons and platelets as granule secreting cells. Altered platelet dense granule morphology was previously reported in three autism spectrum disorder (ASD) patients with chromosomal translocations that disrupted ASD candidate genes NBEA, SCAMP5, and AMYSIN, but a systematic analysis of platelet function in ASD is lacking in contrast to numerous reports of elevated serotonin levels in platelets and blood as potential biomarker for ASD. METHODS: We explored platelet count, size, epinephrine-induced activation, and dense granule ATP secretion in a cohort of 159 ASD patients, their 289 first-degree relatives (103 unaffected siblings, 99 mothers, and 87 fathers), 45 adult controls, and 65 pediatric controls. For each of the responses separately, a linear mixed model with gender as a covariate was used to compare the level between groups. We next investigated the correlation between platelet function outcomes and severity of impairments in social behavior (social responsiveness score (SRS)). RESULTS: The average platelet count was increased in ASD patients and siblings vs. controls (ASD 320.3 × 10(9)/L, p = 0.003; siblings 332.0 × 10(9)/L, p < 0.001; controls 283.0 × 10(9)/L). The maximal platelet secretion-dependent aggregation response to epinephrine was not significantly lower for ASD patients. However, secondary wave responses following stimulation with epinephrine were more frequently delayed or absent compared to controls (ASD 52 %, siblings 45 %, parents 53 %, controls 22 %, p = 0.002). In addition, stimulated release of ATP from dense granules was reduced in ASD patients, siblings, and parents vs. controls following activation of platelets with either collagen (ASD 1.54 µM, p = 0.001; siblings 1.51 µM, p < 0.001; parents 1.67 µM, p = 0.021; controls 2.03 µM) or ADP (ASD 0.96 µM, p = 0.003; siblings 1.00 µM, p = 0.012; parents 1.17 µM, p = 0.21; controls 1.40 µM). Plasma serotonin levels were increased for ASD patients (n = 20, p = 0.005) and siblings (n = 20, p = 0.0001) vs. controls (n = 16). No significant correlations were found in the different groups between SRS scores and count, size, epinephrine aggregation, or ATP release. CONCLUSIONS: We report increased platelet counts, decreased platelet ATP dense granule secretion, and increased serotonin plasma levels not only in ASD patients but also in their first-degree relatives. This suggests that potential genetic factors associated with platelet counts and granule secretion can be associated with, but are not fully penetrant for ASD.

3.
Arterioscler Thromb Vasc Biol ; 34(4): 751-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24504733

ABSTRACT

OBJECTIVE: The ATP-binding cassette (ABC) transporter B6 (ABCB6) is highly expressed in megakaryocyte progenitors, but its role in platelet production and disease has not been elucidated. APPROACH AND RESULTS: Among various ABC transporters, ABCB6 was highly expressed in megakaryocyte progenitors, exhibiting the same pattern of expression of genes involved in heme synthesis pathway. Transplantation of Abcb6 deficient (Abcb6(-/-)) bone marrow into low density lipoprotein receptor deficient recipient mice resulted in expansion and proliferation of megakaryocyte progenitors, attributable to increased reactive oxygen species production in response to porphyrin loading. The enhanced megakaryopoiesis in Abcb6(-/-) bone marrow-transplanted mice was further illustrated by increased platelet counts, mean platelet volume, and platelet activity. Platelets from Abcb6(-/-) bone marrow-transplanted mice had higher levels of chemokine (C-C motif) ligand 5, which was associated with increased plasma chemokine (C-C motif) ligand 5 levels. There were also increased platelet-leukocyte aggregates, which resulted in leukocyte activation. Abcb6(-/-) bone marrow-transplanted mice had accelerated atherosclerosis which was associated with deposition of the chemotactic agent, chemokine (C-C motif) ligand 5 in atherosclerotic plaques, resulting in increased macrophage accumulation. CONCLUSIONS: Our findings identify a new role of ABCB6 in preventing atherosclerosis development by dampening platelet production, reactivity, and chemokine (C-C motif) ligand 5 deposition in atherosclerotic lesions.


