Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 21(9): 2528-2544, 2023 09.
Article in English | MEDLINE | ID: mdl-37085035

ABSTRACT

BACKGROUND: Germline mutations in the ETV6 transcription factor gene are responsible for familial thrombocytopenia and leukemia predisposition syndrome. Although previous studies have shown that ETV6 plays an important role in megakaryocyte (MK) maturation and platelet formation, the mechanisms by which ETV6 dysfunction promotes thrombocytopenia remain unclear. OBJECTIVES: To decipher the transcriptional mechanisms and gene regulatory network linking ETV6 germline mutations and thrombocytopenia. METHODS: Presuming that ETV6 mutations result in selective effects at a particular cell stage, we applied single-cell RNA sequencing to understand gene expression changes during megakaryopoiesis in peripheral CD34+ cells from healthy controls and patients with ETV6-related thrombocytopenia. RESULTS: Analysis of gene expression and regulon activity revealed distinct clusters partitioned into 7 major cell stages: hematopoietic stem/progenitor cells, common-myeloid progenitors (CMPs), MK-primed CMPs, granulocyte-monocyte progenitors, MK-erythroid progenitors (MEPs), progenitor MKs/mature MKs, and platelet-like particles. We observed a differentiation trajectory in which MEPs developed directly from hematopoietic stem/progenitor cells and bypassed the CMP stage. ETV6 deficiency led to the development of aberrant cells as early as the MEP stage, which intensified at the progenitor MK/mature MK stage, with a highly deregulated core "ribosome biogenesis" pathway. Indeed, increased translation levels have been documented in patient CD34+-derived MKs with overexpression of ribosomal protein S6 and phosphorylated ribosomal protein S6 in both CD34+-derived MKs and platelets. Treatment of patient MKs with the ribosomal biogenesis inhibitor CX-5461 resulted in an increase in platelet-like particles. CONCLUSION: These findings provide novel insight into both megakaryopoiesis and the link among ETV6, translation, and platelet production.


Subject(s)
Megakaryocytes , Thrombocytopenia , Humans , Cell Differentiation , Megakaryocytes/metabolism , Ribosomal Protein S6/metabolism , Single-Cell Analysis , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombopoiesis/genetics , Antigens, CD34 , ETS Translocation Variant 6 Protein
2.
J Thromb Haemost ; 19(9): 2287-2301, 2021 09.
Article in English | MEDLINE | ID: mdl-34060193

ABSTRACT

BACKGROUND: GATA1 is an essential transcription factor for both polyploidization and megakaryocyte (MK) differentiation. The polyploidization defect observed in GATA1 variant carriers is not well understood. OBJECTIVE: To extensively phenotype two pedigrees displaying different variants in the GATA1 gene and determine if GATA1 controls MYH10 expression levels, a key modulator of MK polyploidization. METHOD: A total of 146 unrelated propositi with constitutional thrombocytopenia were screened on a multigene panel. We described the genotype-phenotype correlation in GATA1 variant carriers and investigated the effect of these novel variants on MYH10 transcription using luciferase constructs. RESULTS: The clinical profile associated with the p.L268M variant localized in the C terminal zinc finger was unusual in that the patient displayed bleeding and severe platelet aggregation defects without early-onset thrombocytopenia. p.N206I localized in the N terminal zinc finger was associated, on the other hand, with severe thrombocytopenia (15G/L) in early life. High MYH10 levels were evidenced in platelets of GATA1 variant carriers. Analysis of MKs anti-GATA1 chromatin immunoprecipitation-sequencing data revealed two GATA1 binding sites, located in the 3' untranslated region and in intron 8 of the MYH10 gene. Luciferase reporter assays showed their respective role in the regulation of MYH10 gene expression. Both GATA1 variants significantly alter intron 8 driven MYH10 transcription. CONCLUSION: The discovery of an association between MYH10 and GATA1 is a novel one. Overall, this study suggests that impaired MYH10 silencing via an intronic regulatory element is the most likely cause of GATA1-related polyploidization defect.


