Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biosens Bioelectron ; 220: 114826, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36371959

ABSTRACT

Nicotinamide riboside (NR) is a form of vitamin B3 and is one of the most studied compounds for the restoration of cellular NAD+ levels demonstrating clinical potential in many metabolic and age-related disorders. Despite its wide commercial availability as a powerful nutraceutical, our understanding of NR uptake by different cells and tissues is greatly limited by the lack of noninvasive in vivo imaging tools limiting its clinical translation. Here, we report the development and validation of a bioluminescent NR uptake probe (BiNR) for non-invasive longitudinal imaging of NR uptake both in vitro and in vivo. In addition, we optimized an assay that allows monitoring of NR flux without the need to transfect cells with the luciferase gene, enabling the use of the BiNR probe in clinical samples, as demonstrated with human T cells. Lastly, we used BiNR to investigate the role of NR uptake in cancer prevalence and metastases formation in triple negative breast cancer (TNBC) animal model. Our results demonstrate that NR supplementation results in a significant increase in cancer prevalence and metastases of TNBC to the brain. These results outline the important role of powerful nutraceuticals like NR in cancer metabolism and the need to personalize their use in certain patient populations.


Subject(s)
Biosensing Techniques , Triple Negative Breast Neoplasms , Animals , Humans , NAD , Niacinamide/metabolism , Pyridinium Compounds
2.
Mol Metab ; 66: 101605, 2022 12.
Article in English | MEDLINE | ID: mdl-36165811

ABSTRACT

OBJECTIVE: Disturbances in NAD+ metabolism have been described as a hallmark for multiple metabolic and age-related diseases, including type 2 diabetes. While alterations in pancreatic ß-cell function are critical determinants of whole-body glucose homeostasis, the role of NAD+ metabolism in the endocrine pancreas remains poorly explored. Here, we aimed to evaluate the role of nicotinamide riboside (NR) metabolism in maintaining NAD+ levels and pancreatic ß-cell function in pathophysiological conditions. METHODS: Whole body and pancreatic ß-cell-specific NRK1 knockout (KO) mice were metabolically phenotyped in situations of high-fat feeding and aging. We also analyzed pancreatic ß-cell function, ß-cell mass and gene expression. RESULTS: We first demonstrate that NRK1, the essential enzyme for the utilization of NR, is abundantly expressed in pancreatic ß-cells. While NR treatment did not alter glucose-stimulated insulin secretion in pancreatic islets from young healthy mice, NRK1 knockout mice displayed glucose intolerance and compromised ß-cells response to a glucose challenge upon high-fat feeding or aging. Interestingly, ß cell dysfunction stemmed from the functional failure of other organs, such as liver and kidney, and the associated changes in circulating peptides and hormones, as mice lacking NRK1 exclusively in ß-cells did not show altered glucose homeostasis. CONCLUSIONS: This work unveils a new physiological role for NR metabolism in the maintenance of glucose tolerance and pancreatic ß-cell function in high-fat feeding or aging conditions.


Subject(s)
Diabetes Mellitus, Type 2 , NAD , Phosphotransferases (Alcohol Group Acceptor) , Animals , Mice , Diet, High-Fat/adverse effects , Glucose , Mice, Knockout , NAD/metabolism , Niacinamide/pharmacology , Niacinamide/metabolism , Pyridinium Compounds , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Insulin-Secreting Cells/pathology , Aging
3.
Nutrients ; 14(13)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35807932

ABSTRACT

Through evolution, eukaryote organisms have developed the ability to use different molecules as independent precursors to generate nicotinamide adenine dinucleotide (NAD+), an essential molecule for life. However, whether these different precursors act in an additive or complementary manner is not truly well understood. Here, we have evaluated how combinations of different NAD+ precursors influence intracellular NAD+ levels. We identified dihydronicotinic acid riboside (NARH) as a new NAD+ precursor in hepatic cells. Second, we demonstrate how NARH, but not any other NAD+ precursor, can act synergistically with nicotinamide riboside (NR) to increase NAD+ levels in cultured cells and in mice. Finally, we demonstrate that the large increase in NAD+ prompted by the combination of these two precursors is due to their chemical interaction and conversion to dihydronicotinamide riboside (NRH). Altogether, this work demonstrates for the first time that NARH can act as a NAD+ precursor in mammalian cells and how different NAD+ precursors can interact and influence each other when co-administered.