Subject(s)
ATP-Binding Cassette Transporters/deficiency , Atherosclerosis/metabolism , Blood Platelets/metabolism , Megakaryocyte Progenitor Cells/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Bone Marrow Transplantation , Cell Proliferation , Cells, Cultured , Chemokine CCL5/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Reactive Oxygen Species/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Thrombopoiesis , Time Factors
4.
Nat Med ; 19(5): 586-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23584088

ABSTRACT

Platelets have a key role in atherogenesis and its complications. Both hypercholesterolemia and increased platelet production promote atherothrombosis; however, a potential link between altered cholesterol homeostasis and platelet production has not been explored. Here we show that transplantation of bone marrow deficient in ABCG4, a transporter of unknown function, into Ldlr(-/-) mice resulted in thrombocytosis, accelerated thrombosis and atherosclerosis. Although not detected in atherosclerotic lesions, Abcg4 was highly expressed in bone marrow megakaryocyte progenitors (MkPs). Abcg4(-/-) MkPs had defective cholesterol efflux to high-density lipoprotein (HDL), increased cell surface expression of the thrombopoietin (TPO) receptor (c-MPL) and enhanced proliferation. These consequences of ABCG4 deficiency seemed to reflect disruption of negative feedback regulation of c-MPL signaling by the E3 ligase c-CBL and the cholesterol-sensing LYN kinase. HDL infusion reduced platelet counts in Ldlr(-/-) mice and in a mouse model of myeloproliferative neoplasm in an ABCG4-dependent fashion. HDL infusions may offer a new approach to reducing atherothrombotic events associated with increased platelet production.


Subject(s)
Blood Platelets/cytology , Cholesterol/metabolism , Megakaryocytes/cytology , Stem Cells/cytology , Thrombocytosis/metabolism , ATP Binding Cassette Transporter, Subfamily G , ATP-Binding Cassette Transporters/genetics , Animals , Cell Proliferation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Receptors, Thrombopoietin/metabolism , Thrombosis/pathology , Ubiquitin-Protein Ligases/metabolism , src-Family Kinases/metabolism
5.
Metabolism ; 61(1): 99-107, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21816446

ABSTRACT

Obesity and its associated conditions such as type 2 diabetes mellitus are major causes of morbidity and mortality. The iminosugar N-(5-adamantane-1-yl-methoxy-pentyl)-deoxynojirimycin (AMP-DNM) improves insulin sensitivity in rodent models of insulin resistance and type 2 diabetes mellitus. In the current study, we characterized the impact of AMP-DNM on substrate oxidation patterns, food intake, and body weight gain in obese mice. Eight ob/ob mice treated with 100 mg/(kg d) AMP-DNM mixed in the food and 8 control ob/ob mice were placed in metabolic cages during the first, third, and fifth week of the experiment for measurement of substrate oxidation rates, energy expenditure, activity, and food intake. Mice were killed after 6 weeks of treatment. Initiation of treatment with AMP-DNM resulted in a rapid increase in fat oxidation by 129% (P = .05), a decrease in carbohydrate oxidation by 35% (P = .01), and a reduction in food intake by approximately 26% (P < .01) compared with control mice. Treatment with AMP-DNM decreased hepatic triglyceride content by 66% (P < .01) and, in line with the elevated fat oxidation rates, increased hepatic carnitine palmitoyl transferase 1a expression. Treatment with AMP-DNM increased plasma levels of the appetite-regulating peptide YY compared with control mice. Treatment with AMP-DNM rapidly reduces food intake and increases fat oxidation, resulting in improvement of the obese phenotype. These features of AMP-DNM, together with its insulin-sensitizing capacity, make it an attractive candidate drug for the treatment of obesity and its associated metabolic derangements.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Adamantane/analogs & derivatives , Body Weight/drug effects , Eating/drug effects , Obesity/drug therapy , Obesity/metabolism , 1-Deoxynojirimycin/pharmacology , Adamantane/pharmacology , Adipose Tissue/metabolism , Animals , Carbohydrate Metabolism/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Ghrelin/metabolism , Glucose/metabolism , Imino Sugars/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Oxidation-Reduction , Peptide YY/metabolism , Triglycerides/metabolism , Up-Regulation/drug effects , Weight Gain/drug effects
6.
J Clin Invest ; 121(10): 4138-49, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21968112

