Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Turk J Med Sci ; 49(2): 490-496, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30866603

ABSTRACT

Background/aim: Genetic aspects play a role in insulin resistance in children. In this study, for the first time, the association of LRP5 (rs556442) polymorphism and insulin resistance in Iranian children and adolescents was investigated. Materials and methods: The study population comprises children and adolescents aged 9­18 years. Anthropometric and biochemical parameters were assessed. Insulin resistance/sensitivity was determined by the quantitative insulin sensitivity check index (QUICKI), homeostasis model assessment-insulin resistance (HOMA-IR), insulin-to-glucose ratio, McAuley index, revised McAuley index, fasting insulin resistance index (FIRI), and Bennett's index. LRP5 (rs566442) single nucleotide polymorphism (SNP) was identified using restriction fragment length polymorphism (RFLP). Linear regression analysis was used to determine the association between the LRP5 polymorphism (rs556442) and insulin sensitivity indexes. Results: Significant differences were found between GG genotype vs. AG/AA genotypes for McAuley index (P = 0.049) and revised McAuley index (P = 0.044) when adjusted for interaction factors (age, sex, and puberty) in regression models. No significant association was found between LRP5 (rs566442) and other insulin resistance indexes. Also, LRP5 (rs566442) did not show a significant impact on biochemical parameters. Conclusion: This study showed that LRP5 polymorphism (rs556442) was associated with insulin resistance in Iranian children and adolescents.


Subject(s)
Insulin Resistance/genetics , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Polymorphism, Single Nucleotide/genetics , Adolescent , Child , Cross-Sectional Studies , Female , Genetic Predisposition to Disease , Glucose Tolerance Test , Healthy Volunteers , Homeostasis/genetics , Humans , Iran , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Male , Urban Population
2.
J Cell Biochem ; 120(3): 3362-3366, 2019 03.
Article in English | MEDLINE | ID: mdl-30246479

ABSTRACT

We have previously demonstrated that Lrp5/6/ß-catenin plays an important role in valve calcification with a specific osteogenic phenotype defined by increased bone mineral content and overall valve thickening. Recent studies indicate that TIEG1 may be involved in mediating the Wnt signaling pathway in bone, which is known to play critical roles in osteoblast differentiation and bone mineralization. Therefore, we sought to test the role of TIEG1 in mediating Wnt signaling, in an established model of hypercholesterolemic valve disease. Our previous model treated null mice with cholesterol diets: Lrp5 -/- /ApoE -/- mice versus wild-type control (n = 180). Group I (n = 60) normal diet, Group II (n = 60) 0.25% chol diet (w/w), and Group III (n = 60) 0.25% (w/w) chol diet + atorv was tested for gene expression for TIEG1, Lrp6, and Runx2. Real-time polymerase chain reaction confirmed that there is upregulation of the gene expression for TIEG1 and Runx2 in the hypercholesterolemic double knockout and single knockout valves as compared with controls with a mild increase in Lrp6. To confirm the mechanism, coexpression of ß-catenin, TIEG1, and LEF1 in valve cells in vitro, led to the coactivation of the TOPFLASH reporter, which was further confirmed by the observation that TIEG1 and ß-catenin colocalize with one another in the nucleus of valvular interstitial cells (VICs) following stimulation with transforming growth factor-ß treatment, an established activator of TIEG1. Taken together, these data implicate an important role for TIEG1 in mediating valve osteogenesis.


Subject(s)
Aortic Valve Stenosis/pathology , Calcinosis/pathology , DNA-Binding Proteins/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice, Knockout, ApoE/physiology , Osteogenesis , Transcription Factors/physiology , Animals , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Calcinosis/genetics , Calcinosis/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology
3.
FEBS J ; 284(11): 1657-1671, 2017 06.
Article in English | MEDLINE | ID: mdl-28425175