Subject(s)
GATA1 Transcription Factor , Megakaryocytes , Myosin Heavy Chains/genetics , Nonmuscle Myosin Type IIB/genetics , Thrombocytopenia , Blood Platelets , GATA1 Transcription Factor/genetics , Gene Silencing , Humans , Thrombocytopenia/genetics , Thrombopoiesis/genetics , Transcription Factors
3.
J Thromb Haemost ; 19(5): 1186-1199, 2021 05.
Article in English | MEDLINE | ID: mdl-33605529

ABSTRACT

BACKGROUND: Coagulation factor V (FV), present in plasma and platelets, has both pro- and anticoagulant functions. OBJECTIVE: We investigated an FV-deficient patient (FV:C 3%, FV:Ag 4%) paradoxically presenting with recurrent venous thrombosis (11 events) instead of bleeding. METHODS/RESULTS: Thrombophilia screening revealed only heterozygosity for the F2 20210G>A mutation. Although thrombin generation in the patient's platelet-poor plasma was suggestive of a hypocoagulable state, thrombin generation in the patient's platelet-rich plasma (PRP) was higher than in control PRP and extremely resistant to activated protein C (APC). This was partially attributable to the complete abolition of the APC-cofactor activity of FV and a marked reduction of plasma tissue factor pathway inhibitor antigen and activity. The patient was homozygous for a novel missense mutation (Ala2086Asp, FVBesançon ) that favors a "closed conformation" of the C2 domain, predicting impaired binding of FV(a) to phospholipids. Recombinant FVBesançon was hardly secreted, indicating that this mutation is responsible for the patient's FV deficiency. Model system experiments performed using highly diluted plasma as a source of FV showed that, compared with normal FVa, FVaBesançon has slightly (≤1.5-fold) unfavorable kinetic parameters (Km , Vmax ) of prothrombin activation, but also a lower rate of APC-catalyzed inactivation in the presence of protein S. CONCLUSIONS: FVBesançon induces a hypercoagulable state via quantitative (markedly decreased FV level) and qualitative (phospholipid-binding defect) effects that affect anticoagulant pathways (anticoagulant activities of FV, FVa inactivation, tissue factor pathway inhibitor α level) more strongly than the prothrombinase activity of FVa. A possible specific role of platelet FV cannot be excluded.


Subject(s)
Factor V , Thrombophilia , Blood Coagulation Tests , Factor V/genetics , Homozygote , Humans , Mutation , Thrombophilia/genetics
4.
J Thromb Haemost ; 18(3): 693-705, 2020 03.
Article in English | MEDLINE | ID: mdl-31758832

ABSTRACT

BACKGROUND: The small GTPase Rap1 and its guanine nucleotide exchange factor, CalDAG-GEFI (CDGI), are critical for platelet function and hemostatic plug formation. CDGI function is regulated by a calcium binding EF hand regulatory domain and an atypical C1 domain with unknown function. OBJECTIVE: Here, we investigated whether the C1 domain controls CDGI subcellular localization, both in vitro and in vivo. METHODS: CDGI interaction with phosphoinositides was studied by lipid co-sedimentation assays and molecular dynamics simulations. Cellular localization of CDGI was studied in heterologous cells by immunofluorescence and subcellular fractionation assays. RESULTS: Lipid co-sedimentation studies demonstrated that the CDGI C1 domain associates with membranes through exclusive recognition of phosphoinositides, phosphatidylinositol (4,5)-biphosphate (PIP2) and phosphatidylinositol (3,4,5)-triphosphate (PIP3). Molecular dynamics simulations identified a phospholipid recognition motif consisting of residues exclusive to the CDGI C1 domain. Mutation of those residues abolished co-sedimentation of the C1 domain with lipid vesicles and impaired membrane localization of CDGI in heterologous cells. CONCLUSION: Our studies identify a novel interaction between an atypical C1 domain and phosphatidylinositol (4,5)-biphosphate and phosphatidylinositol (3,4,5)-triphosphate in cellular membranes, which is critical for Rap1 signaling in health and disease.


Subject(s)
GTP Phosphohydrolase Activators , Phosphatidylinositols , GTP Phosphohydrolases , Guanine Nucleotide Exchange Factors , Signal Transduction
5.
Res Pract Thromb Haemost ; 3(4): 684-694, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31624788

ABSTRACT

Antiplatelet therapy through inhibition of the adenosine diphosphate (ADP)/P2Y12 pathway is commonly used in the treatment of acute coronary syndrome (ACS). Although efficient in preventing platelet activation and thrombus formation, it increases the risk of bleeding complications. In patients with ACS receiving platelet aggregation inhibitors, that is, P2Y12 blockers (n = 923), we investigated the relationship between plasma and platelet-associated CD40L levels and bleeding events (n = 71). Treatment with P2Y12 inhibitors in patients with ACS did not affect plasma-soluble CD40L levels, but decreased platelet CD40L surface expression (pCD40L) and platelet-released CD40L (rCD40L) levels in response to stimulation as compared to healthy controls. In vitro inhibition of the ADP pathway in healthy control platelets reduced both pCD40L and rCD40L levels. In a multivariable analysis, the reduced pCD40L level observed in ACS patients was significantly associated with the risk of bleeding occurrence (adjusted odds ratio = 0.15; 95% confidence interval = 0.034-0.67). P2Y12 inhibitor-treated (ticagrelor) mice exhibited a 2.5-fold increase in tail bleeding duration compared with controls. A significant reduction in bleeding duration was observed on CD40L+/+ but not CD40L-/- platelet infusion. In addition, CD40L blockade in P2Y12 inhibitor-treated blood samples from a healthy human reduced thrombus growth over immobilized collagen under arterial flow. In conclusion, measurement of pCD40L may offer a novel approach to assessing bleeding risk in patients with ACS who are being treated with P2Y12 inhibitors.

6.
Sci Rep ; 9(1): 9631, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31270351

ABSTRACT

Increased platelet activity occurs in type 2 diabetes mellitus (T2DM) and such platelet dysregulation likely originates from altered megakaryopoiesis. We initiated identification of dysregulated pathways in megakaryocytes in the setting of T2DM. We evaluated through transcriptomic analysis, differential gene expressions in megakaryocytes from leptin receptor-deficient mice (db/db), exhibiting features of human T2DM, and control mice (db/+). Functional gene analysis revealed an upregulation of transcripts related to calcium signaling, coagulation cascade and platelet receptors in diabetic mouse megakaryocytes. We also evidenced an upregulation (7- to 9.7-fold) of genes encoding stefin A (StfA), the human ortholog of Cystatin A (CSTA), inhibitor of cathepsin B, H and L. StfA/CSTA was present in megakaryocytes and platelets and its expression increased during obesity and diabetes in rats and humans. StfA/CSTA was primarily localized at platelet membranes and granules and was released upon agonist stimulation and clot formation through a metalloprotease-dependent mechanism. StfA/CSTA did not affect platelet aggregation, but reduced platelet accumulation on immobilized collagen from flowing whole blood (1200 s-1). In-vivo, upon laser-induced vascular injury, platelet recruitment and thrombus formation were markedly reduced in StfA1-overexpressing mice without affecting bleeding time. The presence of CA-074Me, a cathepsin B specific inhibitor significantly reduced thrombus formation in-vitro and in-vivo in human and mouse, respectively. Our study identifies StfA/CSTA as a key contributor of platelet-dependent thrombus formation in both rodents and humans.


Subject(s)
Blood Platelets/enzymology , Cystatin A/metabolism , Diabetes Mellitus, Experimental/complications , Megakaryocytes/enzymology , Thrombosis/prevention & control , Animals , Calcium Signaling , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Aggregation , Rats , Rats, Wistar , Thrombosis/etiology , Thrombosis/metabolism , Thrombosis/pathology
7.
Hum Mutat ; 40(12): 2258-2269, 2019 12.
Article in English | MEDLINE | ID: mdl-31237726

ABSTRACT

The ACTN1 gene has been implicated in inherited macrothrombocytopenia. To decipher the spectrum of variants and phenotype of ACTN1-related thrombocytopenia, we sequenced the ACTN1 gene in 272 cases of unexplained chronic or familial thrombocytopenia. We identified 15 rare, monoallelic, nonsynonymous and likely pathogenic ACTN1 variants in 20 index cases from 20 unrelated families. Thirty-one family members exhibited thrombocytopenia. Targeted sequencing was carried out on 12 affected relatives, which confirmed presence of the variant. Twenty-eight of 32 cases with monoallelic ACTN1 variants had mild to no bleeding complications. Eleven cases harbored 11 different unreported ACTN1 variants that were monoallelic and likely pathogenic. Nine variants were located in the α-actinin-1 (ACTN1) rod domain and were predicted to hinder dimer formation. These variants displayed a smaller increase in platelet size compared with variants located outside the rod domain. In vitro expression of the new ACTN1 variants induced actin network disorganization and led to increased thickness of actin fibers. These findings expand the repertoire of ACTN1 variants associated with thrombocytopenia and highlight the high frequency of ACTN1-related thrombocytopenia cases. The rod domain, like other ACTN1 functional domains, may be mutated resulting in actin disorganization in vitro and thrombocytopenia with normal platelet size in most cases.


Subject(s)
Actinin/chemistry , Actinin/genetics , Mutation , Sequence Analysis, DNA/methods , Thrombocytopenia/genetics , Adolescent , Adult , Aged , Child , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutagenesis, Site-Directed , Pedigree , Protein Domains , Young Adult
9.
Haematologica ; 102(6): 1006-1016, 2017 06.
Article in English | MEDLINE | ID: mdl-28255014

ABSTRACT

Congenital macrothrombocytopenia is a family of rare diseases, of which a significant fraction remains to be genetically characterized. To analyze cases of unexplained thrombocytopenia, 27 individuals from a patient cohort of the Bleeding and Thrombosis Exploration Center of the University Hospital of Marseille were recruited for a high-throughput gene sequencing study. This strategy led to the identification of two novel FLI1 variants (c.1010G>A and c.1033A>G) responsible for macrothrombocytopenia. The FLI1 variant carriers' platelets exhibited a defect in aggregation induced by low-dose adenosine diphosphate (ADP), collagen and thrombin receptor-activating peptide (TRAP), a defect in adenosine triphosphate (ATP) secretion, a reduced mepacrine uptake and release and a reduced CD63 expression upon TRAP stimulation. Precise ultrastructural analysis of platelet content was performed using transmission electron microscopy and focused ion beam scanning electron microscopy. Remarkably, dense granules were nearly absent in the carriers' platelets, presumably due to a biogenesis defect. Additionally, 25-29% of the platelets displayed giant α-granules, while a smaller proportion displayed vacuoles (7-9%) and autophagosome-like structures (0-3%). In vitro study of megakaryocytes derived from circulating CD34+ cells of the carriers revealed a maturation defect and reduced proplatelet formation potential. The study of the FLI1 variants revealed a significant reduction in protein nuclear accumulation and transcriptional activity properties. Intraplatelet flow cytometry efficiently detected the biomarker MYH10 in FLI1 variant carriers. Overall, this study provides new insights into the phenotype, pathophysiology and diagnosis of FLI1 variant-associated thrombocytopenia.


Subject(s)
Cytoplasmic Granules/metabolism , Thrombocytopenia/etiology , Adult , Blood Platelets/pathology , Blood Platelets/ultrastructure , Cell Nucleus/chemistry , Genetic Variation , Humans , Male , Megakaryocytes/pathology , Middle Aged , Platelet Aggregation/genetics , Proto-Oncogene Protein c-fli-1/genetics , Thrombocytopenia/congenital , Transcription, Genetic , Young Adult
10.
Haematologica ; 102(2): 282-294, 2017 02.
Article in English | MEDLINE | ID: mdl-27663637

ABSTRACT

Variants in ETV6, which encodes a transcription repressor of the E26 transformation-specific family, have recently been reported to be responsible for inherited thrombocytopenia and hematologic malignancy. We sequenced the DNA from cases with unexplained dominant thrombocytopenia and identified six likely pathogenic variants in ETV6, of which five are novel. We observed low repressive activity of all tested ETV6 variants, and variants located in the E26 transformation-specific binding domain (encoding p.A377T, p.Y401N) led to reduced binding to corepressors. We also observed a large expansion of megakaryocyte colony-forming units derived from variant carriers and reduced proplatelet formation with abnormal cytoskeletal organization. The defect in proplatelet formation was also observed in control CD34+ cell-derived megakaryocytes transduced with lentiviral particles encoding mutant ETV6. Reduced expression levels of key regulators of the actin cytoskeleton CDC42 and RHOA were measured. Moreover, changes in the actin structures are typically accompanied by a rounder platelet shape with a highly heterogeneous size, decreased platelet arachidonic response, and spreading and retarded clot retraction in ETV6 deficient platelets. Elevated numbers of circulating CD34+ cells were found in p.P214L and p.Y401N carriers, and two patients from different families suffered from refractory anemia with excess blasts, while one patient from a third family was successfully treated for acute myeloid leukemia. Overall, our study provides novel insights into the role of ETV6 as a driver of cytoskeletal regulatory gene expression during platelet production, and the impact of variants resulting in platelets with altered size, shape and function and potentially also in changes in circulating progenitor levels.


Subject(s)
Blood Platelets/metabolism , Germ-Line Mutation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombopoiesis/genetics , Antigens, CD34/metabolism , Blood Cell Count , Cell Differentiation , Family , Female , Gene Expression Regulation , Genotype , Humans , Hyperplasia , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Megakaryocytes/pathology , Pedigree , Phenotype , Platelet Count , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , ETS Translocation Variant 6 Protein
11.
Thromb Haemost ; 117(2): 325-338, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27853810

ABSTRACT

Cell accumulation is a prerequisite for adipose tissue inflammation. The leukocyte integrin Mac-1 (CD11b/CD18, αMß2) is a classic adhesion receptor critically regulating inflammatory cell recruitment. Here, we tested the hypothesis that a genetic deficiency and a therapeutic modulation of Mac-1 regulate adipose tissue inflammation in a mouse model of diet-induced obesity (DIO). C57Bl6/J mice genetically deficient (Mac-1-/-) or competent for Mac-1 (WT) consumed a high fat diet for 20 weeks. Surprisingly, Mac-1-/- mice presented with increased diet-induced weight gain, decreased insulin sensitivity in skeletal muscle and in the liver in insulin-clamps, insulin secretion deficiency and elevated glucose levels in fasting animals, and dyslipidaemia. Unexpectedly, accumulation of adipose tissue macrophages (ATMs) was unaffected, while gene expression indicated less inflamed adipose tissue and macrophages in Mac-1-/- mice. In contrast, inflammatory gene expression at distant locations, such as in skeletal muscle, was not changed. Treatment of ATMs with an agonistic anti-Mac-1 antibody, M1/70, induced pro-inflammatory genes in cell culture. In vivo, treatment with M1/70 induced a hyper-inflammatory phenotype with increased expression of IL-6 and MCP-1, whereas accumulation of ATMs did not change. Finally, inhibition of Mac-1's adhesive interaction to CD40L by the peptide inhibitor cM7 did not affect myeloid cell accumulation in adipose tissue. We present the surprising finding that adhesive properties of the leukocyte integrin Mac-1 are not required for macrophage accumulation in adipose tissue. Instead, Mac-1 modulates inflammatory gene expression in macrophages. These findings question the net effect of integrin blockade in cardio-metabolic disease.


Subject(s)
CD11b Antigen/metabolism , CD18 Antigens/metabolism , Chemotaxis , Diet/adverse effects , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Leukocytes/metabolism , Macrophage-1 Antigen/metabolism , Macrophages/metabolism , Obesity/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/pharmacology , CD11b Antigen/deficiency , CD11b Antigen/genetics , CD18 Antigens/deficiency , CD18 Antigens/genetics , Cell Adhesion , Cells, Cultured , Chemotaxis/drug effects , Cytokines/metabolism , Disease Models, Animal , Genotype , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Inflammation/genetics , Inflammation/pathology , Insulin Resistance , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Leukocytes/drug effects , Leukocytes/pathology , Macrophage-1 Antigen/genetics , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/pathology , Phenotype , Signal Transduction/drug effects , Weight Gain
12.
Antioxid Redox Signal ; 26(10): 489-500, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27225042

ABSTRACT

SIGNIFICANCE: Immune T cells are present in adipose tissues (AT), and the stoichiometry of the different T cell subsets is altered during diet-induced obesity (DIO). T cells contribute to the early steps of AT inflammation during DIO. Recent Advances: Many factors could potentially be responsible for this altered pro-inflammatory versus anti-inflammatory T cell balance. CRITICAL ISSUES: T cells are potentially activated in AT, which vitamin D might contribute to, as will be discussed in this article. In addition, we will review the different possible contributors to T cell activation in AT, such as the CD28 and CD154 T cell costimulatory molecules in AT. FUTURE DIRECTIONS: The potential antigen presentation capacities of adipocytes should be further investigated. Moreover, the properties of these AT resident (or migrating to AT) T cells must be further assessed. Antioxid. Redox Signal. 26, 489-500.


Subject(s)
Immunomodulation , Lymphocyte Activation/immunology , Obesity/immunology , T-Lymphocytes/immunology , Animals , Biomarkers , Cell Communication , Cell Differentiation/drug effects , Humans , Inflammation/complications , Inflammation/immunology , Inflammation/metabolism , Lymphocyte Activation/drug effects , Mice , Obesity/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Vitamin D/metabolism
13.
J Cell Sci ; 129(11): 2273-84, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27122188

ABSTRACT

Glucose transporter GLUT4 (also known as SLC2A4) plays a major role in glucose homeostasis and is efficiently retained intracellularly in adipocytes and myocytes. To simplify the analysis of its retention, here, various intracellular GLUT4 domains were fused individually to reporter molecules. Of the four short cytoplasmic loops of GLUT4, only the first nine-residue-long loop conferred intracellular retention of truncated forms of the transferrin receptor and CD4 in adipocytes. In contrast, the same loop of GLUT1 was without effect. The reporter molecules to which the first loop of GLUT4 was fused localized, unlike GLUT4, to the trans-Golgi network (TGN), possibly explaining why these molecules did not respond to insulin. The retention induced by the GLUT4 loop was specific to adipocytes as it did not induce retention in preadipocytes. Of the SQWLGRKRA sequence that constitutes this loop, mutation of either the tryptophan or lysine residue abrogated reporter retention. Mutation of these residues individually into alanine residues in the full-length GLUT4 molecule resulted in a decreased retention for GLUT4-W105A. We conclude that the first intracellular loop of GLUT4 contains the retention motif WLGRK, in which W105 plays a prominent role.


Subject(s)
Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Amino Acid Motifs , Animals , CD4 Antigens/metabolism , DNA Mutational Analysis , Genes, Reporter , Insulin/pharmacology , Intracellular Space/metabolism , Mice , Mutation/genetics , Protein Domains , Protein Structure, Secondary , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , trans-Golgi Network/drug effects , trans-Golgi Network/metabolism
14.
J Biol Chem ; 290(5): 2812-21, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25527501

ABSTRACT

Gaining the full activity of the insulin receptor (IR) requires the proteolytic cleavage of its proform by intra-Golgi furin-like activity. In mammalian cells, IR is expressed as two isoforms (IRB and IRA) that are responsible for insulin action. However, only IRA transmits the growth-promoting and mitogenic effects of insulin-like growth factor 2. Here we demonstrate that the two IR isoforms are similarly cleaved by furin, but when this furin-dependent maturation is inefficient, IR proforms move to the cell surface where the proprotein convertase PACE4 selectively supports IRB maturation. Therefore, in situations of impaired furin activity, the proteolytic maturation of IRB is greater than that of IRA, and accordingly, the amount of phosphorylated IRB is also greater than that of IRA. We highlight the ability of a particular proprotein convertase inhibitor to effectively reduce the maturation of IRA and its associated mitogenic signaling without altering the signals emanating from IRB. In conclusion, the selective PACE4-dependent maturation of IRB occurs when furin activity is reduced; accordingly, the pharmacological inhibition of furin reduces IRA maturation and its mitogenic potential without altering the insulin effects.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Proprotein Convertases/metabolism , Receptor, Insulin/metabolism , Serine Endopeptidases/metabolism , 3T3-L1 Cells , Animals , Cell Proliferation , Furin/genetics , Furin/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Mice , Proprotein Convertases/genetics , Real-Time Polymerase Chain Reaction , Receptor, Insulin/genetics , Serine Endopeptidases/genetics
15.
Proc Natl Acad Sci U S A ; 111(7): 2686-91, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24492375

ABSTRACT

The immune system plays an instrumental role in obesity and insulin resistance. Here, we unravel the role of the costimulatory molecule CD40 and its signaling intermediates, TNF receptor-associated factors (TRAFs), in diet-induced obesity (DIO). Although not exhibiting increased weight gain, male CD40(-/-) mice in DIO displayed worsened insulin resistance, compared with wild-type mice. This worsening was associated with excessive inflammation of adipose tissue (AT), characterized by increased accumulation of CD8(+) T cells and M1 macrophages, and enhanced hepatosteatosis. Mice with deficient CD40-TRAF2/3/5 signaling in MHCII(+) cells exhibited a similar phenotype in DIO as CD40(-/-) mice. In contrast, mice with deficient CD40-TRAF6 signaling in MHCII(+) cells displayed no insulin resistance and showed a reduction in both AT inflammation and hepatosteatosis in DIO. To prove the therapeutic potential of inhibition of CD40-TRAF6 in obesity, DIO mice were treated with a small-molecule inhibitor that we designed to specifically block CD40-TRAF6 interactions; this compound improved insulin sensitivity, reduced AT inflammation, and decreased hepatosteatosis. Our study reveals that the CD40-TRAF2/3/5 signaling pathway in MHCII(+) cells protects against AT inflammation and metabolic complications associated with obesity whereas CD40-TRAF6 interactions in MHCII(+) cells aggravate these complications. Inhibition of CD40-TRAF6 signaling by our compound may provide a therapeutic option in obesity-associated insulin resistance.


Subject(s)
CD40 Antigens/metabolism , Insulin Resistance/immunology , Obesity/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 6/metabolism , Adipose Tissue/cytology , Adipose Tissue/immunology , Adipose Tissue/pathology , Analysis of Variance , Animals , Azo Compounds , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Calorimetry , Fatty Liver/etiology , Fatty Liver/pathology , Flow Cytometry , Ligands , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/complications , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Surface Plasmon Resonance , TNF Receptor-Associated Factor 6/antagonists & inhibitors
16.
Biochim Biophys Acta ; 1833(3): 602-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23159491

ABSTRACT

The pleiotropic pro-inflammatory cytokine tumour necrosis factor alpha (TNF) is synthesised as a transmembrane protein that is subject to palmitoylation. In this study, the roles of this acylation on TNF-mediated biological effects were investigated. We found that the lipid raft partitioning of TNF is regulated by its palmitoylation. Furthermore, we demonstrated that this palmitoylation process interferes with the cleavage/degradation of TNF intracellular fragments but is not involved in the regulation of its ectodomain shedding. Moreover, we found that the palmitoylation of TNF hinders the binding of soluble TNF to TNFR1 and regulates the integration/retention of TNFR1 into lipid rafts. Finally, we demonstrate that the transmembrane forms of wild-type and palmitoylation-defective TNF interact differently with TNFR1 and regulate NFκB activity, Erk1/2 phosphorylation and interleukin-6 synthesis differently, strongly suggesting that palmitoylation of TNF is involved in the regulation of TNFR1 signalling. An evidence for the physiological intervention of this regulation is provided by the fact that, in macrophages, the binding of endogenous soluble TNF to TNFR1 is enhanced by inhibition of palmitoylation. Therefore, our data introduce the new concept that palmitoylation of TNF is one of the means by which TNF-producing cells regulate their sensitivity to soluble TNF.


Subject(s)
Gene Expression Regulation , Membrane Microdomains/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Animals , Blotting, Western , Caspase 8/genetics , Caspase 8/metabolism , Cells, Cultured , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoenzyme Techniques , Immunoprecipitation , Interleukin-6/genetics , Interleukin-6/metabolism , Lipoylation , Luciferases/metabolism , Macrophages/cytology , Macrophages/metabolism , Membrane Microdomains/genetics , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Type I/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics
17.
Arterioscler Thromb Vasc Biol ; 32(10): 2394-404, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859493

ABSTRACT

OBJECTIVE: High endogenous thrombin potential (ETP) is associated with venous and arterial thrombosis. Better knowledge of environmental influences on ETP may help to prevent thrombosis. METHODS AND RESULTS: Weaning rats exhibited high ETP values that decreased in low-fat diet and remained elevated on high-fat diet. In adult rats, high-fat diet-induced ETP increase was independent of coagulation factors, obesity, and insulin resistance and negatively associated with polyunsaturated fatty acid levels. Switching from high-fat diet to low-fat diet reversed the procoagulant phenotype with a slower kinetic than the normalization of hyperinsulinemia. In humans, ETP was independent of body weight whereas it was negatively associated with nutritional markers such as the percentage of energy provided by proteins, the protein:fat ratio, circulating phenolic compounds, and omega-3 polyunsaturated fatty acid. A recommended 3-month healthy diet with reduced energy density, including lipids, decreased ETP (-21%; P<0.0001). Changes in ETP were not associated with body weight, insulin sensitivity, or coagulation factor variations, but correlated negatively with plasma docosahexaenoic acid, a nutritional status sensitive fatty acid, and compounds reflecting vegetable intake. CONCLUSIONS: Diet plays a pivotal role in regulating ETP, independently of obesity and insulin resistance. Global nutritional recommendations could be useful in primary prevention of venous thrombosis.


Subject(s)
Diet, Fat-Restricted , Diet, High-Fat , Nutritional Status , Thrombin/metabolism , Thrombosis/epidemiology , Thrombosis/metabolism , Animals , Blood Coagulation/physiology , Fatty Acids, Omega-3/metabolism , Humans , Insulin Resistance/physiology , Middle Aged , Models, Animal , Obesity/physiopathology , Phenols/metabolism , Rats , Rats, Wistar , Risk Factors , Thrombosis/physiopathology , Time Factors
18.
Biochimie ; 94(12): 2569-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22835478

ABSTRACT

We have developed a rapid nonradioisotope chemiluminescent assay adapted to high-throughput screening experiments, to evaluate glucose uptake activity in cultured cells. For chemiluminescence quantification of 2-deoxyglucose, we used a luminol oxidation reaction after an enzymatic dephosphorylation of 2-deoxyglucose-6-phosphate. All reactions were performed at 37 °C by consecutive addition of reagents, and the assay is able to quantify 2DG in picomole per well. To confirm the reliability of this method, we have evaluated the dose-effect of insulin, GLUT4 inhibitors and insulin-sensitizing agent on 2DG uptake into 3T3-L1 cells. The results obtained with the assay for 2DG uptake in vitro in the absence or presence of insulin stimulation, were similar to those obtained by the previous radioisotopic and enzymatic methods. We have also used this assay to evaluate the effect of various reactive carbonyl and oxygen species on insulin-stimulated 2DG-uptake into adipocytes. All reactive carbonyl species tested decreased insulin-stimulated glucose uptake in a time- and dose-dependent manner without affecting basal glucose uptake in 3T3-L1 cells. 4-hydroxynonenal was found to be the most potent in the impairment of glucose uptake. This new enzymatic chemiluminescent assay is rapid and useful for measurement of 2DG uptake in insulin-responsive in cultured cells.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Deoxyglucose/pharmacokinetics , Luminescent Measurements/methods , 3T3-L1 Cells , Acrolein/pharmacology , Aldehydes/pharmacology , Animals , Biological Transport/drug effects , Chromans/pharmacology , Cytochalasin B/pharmacology , Deoxyglucose/metabolism , Hydrogen Peroxide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Luminescent Measurements/instrumentation , Mice , Pyruvaldehyde/pharmacology , Reproducibility of Results , Thiazolidinediones/pharmacology , Troglitazone
19.
Circ Res ; 109(12): 1387-95, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22021930

ABSTRACT

RATIONALE: Unlike conventional dendritic cells, plasmacytoid DCs (PDC) are poor in antigen presentation and critical for type I interferon response. Though proposed to be present in human atherosclerotic lesions, their role in atherosclerosis remains elusive. OBJECTIVE: To investigate the role of PDC in atherosclerosis. METHODS AND RESULTS: We show that PDC are scarcely present in human atherosclerotic lesions and almost absent in mouse plaques. Surprisingly, PDC depletion by 120G8 mAb administration was seen to promote plaque T-cell accumulation and exacerbate lesion development and progression in LDLr⁻/⁻ mice. PDC depletion was accompanied by increased CD4⁺ T-cell proliferation, interferon-γ expression by splenic T cells, and plasma interferon-γ levels. Lymphoid tissue PDC from atherosclerotic mice showed increased indoleamine 2,3-dioxygenase (IDO) expression and IDO blockage abrogated the PDC suppressive effect on T-cell proliferation. CONCLUSIONS: Our data reveal a protective role for PDC in atherosclerosis, possibly by dampening T-cell proliferation and activity in peripheral lymphoid tissue, rendering PDC an interesting target for future therapeutic interventions.


Subject(s)
Atherosclerosis/pathology , Atherosclerosis/physiopathology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Dendritic Cells/pathology , Dendritic Cells/physiology , Animals , Antibodies, Monoclonal/pharmacology , Atherosclerosis/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/drug effects , Disease Models, Animal , Disease Progression , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism
20.
Arterioscler Thromb Vasc Biol ; 31(10): 2251-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21817098

ABSTRACT

OBJECTIVE: Obese adipose tissue shows hallmarks of chronic inflammation, which promotes the development of metabolic disorders. The mechanisms by which immune cells interact with each other or with metabolism-associated cell types, and the players involved, are still unclear. The CD40-CD40L costimulatory dyad plays a pivotal role in immune responses and in diseases such as atherosclerosis and may therefore be a mediator of obesity. Here we investigated whether CD40L is involved in adipose tissue inflammation and its associated metabolic changes. METHODS AND RESULTS: To assess a putative role of CD40L in obesity in vivo, we evaluated metabolic and inflammatory consequences of 18 weeks of high-fat feeding in CD40L(+/+) and CD40L(-/-) mice. In addition, C57Bl6 mice were injected with neutralizing anti-CD40L (αCD40L) antibody for 12 weeks while being fed a high-fat diet. Genetic deficiency of CD40L attenuated the development of diet-induced obesity, hepatic steatosis, and increased systemic insulin sensitivity. In adipose tissue, it impaired obesity-induced immune cell infiltration and the associated deterioration of glucose and lipid metabolism. Accordingly, αCD40L treatment improved systemic insulin sensitivity, glucose tolerance, and CD4(+) T-cell infiltration in adipose tissue with limited effects on adipose tissue weight. CONCLUSIONS: CD40L plays a crucial role in the development of obesity-induced inflammation and metabolic complications.


Subject(s)
Adipose Tissue/immunology , CD40 Ligand/deficiency , Fatty Liver/prevention & control , Insulin Resistance , Obesity/prevention & control , Panniculitis/prevention & control , Adipose Tissue/metabolism , Animals , Antibodies, Neutralizing/administration & dosage , Blood Glucose/metabolism , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/immunology , Fatty Liver/metabolism , Inflammation Mediators/metabolism , Insulin/blood , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/complications , Obesity/genetics , Obesity/immunology , Obesity/metabolism , Obesity/physiopathology , Panniculitis/genetics , Panniculitis/immunology , Panniculitis/metabolism , Time Factors , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL
...