ABSTRACT

Leukocytosis is associated with increased cardiovascular disease risk in humans and develops in hypercholesterolemic atherosclerotic animal models. Leukocytosis is associated with the proliferation of hematopoietic stem and multipotential progenitor cells (HSPCs) in mice with deficiencies of the cholesterol efflux-promoting ABC transporters ABCA1 and ABCG1 in BM cells. Here, we have determined the role of endogenous apolipoprotein-mediated cholesterol efflux pathways in these processes. In Apoe⁻/⁻ mice fed a chow or Western- type diet, monocytosis and neutrophilia developed in association with the proliferation and expansion of HSPCs in the BM. In contrast, Apoa1⁻/⁻ mice showed no monocytosis compared with controls. ApoE was found on the surface of HSPCs, in a proteoglycan-bound pool, where it acted in an ABCA1- and ABCG1-dependent fashion to decrease cell proliferation. Accordingly, competitive BM transplantation experiments showed that ApoE acted cell autonomously to control HSPC proliferation, monocytosis, neutrophilia, and monocyte accumulation in atherosclerotic lesions. Infusion of reconstituted HDL and LXR activator treatment each reduced HSPC proliferation and monocytosis in Apoe⁻/⁻ mice. These studies suggest a specific role for proteoglycanbound ApoE at the surface of HSPCs to promote cholesterol efflux via ABCA1/ABCG1 and decrease cell proliferation, monocytosis, and atherosclerosis. Although endogenous apoA-I was ineffective, pharmacologic approaches to increasing cholesterol efflux suppressed stem cell proliferative responses.


Subject(s)
Apolipoproteins E/physiology , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Monocytes/pathology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/deficiency , Apolipoprotein A-I/genetics , Apolipoprotein A-I/physiology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cell Proliferation , Cholesterol/metabolism , Disease Models, Animal , Humans , Leukocytosis/pathology , Leukocytosis/physiopathology , Lipoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Proteoglycans/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/physiology
7.
Adv Exp Med Biol ; 721: 99-119, 2011.
Article in English | MEDLINE | ID: mdl-21910085

ABSTRACT

Glycosphingolipids are structural membrane components, residing largely in the plasma membrane with their sugar-moieties exposed at the cell's surface. In recent times a crucial role for glycosphingolipids in insulin resistance has been proposed. A chronic state of insulin resistance is a rapidly increasing disease condition in Western and developing countries. It is considered to be the major underlying cause of the metabolic syndrome, a combination of metabolic abnormalities that increases the risk for an individual to develop Type 2 diabetes, obesity, cardiovascular disease, polycystic ovary syndrome and nonalcoholic fatty liver disease. As discussed in this chapter, the evidence for a direct regulatory interaction of glycosphingolipids with insulin signaling is still largely indirect. However, the recent finding in animal models that pharmacological reduction of glycosphingolipid biosynthesis ameliorates insulin resistance and prevents some manifestations of metabolic syndrome, supports the view that somehow glycosphingolipids act as critical regulators, Importantly, since reductions in glycosphingolipid biosynthesis have been found to be well tolerated, such approaches may have a therapeutic potential.


Subject(s)
Glycosphingolipids/metabolism , Insulin Resistance , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Adamantane/analogs & derivatives , Adamantane/therapeutic use , Animals , Cardiovascular Diseases/metabolism , Ceramides/metabolism , Ceramides/toxicity , Diabetes Mellitus, Type 2/metabolism , Dioxanes/therapeutic use , Disease Models, Animal , Fatty Acids/pharmacokinetics , Fatty Liver/metabolism , Gaucher Disease/drug therapy , Gaucher Disease/genetics , Gaucher Disease/metabolism , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/physiology , Humans , Insulin Resistance/physiology , Metabolic Syndrome/metabolism , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease , Obesity/metabolism , Pyrrolidines/therapeutic use , Receptor, Insulin/chemistry , Receptor, Insulin/physiology , Signal Transduction
8.
Hepatology ; 52(2): 656-66, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20683963