ABSTRACT

Snake venom metalloproteases (SVMPs) are members of the a disintegrin and metalloprotease (ADAM) family of proteins, as they possess similar domains. SVMPs are known to elicit snake venom-induced haemorrhage; however, the target proteins and cleavage sites are not known. In this work, we identified a target protein of vascular apoptosis-inducing protein 1 (VAP1), an SVMP, relevant to its ability to induce haemorrhage. VAP1 disrupted cell-cell adhesions by relocating VE-cadherin and γ-catenin from the cell-cell junction to the cytosol, without inducing proteolysis of VE-cadherin. The Wnt receptors low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) are known to promote catenin relocation, and are rendered constitutively active in Wnt signalling by truncation. Thus, we examined whether VAP1 cleaves LRP5/6 to induce catenin relocation. Indeed, we found that VAP1 cleaved the extracellular region of LRP6 and LRP5. This cleavage removes four inhibitory ß-propeller structures, resulting in activation of LRP5/6. Recombinant human ADAM8 and ADAM12 also cleaved LRP6 at the same site. An antibody against a peptide including the LRP6-cleavage site inhibited VAP1-induced VE-cadherin relocation and disruption of cell-cell adhesions in cultured cells, and blocked haemorrhage in mice in vivo. Intriguingly, animals resistant to the effects of haemorrhagic snake venom express variants of LRP5/6 that lack the VAP1-cleavage site, or low-density lipoprotein receptor domain class A domains involved in formation of the constitutively active form. The results validate LRP5/6 as physiological targets of ADAMs. Furthermore, they indicate that SVMP-induced cleavage of LRP5/6 causes disruption of cell-cell adhesion and haemorrhage, potentially opening new avenues for the treatment of snake bites.


Subject(s)
ADAM Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Crotalid Venoms/metabolism , Hemorrhage/chemically induced , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Metalloendopeptidases/metabolism , ADAM Proteins/pharmacology , ADAM12 Protein/metabolism , ADAM12 Protein/pharmacology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Drug Resistance , Fibrinogen/metabolism , Fibronectins/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Hydrophobic and Hydrophilic Interactions , Low Density Lipoprotein Receptor-Related Protein-5/chemistry , Low Density Lipoprotein Receptor-Related Protein-6/chemistry , Male , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Mice , Models, Molecular , Molecular Docking Simulation , Protein Domains , Protein Structure, Secondary/drug effects , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Vertebrates/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology
4.
PLoS One ; 10(11): e0140775, 2015.
Article in English | MEDLINE | ID: mdl-26554834

ABSTRACT

The low density lipoprotein receptor-related protein-5 (LRP5), a co-receptor in the Wnt signaling pathway, modulates bone mass in humans and in mice. Lrp5 knock-out mice have severely impaired responsiveness to mechanical stimulation whereas Lrp5 gain-of-function knock-in and transgenic mice have enhanced responsiveness to mechanical stimulation. Those observations highlight the importance of Lrp5 protein in bone cell mechanotransduction. It is unclear if and how high bone mass-causing (HBM) point mutations in Lrp5 alter the bone-wasting effects of mechanical disuse. To address this issue we explored the skeletal effects of mechanical disuse using two models, tail suspension and Botulinum toxin-induced muscle paralysis, in two different Lrp5 HBM knock-in mouse models. A separate experiment employing estrogen withdrawal-induced bone loss by ovariectomy was also conducted as a control. Both disuse stimuli induced significant bone loss in WT mice, but Lrp5 A214V and G171V were partially or fully protected from the bone loss that normally results from disuse. Trabecular bone parameters among HBM mice were significantly affected by disuse in both models, but these data are consistent with DEXA data showing a failure to continue growing in HBM mice, rather than a loss of pre-existing bone. Ovariectomy in Lrp5 HBM mice resulted in similar protection from catabolism as was observed for the disuse experiments. In conclusion, the Lrp5 HBM alleles offer significant protection from the resorptive effects of disuse and from estrogen withdrawal, and consequently, present a potential mechanism to mimic with pharmaceutical intervention to protect against various bone-wasting stimuli.


Subject(s)
Bone Density/physiology , Bone Diseases, Metabolic/prevention & control , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Mutation, Missense , Point Mutation , Animals , Bone Density/genetics , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/pathology , Botulinum Toxins/toxicity , Disease Models, Animal , Estrogens/deficiency , Estrogens/physiology , Female , Femur/pathology , Gene Knock-In Techniques , Humans , Immobilization/adverse effects , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Mice , Osteoporosis, Postmenopausal/pathology , Osteoporosis, Postmenopausal/prevention & control , Ovariectomy/adverse effects , Paralysis/chemically induced , Paralysis/complications , Paralysis/pathology , Stress, Mechanical , Weight-Bearing
5.
Immunol Cell Biol ; 93(7): 653-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25748163