Subject(s)
NAD , Niacinamide , Animals , Mammals , Mice , Niacinamide/analogs & derivatives , Pyridinium Compounds
4.
Cell Rep ; 36(8): 109565, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433037

ABSTRACT

Mitochondria constantly undergo fusion and fission events, referred as mitochondrial dynamics, which determine mitochondrial architecture and bioenergetics. Cultured cell studies demonstrate that mitochondrial dynamics are acutely regulated by phosphorylation of the mitochondrial fission orchestrator dynamin-related protein 1 (Drp1) at S579 or S600. However, the physiological impact and crosstalk of these phosphorylation sites is poorly understood. Here, we describe the functional interrelation between S579 and S600 phosphorylation sites in vivo and their role on mitochondrial remodeling. Mice carrying a homozygous Drp1 S600A knockin (Drp1 KI) mutation display larger mitochondria and enhanced lipid oxidation and respiratory capacities, granting improved glucose tolerance and thermogenic response upon high-fat feeding. Housing mice at thermoneutrality blunts these differences, suggesting a role for the brown adipose tissue in the protection of Drp1 KI mice against metabolic damage. Overall, we demonstrate crosstalk between Drp1 phosphorylation sites and provide evidence that their modulation could be used in the treatment and prevention of metabolic diseases.


Subject(s)
Adipose Tissue, Brown/metabolism , Dynamins/metabolism , Lipid Metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Animals , Dynamins/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mutation , Oxidation-Reduction , Phosphorylation
6.
Mol Metab ; 30: 192-202, 2019 12.
Article in English | MEDLINE | ID: mdl-31767171

ABSTRACT

OBJECTIVE: A decay in intracellular NAD+ levels is one of the hallmarks of physiological decline in normal tissue functions. Accordingly, dietary supplementation with NAD+ precursors can prevent, alleviate, or even reverse multiple metabolic complications and age-related disorders in diverse model organisms. Within the constellation of NAD+ precursors, nicotinamide riboside (NR) has gained attention due to its potent NAD+ biosynthetic effects in vivo while lacking adverse clinical effects. Nevertheless, NR is not stable in circulation, and its utilization is rate-limited by the expression of nicotinamide riboside kinases (NRKs). Therefore, there is a strong interest in identifying new effective NAD+ precursors that can overcome these limitations. METHODS: Through a combination of metabolomics and pharmacological approaches, we describe how NRH, a reduced form of NR, serves as a potent NAD+ precursor in mammalian cells and mice. RESULTS: NRH acts as a more potent and faster NAD+ precursor than NR in mammalian cells and tissues. Despite the minor structural difference, we found that NRH uses different steps and enzymes to synthesize NAD+, thus revealing a new NRK1-independent pathway for NAD+ synthesis. Finally, we provide evidence that NRH is orally bioavailable in mice and prevents cisplatin-induced acute kidney injury. CONCLUSIONS: Our data identify a new pathway for NAD+ synthesis and classify NRH as a promising new therapeutic strategy to enhance NAD+ levels.


Subject(s)
NAD/biosynthesis , NAD/metabolism , Niacinamide/analogs & derivatives , Animals , Cell Line , Male , Mice , Niacinamide/metabolism , Niacinamide/physiology , Phosphotransferases (Alcohol Group Acceptor) , Pyridinium Compounds , Rats
7.
Nat Commun ; 10(1): 4291, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541116

ABSTRACT

Supplementation with the NAD+ precursor nicotinamide riboside (NR) ameliorates and prevents a broad array of metabolic and aging disorders in mice. However, little is known about the physiological role of endogenous NR metabolism. We have previously shown that NR kinase 1 (NRK1) is rate-limiting and essential for NR-induced NAD+ synthesis in hepatic cells. To understand the relevance of hepatic NR metabolism, we generated whole body and liver-specific NRK1 knockout mice. Here, we show that NRK1 deficiency leads to decreased gluconeogenic potential and impaired mitochondrial function. Upon high-fat feeding, NRK1 deficient mice develop glucose intolerance, insulin resistance and hepatosteatosis. Furthermore, they are more susceptible to diet-induced liver DNA damage, due to compromised PARP1 activity. Our results demonstrate that endogenous NR metabolism is critical to sustain hepatic NAD+ levels and hinder diet-induced metabolic damage, highlighting the relevance of NRK1 as a therapeutic target for metabolic disorders.


Subject(s)
Diet, High-Fat/adverse effects , Liver Diseases/prevention & control , Niacinamide/analogs & derivatives , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protective Agents/metabolism , Protective Agents/pharmacology , Animals , Blood Glucose , DNA Damage , Disease Models, Animal , Gene Knockout Techniques , Genetic Predisposition to Disease/genetics , Glucose Intolerance , Hepatocytes/metabolism , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/pathology , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD/metabolism , Niacinamide/genetics , Niacinamide/metabolism , Niacinamide/pharmacology , Pyridinium Compounds
8.
EMBO J ; 36(11): 1543-1558, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28348166

ABSTRACT

Mitochondrial fusion and fission events, collectively known as mitochondrial dynamics, act as quality control mechanisms to ensure mitochondrial function and fine-tune cellular bioenergetics. Defective mitofusin 2 (Mfn2) expression and enhanced mitochondrial fission in skeletal muscle are hallmarks of insulin-resistant states. Interestingly, Mfn2 is highly expressed in brown adipose tissue (BAT), yet its role remains unexplored. Using adipose-specific Mfn2 knockout (Mfn2-adKO) mice, we demonstrate that Mfn2, but not Mfn1, deficiency in BAT leads to a profound BAT dysfunction, associated with impaired respiratory capacity and a blunted response to adrenergic stimuli. Importantly, Mfn2 directly interacts with perilipin 1, facilitating the interaction between the mitochondria and the lipid droplet in response to adrenergic stimulation. Surprisingly, Mfn2-adKO mice were protected from high-fat diet-induced insulin resistance and hepatic steatosis. Altogether, these results demonstrate that Mfn2 is a mediator of mitochondria to lipid droplet interactions, influencing lipolytic processes and whole-body energy homeostasis.


Subject(s)
Adipose Tissue, Brown/metabolism , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Thermogenesis , Animals , GTP Phosphohydrolases/deficiency , Mice , Mice, Knockout , Perilipin-1/metabolism , Protein Binding
9.
Nat Commun ; 7: 13103, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27725675

ABSTRACT

NAD+ is a vital redox cofactor and a substrate required for activity of various enzyme families, including sirtuins and poly(ADP-ribose) polymerases. Supplementation with NAD+ precursors, such as nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), protects against metabolic disease, neurodegenerative disorders and age-related physiological decline in mammals. Here we show that nicotinamide riboside kinase 1 (NRK1) is necessary and rate-limiting for the use of exogenous NR and NMN for NAD+ synthesis. Using genetic gain- and loss-of-function models, we further demonstrate that the role of NRK1 in driving NAD+ synthesis from other NAD+ precursors, such as nicotinamide or nicotinic acid, is dispensable. Using stable isotope-labelled compounds, we confirm NMN is metabolized extracellularly to NR that is then taken up by the cell and converted into NAD+. Our results indicate that mammalian cells require conversion of extracellular NMN to NR for cellular uptake and NAD+ synthesis, explaining the overlapping metabolic effects observed with the two compounds.


Subject(s)
Mammals/metabolism , Niacinamide/analogs & derivatives , Nicotinamide Mononucleotide/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Hep G2 Cells , Hepatocytes/metabolism , Humans , Injections, Intraperitoneal , Mice, Knockout , NAD/biosynthesis , Niacinamide/metabolism , Pyridinium Compounds
10.
Diabetes ; 65(12): 3552-3560, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27613809

ABSTRACT

Mitochondrial function can be influenced by mitochondrial shape and connectivity with other cellular organelles through fusion and fission processes. Disturbances in mitochondrial architecture and mitochondrial fusion-related genes are observed in situations of type 2 diabetes and obesity, leading to a highly fissioned mitochondrial network. To directly test the effect of reduced mitochondrial fusion on hepatic metabolism, we generated mice with a liver-specific deletion of the Mfn1 gene (Mfn1LKO) and monitored their energy homeostasis, mitochondrial function, and susceptibility to diet-induced insulin resistance. Livers from Mfn1LKO mice displayed a highly fragmented mitochondrial network. This was coupled to an enhanced mitochondrial respiration capacity and a preference for the use of lipids as the main energy source. Although Mfn1LKO mice are similar to control mice fed a low-fat diet, they are protected against insulin resistance induced by a high-fat diet. Importantly, Mfn1 deficiency increased complex I abundance and sensitized animals to the hypoglycemic effect of metformin. Our results suggest that targeting Mfn1 could provide novel avenues to ameliorate glucose homeostasis in obese patients and improve the effectiveness of metformin.


Subject(s)
GTP Phosphohydrolases/deficiency , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Animals , Diet, High-Fat/adverse effects , GTP Phosphohydrolases/genetics , Homeostasis/drug effects , Insulin Resistance/physiology , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism
11.
Cell Rep ; 14(9): 2068-2075, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26923584

ABSTRACT

Caloric restriction (CR) has been shown to prevent the onset of insulin resistance and to delay age-related physiological decline in mammalian organisms. SIRT1, a NAD(+)-dependent deacetylase enzyme, has been suggested to mediate the adaptive responses to CR, leading to the speculation that SIRT1 activation could be therapeutically used as a CR-mimetic strategy. Here, we used a mouse model of moderate SIRT1 overexpression to test whether SIRT1 gain of function could mimic or boost the metabolic benefits induced by every-other-day feeding (EODF). Our results indicate that SIRT1 transgenesis does not affect the ability of EODF to decrease adiposity and improve insulin sensitivity. Transcriptomic analyses revealed that SIRT1 transgenesis and EODF promote very distinct adaptations in individual tissues, some of which can be even be metabolically opposite, as in brown adipose tissue. Therefore, whereas SIRT1 overexpression and CR both improve glucose metabolism and insulin sensitivity, the etiologies of these benefits are largely different.


Subject(s)
Fasting/physiology , Sirtuin 1/physiology , Adaptation, Physiological , Animals , Gene Expression , Glucose/metabolism , Insulin Resistance , Liver/metabolism , Male , Mice, Transgenic , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Transcriptome
12.
Mol Metab ; 4(2): 118-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25685699

ABSTRACT

OBJECTIVE: SIRT1 has been proposed to be a key signaling node linking changes in energy metabolism to transcriptional adaptations. Although SIRT1 overexpression is protective against diverse metabolic complications, especially in response to high-fat diets, studies aiming to understand the etiology of such benefits are scarce. Here, we aimed to identify the key tissues and mechanisms implicated in the beneficial effects of SIRT1 on glucose homeostasis. METHODS: We have used a mouse model of moderate SIRT1 overexpression, under the control of its natural promoter, to evaluate glucose homeostasis and thoroughly characterize how different tissues could influence insulin sensitivity. RESULTS: Mice with moderate overexpression of SIRT1 exhibit better glucose tolerance and insulin sensitivity even on a low fat diet. Euglycemic-hyperinsulinemic clamps and in-depth tissue analyses revealed that enhanced insulin sensitivity was achieved through a higher brown adipose tissue activity and was fully reversed by housing the mice at thermoneutrality. SIRT1 did not influence brown adipocyte differentiation, but dramatically enhanced the metabolic transcriptional responses to ß3-adrenergic stimuli in differentiated adipocytes. CONCLUSIONS: Our work demonstrates that SIRT1 improves glucose homeostasis by enhancing BAT function. This is not consequent to an alteration in the brown adipocyte differentiation process, but as a result of potentiating the response to ß3-adrenergic stimuli.

13.
J Transl Med ; 11: 5, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23294527

ABSTRACT

BACKGROUND: EMD 521873 (Selectikine or NHS-IL2LT) is a fusion protein consisting of modified human IL-2 which binds specifically to the high-affinity IL-2 receptor, and an antibody specific for both single- and double-stranded DNA, designed to facilitate the enrichment of IL-2 in tumor tissue. METHODS: An extensive analysis of pharmacodynamic (PD) markers associated with target modulation was assessed during a first-in-human phase I dose-escalation trial of Selectikine. RESULTS: Thirty-nine patients with metastatic or locally advanced tumors refractory to standard treatments were treated with increasing doses of Selectikine, and nine further patients received additional cyclophosphamide. PD analysis, assessed during the first two treatment cycles, revealed strong activation of both CD4+ and CD8+ T-cells and only weak NK cell activation. No dose response was observed. As expected, Treg cells responded actively to Selectikine but remained at lower frequency than effector CD4+ T-cells. Interestingly, patient survival correlated positively with both high lymphocyte counts and low levels of activated CD8+ T-cells at baseline, the latter of which was associated with enhanced T-cell responses to the treatment. CONCLUSIONS: The results confirm the selectivity of Selectikine with predominant T-cell and low NK cell activation, supporting follow-up studies assessing the clinical efficacy of Selectikine for cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA/immunology , Interleukin-2/immunology , Lymphocyte Activation , Neoplasms/drug therapy , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/cytology , Cell Proliferation , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Count , Survival Analysis
14.
J Biol Chem ; 285(40): 31011-23, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20610391

ABSTRACT

Accumulation of fat in the liver increases the risk to develop fibrosis and cirrhosis and is associated with development of the metabolic syndrome. Here, to identify genes or gene pathways that may underlie the genetic susceptibility to fat accumulation in liver, we studied A/J and C57Bl/6 mice that are resistant and sensitive to diet-induced hepatosteatosis and obesity, respectively. We performed comparative transcriptomic and lipidomic analysis of the livers of both strains of mice fed a high fat diet for 2, 10, and 30 days. We found that resistance to steatosis in A/J mice was associated with the following: (i) a coordinated up-regulation of 10 genes controlling peroxisome biogenesis and ß-oxidation; (ii) an increased expression of the elongase Elovl5 and desaturases Fads1 and Fads2. In agreement with these observations, peroxisomal ß-oxidation was increased in livers of A/J mice, and lipidomic analysis showed increased concentrations of long chain fatty acid-containing triglycerides, arachidonic acid-containing lysophosphatidylcholine, and 2-arachidonylglycerol, a cannabinoid receptor agonist. We found that the anti-inflammatory CB2 receptor was the main hepatic cannabinoid receptor, which was highly expressed in Kupffer cells. We further found that A/J mice had a lower pro-inflammatory state as determined by lower plasma levels and IL-1ß and granulocyte-CSF and reduced hepatic expression of their mRNAs, which were found only in Kupffer cells. This suggests that increased 2-arachidonylglycerol production may limit Kupffer cell activity. Collectively, our data suggest that genetic variations in the expression of peroxisomal ß-oxidation genes and of genes controlling the production of an anti-inflammatory lipid may underlie the differential susceptibility to diet-induced hepatic steatosis and pro-inflammatory state.


Subject(s)
Dietary Fats/adverse effects , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Microsomes, Liver/metabolism , Peroxisomes/metabolism , Animals , Dietary Fats/pharmacology , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression Regulation/genetics , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Kupffer Cells/metabolism , Kupffer Cells/pathology , Lipid Metabolism/genetics , Lipids/biosynthesis , Lipids/genetics , Male , Mice , Microsomes, Liver/pathology , Peroxisomes/genetics , Peroxisomes/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/genetics , Species Specificity , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
15.
J Biol Chem ; 285(14): 10538-45, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20145256

ABSTRACT

Glucagon-like peptide-1 (GLP-1) protects beta-cells against apoptosis, increases their glucose competence, and induces their proliferation. We previously demonstrated that the anti-apoptotic effect was mediated by an increase in insulin-like growth factor-1 receptor (IGF-1R) expression and signaling, which was dependent on autocrine secretion of insulin-like growth factor 2 (IGF-2). Here, we further investigated how GLP-1 induces IGF-1R expression and whether the IGF-2/IGF-1R autocrine loop is also involved in mediating GLP-1-increase in glucose competence and proliferation. We show that GLP-1 up-regulated IGF-1R expression by a protein kinase A-dependent translational control mechanism, whereas isobutylmethylxanthine, which led to higher intracellular accumulation of cAMP than GLP-1, increased both IGF-1R transcription and translation. We then demonstrated, using MIN6 cells and primary islets, that the glucose competence of these cells was dependent on the level of IGF-1R expression and on IGF-2 secretion. We showed that GLP-1-induced primary beta-cell proliferation was suppressed by Igf-1r gene inactivation and by IGF-2 immunoneutralization or knockdown. Together our data show that regulation of beta-cell number and function by GLP-1 depends on the cAMP/protein kinase A mediated-induction of IGF-1R expression and the increased activity of an IGF-2/IGF-1R autocrine loop.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Protein Biosynthesis , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fluorescent Antibody Technique , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription, Genetic
16.
J Biol Chem ; 283(42): 28536-45, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18713749

ABSTRACT

P-selectin glycoprotein ligand-1 (PSGL-1) interacts with selectins to support leukocyte rolling along vascular wall. L- and P-selectin bind to N-terminal tyrosine sulfate residues and to core-2 O-glycans attached to Thr-57, whereas tyrosine sulfation is not required for E-selectin binding. PSGL-1 extracellular domain contains decameric repeats, which extend L- and P-selectin binding sites far above the plasma membrane. We hypothesized that decamers may play a role in regulating PSGL-1 interactions with selectins. Chinese hamster ovary cells expressing wild-type PSGL-1 or PSGL-1 molecules exhibiting deletion or substitution of decamers with the tandem repeats of platelet glycoprotein Ibalpha were compared in their ability to roll on selectins and to bind soluble L- or P-selectin. Deletion of decamers abrogated soluble L-selectin binding and cell rolling on L-selectin, whereas their substitution partially reversed these diminutions. P-selectin-dependent interactions with PSGL-1 were less affected by decamer deletion. Videomicroscopy analysis showed that decamers are required to stabilize L-selectin-dependent rolling. Importantly, adhesion assays performed on recombinant decamers demonstrated that they directly bind to E-selectin and promote slow rolling. Our results indicate that the role of decamers is to extend PSGL-1 N terminus far above the cell surface to support and stabilize leukocyte rolling on L- or P-selectin. In addition, they function as a cell adhesion receptor, which supports approximately 80% of E-selectin-dependent rolling.


Subject(s)
Membrane Glycoproteins/physiology , Selectins/chemistry , Animals , CHO Cells , Cell Adhesion , Cell Membrane/metabolism , Cricetinae , Cricetulus , Humans , K562 Cells , Leukocyte Rolling , Membrane Glycoproteins/metabolism , Microscopy, Video , Models, Biological , Protein Structure, Tertiary , Tyrosine/analogs & derivatives , Tyrosine/chemistry
17.
J Biol Chem ; 280(7): 5378-90, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15579466

ABSTRACT

P-selectin glycoprotein ligand-1 (PSGL-1) interactions with selectins regulate leukocyte migration in inflammatory lesions. In mice, selectin ligand activity regulating leukocyte recruitment and lymphocyte homing into lymph nodes results from the sum of unequal contributions of fucosyltransferase (FucT)-IV and FucT-VII, with FucT-VII playing a predominant role. Here we have examined the role of human FucT-IV and -VII in conferring L-selectin, P-selectin, and E-selectin binding activities to PSGL-1. Lewis x (Le(x)) carbohydrate was generated at the CHO(dhfr)(-) cell surface by FucT-IV expression, whereas sialyl Le(x) (sLe(x)) was synthesized by FucT-VII. Both human FucT-IV and -VII had the ability to generate carbohydrate ligands that support L-selectin-, P-selectin-, and E-selectin-dependent rolling on PSGL-1, with FucT-VII playing a major role. Cooperation was observed between FucT-IV and -VII in recruiting L-, P-, or E-selectin-expressing cells on PSGL-1 and in regulating cell rolling velocity and stability. Additional rolling adhesion assays were performed to assess the role of Thr-57-linked core-2 O-glycans in supporting L-selectin-, P-selectin-, and E-selectin-dependent rolling on PSGL-1. These studies confirmed that core-2 O-glycans attached to Thr-57 play a critical role in supporting L- and P-selectin-dependent rolling and revealed that additional binding sites support >75% of E-selectin-mediated rolling. The observations presented here indicate that human FucT-IV and -VII both contribute and cooperate in regulating L-selectin-, P-selectin-, and E-selectin-dependent rolling on PSGL-1, with FucT-VII playing a predominant role in conferring selectin binding activity to PSGL-1.


Subject(s)
Fucosyltransferases/metabolism , Membrane Glycoproteins/metabolism , Selectins/metabolism , Animals , CHO Cells , Cell Adhesion , Cells, Cultured , Cricetinae , Epitopes/chemistry , Epitopes/metabolism , Flow Cytometry , Fucosyltransferases/genetics , Glycosylation , Humans , Leukocyte Rolling , Lewis X Antigen/chemistry , Lewis X Antigen/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Neutrophils/cytology , Neutrophils/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Protein Binding/drug effects , Selectins/genetics , Sialyl Lewis X Antigen , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...