ABSTRACT

UNLABELLED: De novo bile acid synthesis in the liver needs to be tightly regulated in order to maintain optimal bile flow and prevent cholestasis. In the liver, fibroblast growth factor 19 (FGF19) regulates bile acid synthesis by down-regulating messenger RNA levels of cytochrome P450 7A1 (CYP7A1). FGF19 acts through fibroblast growth factor receptor 4 (FGFR4), and beta-Klotho has recently been recognized as a modulator of FGFR4 activity. However, its precise mechanism of action has not been thoroughly described. We show here that beta-Klotho is an endoplasmic reticulum-resident protein that affects the cellular abundance of different FGFR4 glycoforms. beta-Klotho binds and directs the core glycoform of FGFR4 to the proteasome, and it allows only a terminal glycoform to reach the plasma membrane. Only the terminal FGFR4 glycoform is phosphorylated upon FGF19 treatment of HepG2 cells, and this shows that only fully glycosylated FGFR4 is active in CYP7A1 down-regulation. CONCLUSION: beta-Klotho enhances FGF19 signaling by binding the inactive, core-glycosylated FGFR4 and preventing it from reaching the surface. These results indicate that beta-Klotho is an indirect regulator of FGFR4, whereas glycosylation is the master switch for FGF19 activity and regulation of bile acid synthesis.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Down-Regulation , Fibroblast Growth Factors/physiology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Glucuronidase/physiology , Glycosylation , Humans , Klotho Proteins , Phosphorylation
9.
J Med Chem ; 53(2): 689-98, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-20000679

ABSTRACT

The lipophilic iminosugar N-[5-(adamantan-1-ylmethoxy)pentyl]-1-deoxynojirimycin (2, AMP-DNM) potently controls hyperglycemia in obese rodent models of insulin resistance. The reduction of visceral glycosphingolipids by 2 is thought to underlie its beneficial action. It cannot, however, be excluded that concomitant inhibition of intestinal glycosidases and associated buffering of carbohydrate assimilation add to this. To firmly establish the mode of action of 2, we developed a panel of lipophilic iminosugars varying in configuration at C-4/C-5 and N-substitution of the iminosugar. From these we identified the l-ido derivative of 2, l-ido-AMP-DNM (4), as a selective inhibitor of glycosphingolipid synthesis. Compound 4 lowered visceral glycosphingolipids in ob/ob mice and ZDF rats on a par with 2. In contrast to 2, 4 did not inhibit sucrase activity or sucrose assimilation. Treatment with 4 was significantly less effective in reducing blood glucose and HbA1c. We conclude that the combination of reduction of glycosphingolipids in tissue and buffering of carbohydrate assimilation by 2 produces a superior glucose homeostasis.


Subject(s)
Blood Glucose/drug effects , Carbohydrate Metabolism/drug effects , Glycosphingolipids/metabolism , Imino Sugars/pharmacology , Obesity/drug therapy , Absorption/drug effects , Animals , Glycated Hemoglobin/drug effects , Imino Sugars/chemistry , Imino Sugars/therapeutic use , Mice , Mice, Obese , Rats , Rats, Zucker , Structure-Activity Relationship , Viscera/metabolism
10.
Hepatology ; 50(5): 1431-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19731235