ABSTRACT

Inflammation is triggered after invasion or injury to restore homeostasis. Although the activation of Wnt/ß-catenin signaling is one of the first molecular responses to cellular damage, its role in inflammation is still unclear. It was our hypothesis that the low-density lipoprotein (LDL) receptor-related protein 5 (LRP5) and the canonical Wnt signaling pathway are modulators of inflammatory mechanisms. Wild-type (WT) and LRP5(-/-) mice were fed a hypercholesterolemic (HC) diet to trigger dislipidemia and chronic inflammation. Diets were supplemented with plant sterol esters (PSEs) to induce LDL cholesterol lowering and the reduction of inflammation. HC WT mice showed increased serum cholesterol levels that correlated with increased Lrp5 and Wnt/ß-catenin gene expression while in the HC LRP5(-/-) mice Wnt/ß-catenin pathway was shut down. Functionally, HC induced pro-inflammatory gene expression in LRP5(-/-) mice, suggesting an inhibitory role of the Wnt pathway in inflammation. Dietary PSE administration downregulated serum cholesterol levels in WT and LRP5(-/-) mice. Furthermore, in WT mice PSE increased anti-inflammatory genes expression and inhibited Wnt/ß-catenin activation. Hepatic gene expression of Vldlr, Lrp2 and Lrp6 was increased after HC feeding in WT mice but not in LRP5(-/-) mice, suggesting a role for these receptors in the clearance of plasmatic lipoproteins. Finally, an antiatherogenic role for LRP5 was demonstrated as HC LRP5(-/-) mice developed larger aortic atherosclerotic lesions than WT mice. Our results show an anti-inflammatory, pro-survival role for LRP5 and the Wnt signaling pathway in peripheral blood leukocytes.


Subject(s)
Cholesterol/blood , Hypercholesterolemia/blood , Leukocytes/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Wnt Signaling Pathway , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/therapy , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/therapy , Cholesterol, Dietary/toxicity , Cholesterol, HDL/blood , Humans , Hypercholesterolemia/diet therapy , Jejunum/metabolism , Lipoproteins/biosynthesis , Lipoproteins/genetics , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/deficiency , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Phytosterols/therapeutic use , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Spleen/metabolism
6.
J Clin Invest ; 124(9): 3825-46, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25083995

ABSTRACT

Canonical WNT signaling is required for proper vascularization of the CNS during embryonic development. Here, we used mice with targeted mutations in genes encoding canonical WNT pathway members to evaluate the exact contribution of these components in CNS vascular development and in specification of the blood-brain barrier (BBB) and blood-retina barrier (BRB). We determined that vasculature in various CNS regions is differentially sensitive to perturbations in canonical WNT signaling. The closely related WNT signaling coreceptors LDL receptor-related protein 5 (LRP5) and LRP6 had redundant functions in brain vascular development and barrier maintenance; however, loss of LRP5 alone dramatically altered development of the retinal vasculature. The BBB in the cerebellum and pons/interpeduncular nuclei was highly sensitive to decrements in canonical WNT signaling, and WNT signaling was required to maintain plasticity of barrier properties in mature CNS vasculature. Brain and retinal vascular defects resulting from ablation of Norrin/Frizzled4 signaling were ameliorated by stabilizing ß-catenin, while inhibition of ß-catenin-dependent transcription recapitulated the vascular development and barrier defects associated with loss of receptor, coreceptor, or ligand, indicating that Norrin/Frizzled4 signaling acts predominantly through ß-catenin-dependent transcriptional regulation. Together, these data strongly support a model in which identical or nearly identical canonical WNT signaling mechanisms mediate neural tube and retinal vascularization and maintain the BBB and BRB.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Retinal Barrier/physiology , Neovascularization, Physiologic/physiology , Wnt Signaling Pathway/physiology , Animals , Eye Proteins/genetics , Frizzled Receptors/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Mice , Nerve Tissue Proteins/genetics , Retina/physiology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , beta Catenin/physiology
7.
Clin Calcium ; 24(6): 853-61, 2014 Jun.
Article in Japanese | MEDLINE | ID: mdl-24870836

ABSTRACT

Bone is constantly renewed by the balanced action of osteoblastic bone formation and osteoclastic bone resorption both of which mainly occur at the bone surface. This restructuring process called "bone remodeling" is important not only for normal bone mass and strength, but also for mineral homeostasis. Coupling has been understood as a balanced induction of osteoblastic bone formation in response to osteoclastic bone resorption. An imbalance of this coupling is often linked to various bone diseases. TGF-ß and IGF released from bone matrix during osteoclastic bone resorption are the favored candidates as classical coupling factor. Recently, several reports suggest that osteoclast-derived molecules/cytokines (clastokine) mediate directional signaling between osteoblasts and osteoclasts into the bone microenvironment. Thus, the elucidation of the regulatory mechanisms involved in bone cell communication and coupling is critical for a deeper understanding of the skeletal system in health and disease.


Subject(s)
Bone and Bones/cytology , Bone and Bones/physiology , Cell Communication/genetics , Cell Communication/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Animals , Antigens, CD/physiology , Bone Morphogenetic Protein 6/physiology , Bone Remodeling/genetics , Bone Remodeling/physiology , Cellular Microenvironment/genetics , Cellular Microenvironment/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Mice , NFATC Transcription Factors/metabolism , Osteoprotegerin/physiology , Proprotein Convertases/physiology , RANK Ligand/physiology , Semaphorin-3A/physiology , Semaphorins/physiology , Serine Endopeptidases/physiology , Somatomedins/physiology , Transforming Growth Factor beta/physiology , Wnt Signaling Pathway/physiology
10.
Sci Transl Med ; 5(211): 211ra158, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24225945

ABSTRACT

Osteoporosis pseudoglioma syndrome (OPPG) is a rare genetic disease that produces debilitating effects in the skeleton. OPPG is caused by mutations in LRP5, a WNT co-receptor that mediates osteoblast activity. WNT signaling through LRP5, and also through the closely related receptor LRP6, is inhibited by the protein sclerostin (SOST). It is unclear whether OPPG patients might benefit from the anabolic action of sclerostin neutralization therapy (an approach currently being pursued in clinical trials for postmenopausal osteoporosis) in light of their LRP5 deficiency and consequent osteoblast impairment. To assess whether loss of sclerostin is anabolic in OPPG, we measured bone properties in a mouse model of OPPG (Lrp5(-/-)), a mouse model of sclerosteosis (Sost(-/-)), and in mice with both genes knocked out (Lrp5(-/-);Sost(-/-)). Lrp5(-/-);Sost(-/-) mice have larger, denser, and stronger bones than do Lrp5(-/-) mice, indicating that SOST deficiency can improve bone properties via pathways that do not require LRP5. Next, we determined whether the anabolic effects of sclerostin depletion in Lrp5(-/-) mice are retained in adult mice by treating 17-week-old Lrp5(-/-) mice with a sclerostin antibody for 3 weeks. Lrp5(+/+) and Lrp5(-/-) mice each exhibited osteoanabolic responses to antibody therapy, as indicated by increased bone mineral density, content, and formation rates. Collectively, our data show that inhibiting sclerostin can improve bone mass whether LRP5 is present or not. In the absence of LRP5, the anabolic effects of SOST depletion can occur via other receptors (such as LRP4/6). Regardless of the mechanism, our results suggest that humans with OPPG might benefit from sclerostin neutralization therapies.


Subject(s)
Bone and Bones/physiopathology , Disease Models, Animal , Glycoproteins/genetics , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Osteogenesis Imperfecta/physiopathology , Adaptor Proteins, Signal Transducing , Animals , Bone Development , Glycoproteins/physiology , Intercellular Signaling Peptides and Proteins , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mice , Mice, Knockout , Organ Size
11.
Nat Med ; 19(2): 179-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23389618

ABSTRACT

Low bone mass and strength lead to fragility fractures, for example, in elderly individuals affected by osteoporosis or children with osteogenesis imperfecta. A decade ago, rare human mutations affecting bone negatively (osteoporosis-pseudoglioma syndrome) or positively (high-bone mass phenotype, sclerosteosis and Van Buchem disease) have been identified and found to all reside in components of the canonical WNT signaling machinery. Mouse genetics confirmed the importance of canonical Wnt signaling in the regulation of bone homeostasis, with activation of the pathway leading to increased, and inhibition leading to decreased, bone mass and strength. The importance of WNT signaling for bone has also been highlighted since then in the general population in numerous genome-wide association studies. The pathway is now the target for therapeutic intervention to restore bone strength in millions of patients at risk for fracture. This paper reviews our current understanding of the mechanisms by which WNT signalng regulates bone homeostasis.


Subject(s)
Bone and Bones/metabolism , Homeostasis , Mutation , Wnt Signaling Pathway/physiology , Animals , Bone Density , Bone Development , Bone Morphogenetic Proteins/physiology , Hematopoiesis , Humans , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Mice , beta Catenin/physiology
12.
J Clin Invest ; 122(10): 3490-503, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945629

ABSTRACT

Serotonin is a critical regulator of bone mass, fulfilling different functions depending on its site of synthesis. Brain-derived serotonin promotes osteoblast proliferation, whereas duodenal-derived serotonin suppresses it. To understand the molecular mechanisms of duodenal-derived serotonin action on osteoblasts, we explored its transcriptional mediation in mice. We found that the transcription factor FOXO1 is a crucial determinant of the effects of duodenum-derived serotonin on bone formation We identified two key FOXO1 complexes in osteoblasts, one with the transcription factor cAMP-responsive element-binding protein 1 (CREB) and another with activating transcription factor 4 (ATF4). Under normal levels of circulating serotonin, the proliferative activity of FOXO1 was promoted by a balance between its interaction with CREB and ATF4. However, high circulating serotonin levels prevented the association of FOXO1 with CREB, resulting in suppressed osteoblast proliferation. These observations identify FOXO1 as the molecular node of an intricate transcriptional machinery that confers the signal of duodenal-derived serotonin to inhibit bone formation.


Subject(s)
Bone Remodeling/physiology , Duodenum/metabolism , Forkhead Transcription Factors/physiology , Osteoblasts/physiology , Serotonin/physiology , Activating Transcription Factor 4/physiology , Animals , Blood-Brain Barrier , Cell Division/drug effects , Cells, Cultured/drug effects , Cyclic AMP Response Element-Binding Protein/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genes, Reporter , Genotype , Homeostasis/physiology , Insulin-Like Growth Factor I/genetics , Low Density Lipoprotein Receptor-Related Protein-5/deficiency , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/physiology , MAP Kinase Signaling System/drug effects , Mice , Organ Specificity , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/physiology , Promoter Regions, Genetic , Receptor, Serotonin, 5-HT1B/deficiency , Receptor, Serotonin, 5-HT1B/genetics , Receptor, Serotonin, 5-HT1B/physiology , Serotonin/blood , Serotonin/pharmacology , Stress, Physiological/genetics , Transcription, Genetic
13.
J Biol Chem ; 287(15): 12016-26, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22337886

ABSTRACT

Disturbed Wnt signaling has been implicated in numerous diseases, including type 2 diabetes and the metabolic syndrome. In the present study, we have investigated cross-talk between insulin and Wnt signaling pathways using preadipocytes with and without knockdown of the Wnt co-receptors LRP5 and LRP6 and with and without knock-out of insulin and IGF-1 receptors. We find that Wnt stimulation leads to phosphorylation of insulin signaling key mediators, including Akt, GSK3ß, and ERK1/2, although with a lower fold stimulation and slower time course than observed for insulin. These Wnt effects are insulin/IGF-1 receptor-dependent and are lost in insulin/IGF-1 receptor double knock-out cells. Conversely, in LRP5 knockdown preadipocytes, insulin-induced phosphorylation of IRS1, Akt, GSK3ß, and ERK1/2 is highly reduced. This effect is specific to insulin, as compared with IGF-1, stimulation and appears to be due to an inducible interaction between LRP5 and the insulin receptor as demonstrated by co-immunoprecipitation. These data demonstrate that Wnt and insulin signaling pathways exhibit cross-talk at multiple levels. Wnt induces phosphorylation of Akt, ERK1/2, and GSK3ß, and this is dependent on insulin/IGF-1 receptors. Insulin signaling also involves the Wnt co-receptor LRP5, which has a positive effect on insulin signaling. Thus, altered Wnt and LRP5 activity can serve as modifiers of insulin action and insulin resistance in the pathophysiology of diabetes and metabolic syndrome.


Subject(s)
Adipocytes/metabolism , Insulin/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Receptor Cross-Talk , Wnt Signaling Pathway , 3T3-L1 Cells , Animals , Gene Expression Regulation , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunoprecipitation , Insulin/metabolism , Kinetics , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , MAP Kinase Signaling System , Mice , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Wnt3A Protein/physiology , beta Catenin/metabolism
14.
Endocrinol Nutr ; 59(3): 207-14, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22325788

ABSTRACT

The aim of this paper is to review the main aspects related to high bone density (HBD) as well as to discuss the physiologic mechanisms involved in bone health. There are still no well-defined criteria for identification of individuals with HBD and there are few studies on the topic. Most studies demonstrate that overweight, male gender, black ethnic background, physical activity, calcium and fluoride intake and use of medications such as statins and thiazide diuretics play a relevant and positive role on bone mineral density. Moreover, it is known that individuals with certain diseases such as obesity, diabetes, estrogen receptor-positive breast or endometrial cancer have greater bone density than healthy individuals, as well as athletes having higher bone density than non-athletes does not necessarily mean that they have healthy bones. A better understanding of risk and protective factors may help in the management of patients with bone frailty and have applicability in the treatment and in the prevention of osteoporosis, especially intervening on non-modifiable risk factors.


Subject(s)
Bone Density , Bone and Bones/metabolism , Hyperostosis/etiology , Animals , Athletes , Body Composition , Body Weight , Bone Density/genetics , Bone Density/physiology , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/physiopathology , Bone Remodeling , Calcium/metabolism , Diet , Diphosphonates/pharmacology , Endocrine System Diseases/complications , Endocrine System Diseases/physiopathology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hyperostosis/genetics , Hyperostosis/physiopathology , Lipid Metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Male , Osteoblasts/metabolism , Sodium Chloride Symporter Inhibitors/pharmacology , Vitamin D/pharmacology , Weight-Bearing , Wnt Proteins/physiology
15.
Curr Osteoporos Rep ; 10(1): 93-100, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22210558

ABSTRACT

Osteoporosis is a common disorder in which diminished bone mass leads to progressive microarchitectural skeletal deterioration and increased fracture risk. Our understanding of both normal and pathologic bone biology continues to evolve, and with it our grasp of the highly coordinated relationships between primary bone cells (osteoblasts, osteoclasts, and osteocytes) and the complex molecular signals bone cells use to integrate signals derived from other organ systems, including the immune, hematopoietic, gastrointestinal, and central nervous systems. It is now clear that the Wnt signaling pathway is central to regulation of both skeletal modeling and remodeling. Herein, we discuss components of the Wnt signaling pathway (DKK1, an endogenous soluble inhibitor of Wnt signaling) and LRP5 (a plasma membrane-localized Wnt co-receptor) as potential future targets for osteoporosis therapy. Finally, we discuss the current controversial role for serotonin in skeletal metabolism, and the potential role of future therapies targeting serotonin for osteoporosis treatment.


Subject(s)
Bone and Bones/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Osteoporosis/drug therapy , Serotonin/physiology , Wnt Signaling Pathway/drug effects , Animals , Antibodies, Neutralizing , Bone Resorption/physiopathology , Bone and Bones/drug effects , Humans , Osteoblasts/physiology , Osteoporosis/metabolism , Osteoporosis/physiopathology , Wnt Signaling Pathway/physiology
16.
Circulation ; 124(17): 1871-81, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21969016

ABSTRACT

BACKGROUND: Ischemic proliferative retinopathy, characterized by pathological retinal neovascularization, is a major cause of blindness in working-age adults and children. Defining the molecular pathways distinguishing pathological neovascularization from normal vessels is critical to controlling these blinding diseases with targeted therapy. Because mutations in Wnt signaling cause defective retinal vasculature in humans with some characteristics of the pathological vessels in retinopathy, we investigated the potential role of Wnt signaling in pathological retinal vascular growth in proliferative retinopathy. METHODS AND RESULTS: In this study, we show that Wnt receptors (Frizzled4 and low-density lipoprotein receptor-related protein5 [Lrp5]) and activity are significantly increased in pathological neovascularization in a mouse model of oxygen-induced proliferative retinopathy. Loss of Wnt coreceptor Lrp5 and downstream signaling molecule dishevelled2 significantly decreases the formation of pathological retinal neovascularization in retinopathy. Loss of Lrp5 also affects retinal angiogenesis during development and formation of the blood-retinal barrier, which is linked to significant downregulation of tight junction protein claudin5 in Lrp5(-/-) vessels. Blocking claudin5 significantly suppresses Wnt pathway-driven endothelial cell sprouting in vitro and developmental and pathological vascular growth in retinopathy in vivo. CONCLUSIONS: These results demonstrate an important role of Wnt signaling in pathological vascular development in retinopathy and show a novel function of Cln5 in promoting angiogenesis.


Subject(s)
Cell Proliferation , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Frizzled Receptors/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Neovascularization, Pathologic/metabolism , Receptors, Wnt/physiology , Retina/pathology , Wnt Signaling Pathway/physiology , Animals , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/growth & development , Frizzled Receptors/biosynthesis , Humans , Low Density Lipoprotein Receptor-Related Protein-5/biosynthesis , Lysosomal Membrane Proteins , Membrane Glycoproteins/biosynthesis , Mice , Mice, Knockout , Neovascularization, Pathologic/pathology , Receptors, Wnt/biosynthesis , Retina/growth & development , Retina/physiology
17.
FEBS J ; 278(24): 4704-16, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21902810

ABSTRACT

Tissue transglutaminase (TG2) is a ubiquitously expressed member of the transglutaminase family of Ca(2+)-dependent crosslinking enzymes. Unlike other family members, TG2 is a multifunctional protein, which has several other well documented enzymatic and non-enzymatic functions. A significant body of evidence accumulated over the last decade reveals multiple and complex activities of this protein on the cell surface and in the extracellular matrix (ECM), including its role in the regulation of cell-ECM interactions and outside-in signaling by several types of transmembrane receptors. Moreover, recent findings indicate a dynamic regulation of the levels and functions of extracellular TG2 by several complementary mechanisms. This review summarizes and assesses recent research into the emerging functions and regulation of extracellular TG2.


Subject(s)
Extracellular Matrix/metabolism , GTP-Binding Proteins/physiology , Transglutaminases/physiology , Cell Adhesion/physiology , Cross-Linking Reagents/metabolism , Cysteine/metabolism , Enzyme Activation , Fibronectins/metabolism , GTP-Binding Proteins/metabolism , Humans , Integrins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Macrophages/physiology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Mesenchymal Stem Cell Transplantation , Protein Conformation/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport/physiology , Receptors, Growth Factor/physiology , Signal Transduction/physiology , Syndecan-4/physiology , Transglutaminases/metabolism , beta Catenin/physiology
18.
Eur Heart J ; 32(22): 2841-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21398644

ABSTRACT

AIMS: Atherosclerosis plaque development includes infiltration of inflammatory cells, accumulation of lipids and fibrous cap formation. Low-density lipoprotein receptor-related protein 1 (LRP1) is expressed on atherosclerotic lesions associated with macrophages and vascular smooth muscle cells. The aim of this work is to analyse the role in atherosclerosis lesion progression of another member of the LDL receptor protein family, low-density lipoprotein receptor-related protein 5 (LRP5), a co-receptor with Frizzled known to activate the Wnt signalling pathway in several cell types. METHODS AND RESULTS: LRP5 is expressed in human vascular and innate inflammatory cells. LRP5 is transcriptionally regulated by aggregated LDL (agLDL), participating in the lipid uptake and transformation of macrophages into foam cells, a critical step in atherosclerosis progression. AgLDL-treated macrophages show up-regulated expression of ß-catenin, LEF1, c-jun, cyclinD1, bone morphogenetic protein 2 (BMP2), and osteopontin (OPN), proteins and targets of the Wnt signalling pathway, whereas LRP5-silenced macrophages show a significant down-regulation of OPN and BMP2 expression. Furthermore, LRP5-deficient macrophages exhibit an impaired migration both in wound-repair and modified Boyden chambers models. CONCLUSION: These results demonstrate the involvement of LRP5 in the innate inflammatory reaction to lipid infiltration in atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Cell Movement/physiology , Lipid Metabolism/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Macrophages/metabolism , Wnt Signaling Pathway/physiology , Apoptosis/physiology , Atherosclerosis/metabolism , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Foam Cells/physiology , Humans , Lipoproteins, LDL/pharmacology , Low Density Lipoprotein Receptor-Related Protein-5/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Monocytes/metabolism , Osteopontin/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...