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is associated with obesity, insulin resistance, and type 2 diabetes. The hyperinsulinemia that occurs as a consequence of insulin resistance is thought to be an important contributor to the development of fatty liver. We have shown that the iminosugar N-(5'-adamantane-1'-yl-methoxy)-pentyl-1-deoxynojirimycin (AMP-DNM), an inhibitor of the enzyme glucosylceramide synthase, is a potent enhancer of insulin signaling in rodent models for insulin resistance and type 2 diabetes. The present study was designed to assess the impact of AMP-DNM on insulin levels, liver triglyceride synthesis, and gene expression profile. Treatment of ob/ob mice with AMP-DNM restored insulin signaling in the liver, corrected blood glucose values to levels found in lean mice, and decreased insulin concentration. The expression of sterol regulatory element-binding protein 1c target genes involved in fatty acid synthesis normalized. AMP-DNM treatment significantly reduced liver to body weight ratio and reversed hepatic steatosis, comprising fat as well as inflammatory markers. In addition, AMP-DNM treatment corrected to a large extent the gene expression profile of ob/ob mice livers toward the profile of lean mice. CONCLUSION: Pharmacological lowering of glycosphingolipids with the iminosugar AMP-DNM is a promising approach to restore insulin signaling and improve glucose homeostasis as well as hepatic steatosis.


Subject(s)
Fatty Liver/metabolism , Glycosphingolipids/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Liver/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Adamantane/analogs & derivatives , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Disease Models, Animal , Fatty Liver/drug therapy , Glucose/metabolism , Glycosphingolipids/antagonists & inhibitors , Homeostasis/drug effects , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Triglycerides/metabolism
11.
PLoS One ; 4(3): e4723, 2009.
Article in English | MEDLINE | ID: mdl-19305508

ABSTRACT

Adipose tissue is a critical mediator in obesity-induced insulin resistance. Previously we have demonstrated that pharmacological lowering of glycosphingolipids and subsequently GM3 by using the iminosugar AMP-DNM, strikingly improves glycemic control. Here we studied the effects of AMP-DNM on adipose tissue function and inflammation in detail to provide an explanation for the observed improved glucose homeostasis. Leptin-deficient obese (Lep(Ob)) mice were fed AMP-DNM and its effects on insulin signalling, adipogenesis and inflammation were monitored in fat tissue. We show that reduction of glycosphingolipid biosynthesis in adipose tissue of Lep(Ob) mice restores insulin signalling in isolated ex vivo insulin-stimulated adipocytes. We observed improved adipogenesis as the number of larger adipocytes was reduced and expression of genes like peroxisome proliferator-activated receptor (PPAR) gamma, insulin responsive glucose transporter (GLUT)-4 and adipsin increased. In addition, we found that adiponectin gene expression and protein were increased by AMP-DNM. As a consequence of this improved function of fat tissue we observed less inflammation, which was characterized by reduced numbers of adipose tissue macrophages (crown-like structures) and reduced levels of the macrophage chemo attractants monocyte-chemoattractant protein-1 (Mcp-1/Ccl2) and osteopontin (OPN). In conclusion, pharmacological lowering of glycosphingolipids by inhibition of glucosylceramide biosynthesis improves adipocyte function and as a consequence reduces inflammation in adipose tissue of obese animals.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Adamantane/analogs & derivatives , Adipogenesis/physiology , Adipose Tissue/metabolism , Glycosphingolipids/metabolism , Inflammation/metabolism , Insulin Resistance/physiology , Mice, Obese , 1-Deoxynojirimycin/metabolism , Adamantane/metabolism , Adiponectin/metabolism , Adipose Tissue/cytology , Animals , Chemokine CCL2/metabolism , Glucose/metabolism , Homeostasis , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
12.
Hepatology ; 49(2): 637-45, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19072830

ABSTRACT

UNLABELLED: Recent reports indicate that glycosphingolipids play an important role in regulation of carbohydrate metabolism. We have shown that the iminosugar N-(5'-adamantane-1'-yl-methoxy)-pentyl-1-deoxynojirimycin (AMP-DNM), an inhibitor of the enzyme glucosylceramide synthase, is a potent enhancer of insulin signaling in rodent models for insulin resistance and type 2 diabetes. In this study, we determined whether AMP-DNM also affects lipid homeostasis and, in particular, the reverse cholesterol transport pathway. Treatment of C57BL/6J mice with AMP-DNM for 5 weeks decreased plasma levels of triglycerides and cholesterol by 35%, whereas neutral sterol excretion increased twofold. Secretion of biliary lipid also increased twofold, which resulted in a similar rise in bile flow. This effect was not due to altered expression levels or kinetics of the various export pumps involved in bile formation. However, the bile salt pool size increased and the expression of Cyp7A1 was up-regulated. In vitro experiments using HepG2 hepatoma cell line revealed this to be due to inhibition of fibroblast growth factor-19 (FGF19)-mediated suppression of Cyp7A1 via the FGF receptor. CONCLUSION: Pharmacological modulation of glycosphingolipid metabolism showed surprising effects on lipid homeostasis in C57BL/6J mice. Upon administration of 100 mg AMP-DNM/kg body weight/day, plasma cholesterol and triglyceride levels decreased, biliary lipid secretion doubled and also the endpoint of reverse cholesterol transport, neutral sterol excretion, doubled.


Subject(s)
Bile/metabolism , Gallbladder/metabolism , Glycosphingolipids/biosynthesis , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Cell Line, Tumor , Cells, Cultured , Cholesterol/blood , Cholesterol, HDL/metabolism , Gallbladder/physiology , Gangliosides/physiology , Glycosphingolipids/antagonists & inhibitors , Humans , Lipid Metabolism , Lipoproteins, HDL/metabolism , Liver Neoplasms , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Signal Transduction , Triglycerides/blood
13.
J Pharmacol Exp Ther ; 326(3): 849-55, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18550691

ABSTRACT

Recent findings have implicated glycosphingolipids as modulators of insulin receptor activity. Studies with C57BL/6J ob/ob mice have shown that insulin sensitivity is enhanced by the synthetic hydrophobic iminosugar N-(5-adamantane-1-yl-methoxy-pentyl)-deoxynojirimycin (AMP-DNM) that inhibits glucosylceramide synthase. Here, we treated the liver hepatoma cell line HepG2 with AMP-DNM, resulting in a 70% reduction of glycosphingolipids, and we analyzed the effect on gene expression. Using whole human genome 44K oligonucleotide arrays, we identified 89 genes that were significantly (p < 0.01) up- or down-regulated by AMP-DNM treatment. Of the 56 up-regulated genes, 17 were direct target genes for transcription factors sterol regulatory element-binding protein (SREBP) 1 or SREBP2, which activate genes in the sterol biosynthesis pathway. An increase in cholesterol production rate confirmed that the induction of SREBP target genes seen at the mRNA level resulted in activation of the cholesterol biosynthesis pathway. It is interesting to note that the cholesterol content of the cells did not increase. It is noteworthy that no effects were found on expression of genes related to cell receptor signaling pathways, neither on toxicity nor cell growth. Our findings indicate that inhibition of glucosylceramide synthase with AMP-DNM leads to activation of SREBP target genes and synthesis of cholesterol in HepG2 cells.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Adamantane/analogs & derivatives , Cholesterol/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Glucosyltransferases/antagonists & inhibitors , Sterol Regulatory Element Binding Proteins/biosynthesis , Sterol Regulatory Element Binding Proteins/genetics , 1-Deoxynojirimycin/pharmacology , Adamantane/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucosyltransferases/metabolism , Humans
14.
J Virol ; 80(14): 6982-92, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809304

ABSTRACT

Monoclonal antibodies (MAbs) neutralizing West Nile Virus (WNV) have been shown to protect against infection in animal models and have been identified as a correlate of protection in WNV vaccine studies. In the present study, antibody repertoires from three convalescent WNV-infected patients were cloned into an scFv phage library, and 138 human MAbs binding to WNV were identified. One hundred twenty-one MAbs specifically bound to the viral envelope (E) protein and four MAbs to the premembrane (prM) protein. Enzyme-linked immunosorbent assay-based competitive-binding assays with representative E protein-specific MAbs demonstrated that 24/51 (47%) bound to domain II while only 4/51 (8%) targeted domain III. In vitro neutralizing activity was demonstrated for 12 MAbs, and two of these, CR4374 and CR4353, protected mice from lethal WNV challenge at 50% protective doses of 12.9 and 357 mug/kg of body weight, respectively. Our data analyzing three infected individuals suggest that the human anti-WNV repertoire after natural infection is dominated by nonneutralizing or weakly neutralizing MAbs binding to domain II of the E protein, while domain III-binding MAbs able to potently neutralize WNV in vitro and in vivo are rare.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile virus/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Viral/genetics , Antibody Specificity/genetics , Antibody Specificity/immunology , Binding Sites, Antibody/genetics , Binding Sites, Antibody/immunology , Cloning, Molecular , Humans , Mice , Protein Structure, Tertiary
15.
Eur J Cancer ; 41(1): 178-87, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15618003

ABSTRACT

Tumour-associated cell surface markers are potential targets for antibody-based therapies. We have obtained a panel of myeloid cell binding single chain variable fragments (scFv) by applying phage display selection on myeloid cell lines followed by a selection round on freshly isolated acute myeloid leukaemia (AML) blasts using flow cytometry. To identify the target antigens, the scFv were recloned and expressed in an IgG(1) format and tested for their ability to immunoprecipitate cell surface proteins. The IgGs that reacted with distinct cell membrane extractable proteins were used in large-scale affinity purification of the target antigen followed by mass-spectrometry-based identification. Well-characterised cell surface antigens, such as leukocyte antigen-related receptor protein tyrosine phosphatase (LAR PTP) and activated leukocyte adhesion molecule (ALCAM) in addition to several unknown proteins, like ATAD3A, were identified. These experiments demonstrate that phage antibody selection in combination with affinity chromatography and mass spectrometry can be exploited successfully to identify novel antibody target molecules on malignant cells.


Subject(s)
Antigens, Neoplasm/analysis , Leukemia, Myeloid/genetics , Proteomics , Activated-Leukocyte Cell Adhesion Molecule , Acute Disease , Antigens, Surface/metabolism , Bacteriophages/metabolism , Cell Line, Tumor , Clone Cells , Flow Cytometry/methods , Humans , Immunoglobulin G/metabolism , Leukocytes, Mononuclear/metabolism , Mass Spectrometry , Myeloid Cells/metabolism , Transfection
16.
Cancer Res ; 64(22): 8443-50, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15548716

ABSTRACT

Acute myeloid leukemia (AML) has a poor prognosis due to treatment-resistant relapses. A humanized anti-CD33 antibody (Mylotarg) showed a limited response rate in relapsed AML. To discover novel AML antibody targets, we selected a panel of single chain Fv fragments using phage display technology combined with flow cytometry on AML tumor samples. One selected single chain Fv fragment broadly reacted with AML samples and with myeloid cell lineages within peripheral blood. Expression cloning identified the antigen recognized as C-type lectin-like molecule-1 (CLL-1), a previously undescribed transmembrane glycoprotein. CLL-1 expression was analyzed with a human anti-CLL-1 antibody that was generated from the single chain Fv fragment. CLL-1 is restricted to the hematopoietic lineage, in particular to myeloid cells present in peripheral blood and bone marrow. CLL-1 is absent on uncommitted CD34(+)/CD38(-) or CD34(+)/CD33(-) stem cells and present on subsets of CD34(+)/CD38(+) or CD34(+)/CD33(+) progenitor cells. CLL-1 is not expressed in any other tissue. In contrast, analysis of primary AMLs demonstrated CLL-1 expression in 92% (68 of 74) of the samples. As an AML marker, CLL-1 was able to complement CD33, because 67% (8 of 12) of the CD33(-) AMLs expressed CLL-1. CLL-1 showed variable expression (10-60%) in CD34(+) cells in chronic myelogenous leukemia and myelodysplastic syndrome but was absent in 12 of 13 cases of acute lymphoblastic leukemia. The AML reactivity combined with the restricted expression on normal cells identifies CLL-1 as a novel potential target for AML treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Lectins, C-Type/metabolism , Leukemia, Myeloid/metabolism , Acute Disease , Amino Acid Sequence , Base Sequence , Cell Membrane/metabolism , Flow Cytometry , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL