Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Front Immunol ; 12: 768682, 2021.
Article in English | MEDLINE | ID: mdl-34745143

ABSTRACT

Background: Previous studies have suggested essential roles of growth factors on the risk of Multiple Sclerosis (MS), but it remains undefined whether the effects are causal. Objective: We applied Mendelian randomization (MR) approaches to disentangle the causal relationship between genetically predicted circulating levels of growth factors and the risk of MS. Methods: Genetic instrumental variables for fibroblast growth factor (FGF) 23, growth differentiation factor 15 (GDF15), insulin growth factor 1 (IGF1), insulin-like growth factor binding proteins 3 (IGFBP3) and vascular endothelial growth factor (VEGF) were obtained from up-to-date genome-wide association studies (GWAS). Summary-level statistics of MS were obtained from the International Multiple Sclerosis Genetics Consortium, incorporating 14,802 subjects with MS and 26,703 healthy controls of European ancestry. Inverse-variance weighted (IVW) MR was used as the primary method and multiple sensitivity analyses were employed in this study. Results: Genetically predicted circulating levels of FGF23 were associated with risk of MS. The odds ratio (OR) of IVW was 0.63 (95% confidence interval [CI], 0.49-0.82; p < 0.001) per one standard deviation increase in circulating FGF23 levels. Weighted median estimators also suggested FGF23 associated with lower MS risk (OR = 0.67; 95% CI, 0.51-0.87; p = 0.003). While MR-Egger approach provided no evidence of horizontal pleiotropy (intercept = -0.003, p = 0.95). Results of IVW methods provided no evidence for causal roles of GDF1, IGF1, IGFBP3 and VEGF on MS risks, and additional sensitivity analyses confirmed the robustness of these null findings. Conclusion: Our results implied a causal relationship between FGF23 and the risk of MS. Further studies are warranted to confirm FGF23 as a genetically valid target for MS.


Subject(s)
Fibroblast Growth Factor-23/physiology , Multiple Sclerosis/etiology , Adult , Aged , Female , Genome-Wide Association Study , Growth Differentiation Factor 15/physiology , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/physiology , Male , Mendelian Randomization Analysis , Middle Aged , Vascular Endothelial Growth Factor A/physiology
2.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502376

ABSTRACT

Inflammation induces a wide response of the neuroendocrine system, which leads to modifications in all the endocrine axes. The hypothalamic-growth hormone (GH)-insulin-like growth factor-1 (IGF-1) axis is deeply affected by inflammation, its response being characterized by GH resistance and a decrease in circulating levels of IGF-1. The endocrine and metabolic responses to inflammation allow the organism to survive. However, in chronic inflammatory conditions, the inhibition of the hypothalamic-GH-IGF-1 axis contributes to the catabolic process, with skeletal muscle atrophy and cachexia. Here, we review the changes in pituitary GH secretion, IGF-1, and IGF-1 binding protein-3 (IGFBP-3), as well as the mechanism that mediated those responses. The contribution of GH and IGF-1 to muscle wasting during inflammation has also been analyzed.


Subject(s)
Cachexia/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Cachexia/physiopathology , Growth Hormone/metabolism , Human Growth Hormone/metabolism , Humans , Hypothalamus/metabolism , Inflammation/physiopathology , Insulin/metabolism , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology
3.
Life Sci ; 265: 118758, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33188835

ABSTRACT

AIMS: Insulin-like growth factor (IGF) signaling has been documented in several human malignancies and is thought to contribute to cellular differentiation and migration, as well as malignant progression. A major binding molecule of IGF, IGF-binding protein 3 (IGFBP-3), regulates multiple IGF effects. Here, we focused on the effect of IGFBP-3 in the motility of osteosarcoma cells and examined signaling regulation. MATERIALS AND METHODS: Using a human osteosarcoma tissue array, immunohistochemical staining determined levels of IGFBP-3 expression in osteosarcoma tissue and in normal tissue. The wound healing migration assay, Transwell migration assay, luciferase reporter assay, immunofluorescence staining, Western blot and real-time quantitative PCR were performed to examine whether IGFBP-3 facilitates VCAM-1-dependent migration of osteosarcoma cells. KEY FINDINGS: In this study, we found significantly higher IGFBP-3 levels in osteosarcoma tissue compared with normal healthy tissue. IGFBP-3 treatment of two human osteosarcoma cell lines promoted cell migration and upregulated levels of VCAM-1 expression via PI3K/Akt and AP-1 signaling. SIGNIFICANCE: IGFBP-3 appears to be a novel therapeutic target in metastatic osteosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Cell Movement , Insulin-Like Growth Factor Binding Protein 3/metabolism , Osteosarcoma/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Up-Regulation
4.
J Cell Mol Med ; 24(18): 10525-10541, 2020 09.
Article in English | MEDLINE | ID: mdl-32798252

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of the cancer-related death in the world. Human amniotic mesenchymal stem cells (hAMSCs) have been characterized with a pluripotency, low immunogenicity and no tumorigenicity. Especially, the immunosuppressive and anti-inflammatory effects of hAMSCs make them suitable for treating HCC. Here, we reported that hAMSCs administrated by intravenous injection significantly inhibited HCC through suppressing cell proliferation and inducing cell apoptosis in tumour-bearing mice with Hepg2 cells. Cell tracking experiments with GFP-labelled hAMSCs showed that the stem cells possessed the ability of migrating to the tumorigenic sites for suppressing tumour growth. Importantly, both hAMSCs and the conditional media (hAMSC-CM) have the similar antitumour effects in vitro, suggesting that hAMSCs-derived cytokines might be involved in their antitumour effects. Antibody array assay showed that hAMSCs highly expressed dickkopf-3 (DKK-3), dickkopf-1 (DKK-1) and insulin-like growth factor-binding protein 3 (IGFBP-3). Furthermore, the antitumour effects of hAMSCs were further confirmed by applications of the antibodies or the specific siRNAs of DKK-3, DKK-1 and IGFBP-3 in vitro. Mechanically, hAMSCs-derived DKK-3, DKK-1 and IGFBP-3 markedly inhibited cell proliferation and promoted apoptosis of Hepg2 cells through suppressing the Wnt/ß-catenin signalling pathway and IGF-1R-mediated PI3K/AKT signalling pathway, respectively. Taken together, our study demonstrated that hAMSCs possess significant antitumour effects in vivo and in vitro and might provide a novel strategy for HCC treatment clinically.


Subject(s)
Amnion/cytology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Mesenchymal Stem Cell Transplantation , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Adipogenesis , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Female , Genes, Reporter , Hep G2 Cells/transplantation , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/physiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Osteogenesis , Paracrine Communication , Pregnancy , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
5.
Mol Biol Rep ; 47(4): 2561-2572, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32133604

ABSTRACT

PURPOSE: We have previously identified insulin-like growth factor 2 (IGF2) and insulin-like growth factor 1 receptor (IGF1R) as essential proteins for tip cell maintenance and sprouting angiogenesis. In this study, we aim to identify other IGF family members involved in endothelial sprouting angiogenesis. METHODS: Effects on sprouting were analyzed in human umbilical vein endothelial cells (HUVECs) using the spheroid-based sprouting model, and were quantified as mean number of sprouts per spheroid and average sprout length. RNA silencing technology was used to knockdown gene expression. Recombinant forms of the ligands (IGF1 and IGF2, insulin) and the IGF-binding proteins (IGFBP) 3 and 4 were used to induce excess effects. Effects on the tip cell phenotype were analyzed by measuring the fraction of CD34+ tip cells using flow cytometry and immunohistochemistry in a 3D angiogenesis model. Experiments were performed in the presence and absence of serum. RESULTS: Knockdown of IGF2 inhibited sprouting in HUVECs, in particular when cultured in the absence of serum, suggesting that components in serum influence the signaling of IGF2 in angiogenesis in vitro. We then determined the effects of IGFBP3 and IGFBP4, which are both present in serum, on IGF2-IGF1R signaling in sprouting angiogenesis in the absence of serum: knockdown of IGFBP3 significantly reduced sprouting angiogenesis, whereas knockdown of IGFBP4 resulted in increased sprouting angiogenesis in both flow cytometry analysis and immunohistochemical analysis of the 3D angiogenesis model. Other IGF family members except INSR did not affect IGF2-IGF1R signaling. CONCLUSIONS: Serum components and IGF binding proteins regulate IGF2 effects on sprouting angiogenesis. Whereas IGFBP3 acts as co-factor for IGF2-IGF1R binding, IGFBP4 inhibits IGF2 signaling.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor Binding Protein 4/physiology , Insulin-Like Growth Factor I , Insulin-Like Growth Factor II , Morphogenesis , Neovascularization, Pathologic/metabolism , Organoids/metabolism , Receptor, IGF Type 1 , Receptor, IGF Type 2 , Signal Transduction
6.
Sci Rep ; 10(1): 5083, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32193421

ABSTRACT

Insulin-like growth factor binding protein-3 (IGFBP-3) belongs to a family of six IGF binding proteins. We previously found that IGFBP-3 exerts its cytotoxic effects on A549 (p53 wild-type) cell survival through a mechanism that depends on hyaluronan-CD44 interactions. To shed light on the mechanism employed, we used CD44-negative normal human lung cells (HFL1), A549, and H1299 (p53-null) lung cancer cells. A synthetic IGFBP-3 peptide (215-KKGFYKKKQCRPSKGRKR-232) but not the mutant (K228AR230A), was able to bind hyaluronan more efficiently than the analogous sequences from the other IGFBPs. In a manner comparable to that of the IGFBP-3 protein, the peptide blocked hyaluronan-CD44 signaling, and more effectively inhibited viability of A549 cells than viability of either H1299 or HFL1 cell lines. Treatment with the IGFBP-3 protein or its peptide resulted in increased acetylcholinesterase concentration and activity in the A549 cell media but not in the media of either HFL1 or H1299, an effect that correlated with increased apoptosis and decreased cell viability. These effects were diminished upon the same treatment of A549 cells transfected with either p53 siRNA or acetylcholinesterase siRNA. Taken together, our results show that IGFBP-3 or its peptide blocks hyaluronan-CD44 signaling via a mechanism that depends on both p53 and acetylcholinesterase.


Subject(s)
Acetylcholinesterase/metabolism , Apoptosis/genetics , Culture Media/metabolism , Hyaluronan Receptors/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor Binding Protein 3/physiology , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , A549 Cells , Apoptosis/drug effects , Cell Survival/drug effects , Humans , Hyaluronic Acid/metabolism , Signal Transduction/drug effects
7.
Invest Ophthalmol Vis Sci ; 61(2): 46, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32106295

ABSTRACT

Purpose: The goal of this study was to determine the role of insulin-like growth factor-binding protein-3 (IGFBP-3) in the pathogenesis of herpes stromal keratitis (HSK). Methods: In an unbiased approach, a membrane-based protein array was carried out to determine the level of expression of pro- and anti-angiogenic molecules in uninfected and HSV-1 infected corneas. Quantitative RT-PCR and ELISA assays were performed to measure the amounts of IGFBP-3 at mRNA and protein levels. Confocal microscopy documented the localization of IGFBP-3 in uninfected and infected corneal tissue. Flow cytometry assay showed the frequency of immune cell types in infected corneas from C57BL/6J (B6) and IGFBP-3 knockout (IGFBP-3-/-) mice. Slit-lamp microscopy was used to quantitate the development of opacity and neovascularization in infected corneas from both groups of mice. Results: Quantitation of protein array dot blot showed an increased level of IGFBP-3 protein in HSV-1 infected than uninfected corneas and was confirmed with ELISA and quantitative RT-PCR assays. Cytosolic and nuclear localization of IGFBP-3 were detected in the cells of corneal epithelium, whereas scattered IGFBP-3 staining was evident in the stroma of HSK developing corneas. Increased opacity and hemangiogenesis were noted in the corneas of IGFBP-3-/- than B6 mice during the clinical period of HSK. Furthermore, an increased number of leukocytes comprising of neutrophils and CD4 T cells were found in HSK developing corneas of IGFBP-3-/- than B6 mice. Conclusions: Our data showed that lack of IGFBP-3 exacerbates HSK, suggesting the protective effect of IGFBP-3 protein in regulating the severity of HSK.


Subject(s)
Cornea/metabolism , Insulin-Like Growth Factor Binding Protein 3/physiology , Keratitis, Herpetic/metabolism , Animals , Corneal Neovascularization/pathology , Corneal Opacity/pathology , Corneal Stroma/metabolism , Herpesvirus 1, Human , Keratitis, Herpetic/pathology , Keratitis, Herpetic/virology , Mice , Mice, Inbred C57BL
8.
Atherosclerosis ; 296: 59-65, 2020 03.
Article in English | MEDLINE | ID: mdl-32065979

ABSTRACT

BACKGROUND AND AIMS: The insulin-like growth factor (IGF)-1 signalling pathway has been implicated in the pathogenesis of atherosclerosis; however, the mechanism underlying its role in stroke remains unexplained. Herein, we aimed to explore the effects of genetic polymorphisms in the IGF1 pathway on stroke in the Chinese Han population. METHODS: Twenty-six single-nucleotide polymorphisms (SNPs) in IGF1 pathway genes were genotyped in a case-control study consisting of 2070 stroke cases and 2243 controls. Main genetic effects and gene-gene interactive effects of the IGF1 pathway were evaluated. Weighted genetic risk scores (wGRS) were computed, and the associations between wGRS and gene expression were analysed. RESULTS: The variants at GHRH rs6032470 were significantly associated with high risk of hemorrhagic stroke (HS), and the adjusted OR (95%CI) was 1.368 (1.136-1.647). Significant additive interaction between rs6032470 and gender was detected for HS and ischemic stroke (IS). The association of rs6032470 and stroke was stronger in males than in females. Additionally, a significant gene-gene interaction of rs6032470-rs1874479 (IGFBP1) in relation to HS risk was identified (p < 0.05). IGF1 mRNA expression was significantly upregulated in IS, while it was linearly downregulated across rs6214 genotypes. In addition, IGFBP3 transcript variant 2 mRNA level was negatively correlated with wGRS (r = -0.285, p = 0.005). CONCLUSIONS: Our findings indicated that the IGF1 signalling pathway genes potentiated the risk of stroke through both main effects and gene-gene interactions. The genetic effect of GHRH rs6032470 on stroke was gender dependent. The wGRS of IGF1 pathway genes may be an independent predictor of stroke risk.


Subject(s)
Growth Hormone-Releasing Hormone/genetics , Insulin-Like Growth Factor I/genetics , Polymorphism, Single Nucleotide , Signal Transduction/genetics , Stroke/genetics , Adult , Age Distribution , Aged , Case-Control Studies , China/epidemiology , Diabetes Mellitus/epidemiology , Dyslipidemias/epidemiology , Epistasis, Genetic , Ethnicity/genetics , Female , Genetic Predisposition to Disease , Growth Hormone-Releasing Hormone/physiology , Humans , Hypertension/epidemiology , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/physiology , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Male , Middle Aged , RNA, Messenger/biosynthesis , Stroke/epidemiology , Transcription, Genetic
9.
Oncotarget ; 7(34): 55491-55505, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27448965

ABSTRACT

Obesity is associated epidemiologically with poor breast cancer prognosis, but the mechanisms remain unclear. Since IGF binding protein-3 (IGFBP-3) influences both breast cancer growth and adipocyte maturation, it may impact on how obesity promotes breast oncogenesis. This study investigated the role of endogenous IGFBP-3 on the development of obesity and subsequently on breast tumor growth. Wild-type (WT) C57BL/6 or IGFBP-3-null (BP3KO) mice were fed a high-fat diet (HFD) or control chow-diet for 15 weeks before orthotopic injection with syngeneic EO771 murine breast cancer cells. When the largest tumor reached 1000 mm3, tissues and tumors were excised for analysis. Compared to WT, BP3KO mice showed significantly reduced weight gain and mammary fat pad mass (contralateral to tumor) in response to HFD, despite similar food intake. EO771 tumor weight and volume were increased by HFD and decreased by BP3KO. Despite differences in tumor size, tumors in BP3KO mice showed no differences from WT in the number of mitotically active (Ki67+) and apoptotic (cleaved caspase-3+) cells, but had greater infiltration of CD3+ T-cells. These data suggest that endogenous (circulating and/or stromal) IGFBP-3 is stimulatory to adipose tissue expansion and enhances mammary tumor growth in immune-competent mice, potentially by suppressing T-cell infiltration into tumors.


Subject(s)
Breast Neoplasms/etiology , Insulin-Like Growth Factor Binding Protein 3/physiology , Obesity/etiology , Adipose Tissue/metabolism , Animals , Apoptosis , Breast Neoplasms/blood supply , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Movement , Diet, High-Fat , Disease Progression , Female , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Weight Gain
10.
Clin Epigenetics ; 8: 11, 2016.
Article in English | MEDLINE | ID: mdl-26823688

ABSTRACT

BACKGROUND: Insulin-like growth factors 1 and 2 (IGF1 and IGF2) and their binding proteins (IGFBPs) are expressed in the placenta and known to regulate fetal growth. DNA methylation is an epigenetic mechanism which involves addition of methyl group to a cytosine base in the DNA forming a methylated cytosine-phosphate-guanine (CpG) dinucleotide which is known to silence gene expression. This silences gene expression, potentially altering the expression of IGFs and their binding proteins. This study investigates the relationship between DNA methylation of components of the IGF axis in the placenta and disorders in fetal growth. Placental samples were obtained from cord insertions immediately after delivery from appropriate, small (defined as birthweight <10th percentile for the gestation [SGA]) and macrosomic (defined as birthweight > the 90th percentile for the gestation [LGA]) neonates. Placental DNA methylation, mRNA expression and protein levels of components of the IGF axis were determined by pyrosequencing, rtPCR and Western blotting. RESULTS: In the placenta from small for gestational age (SGA) neonates (n = 16), mRNA and protein levels of IGF1 were lower and of IGFBPs (1, 2, 3, 4 and 7) were higher (p < 0.05) compared to appropriately grown neonates (n = 37). In contrast, in the placenta from large for gestational age (LGA) neonates (n = 20), mRNA and protein levels of IGF1 was not different and those of IGFBPs (1, 2, 3 and 4) were lower (p < 0.05) compared to appropriately grown neonates. Compared to appropriately grown neonates, CpG methylation of the promoter regions of IGF1 was higher in SGA neonates. The CpG methylation of the promoter regions of IGFBP1, IGFBP2, IGFBP3, IGFBP4 and IGFBP7 was lower in the placenta from SGA neonates as compared to appropriately grown neonates, but was unchanged in the placenta from LGA neonates. CONCLUSIONS: Our results suggest that changes in CpG methylation contribute to the changes in gene expression of components of the IGF axis in fetal growth disorders. Differential methylation of the IGF1 gene and its binding proteins is likely to play a role in the pathogenesis of SGA neonates.


Subject(s)
DNA Methylation , Fetal Growth Retardation/etiology , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor I/genetics , Adult , Case-Control Studies , Female , Fetal Growth Retardation/genetics , Humans , Infant, Newborn , Infant, Small for Gestational Age , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/physiology , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/physiology , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/physiology , Insulin-Like Growth Factor Binding Proteins/physiology , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/physiology , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic/genetics
11.
Aging (Albany NY) ; 7(11): 956-63, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26568155

ABSTRACT

Reduced insulin/insulin-like growth factor 1 (IGF-1) signaling has been associated with longevity in various model organisms. However, the role of insulin/IGF-1 signaling in human survival remains controversial. The aim of this study was to test whether circulating IGF-1 axis parameters associate with old age survival and functional status in nonagenarians from the Leiden Longevity Study. This study examined 858 Dutch nonagenarian (males≥89 years; females≥91 years) siblings from 409 families, without selection on health or demographic characteristics. Nonagenarians were divided over sex-specific strata according to their levels of IGF-1, IGF binding protein 3 and IGF-1/IGFBP3 molar ratio. We found that lower IGF-1/IGFBP3 ratios were associated with improved survival: nonagenarians in the quartile of the lowest ratio had a lower estimated hazard ratio (95% confidence interval) of 0.73 (0.59 - 0.91) compared to the quartile with the highest ratio (ptrend=0.002). Functional status was assessed by (Instrumental) Activities of Daily Living ((I)ADL) scales. Compared to those in the quartile with the highest IGF-1/IGFBP3 ratio, nonagenarians in the lowest quartile had higher scores for ADL (ptrend=0.001) and IADL (ptrend=0.003). These findings suggest that IGF-1 axis parameters are associated with increased old age survival and better functional status in nonagenarians from the Leiden Longevity Study.


Subject(s)
Insulin-Like Growth Factor I/physiology , Longevity , Aged , Aged, 80 and over , Female , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Signal Transduction/physiology
12.
Mater Sci Eng C Mater Biol Appl ; 55: 490-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26117781

ABSTRACT

Insulin like growth factor binding protein-3 (IGFBP-3) in bone cells and its utilization in dental implants have not been well studied. The aim of this study was to determine the osteogenic efficacy of chitosan gold nanoparticles (Ch-GNPs) conjugated with IGFBP-3 coated titanium (Ti) implants. Ch-GNPs were conjugated with IGFBP-3 plasmid DNA through a coacervation process. Conjugation was cast over Ti surfaces, and cells were seeded on coated surfaces. For in vitro analysis the expression of different proteins was analyzed by immunoblotting. For in vivo analysis, Ch-GNP/IGFBP-3 coated implants were installed in rat mandibles. Four weeks post-implantation, mandibles were examined by microcomputed tomography (µCT), immunohistochemistry, hematoxylin & eosin and tartrate resistance acid phosphatase staining. In vitro overexpressed Ch-GNP/IGFBP-3 coated Ti surfaces was associated with activation of extracellular signal related kinase (ERK), inhibition of the stress activated protein c-Jun N-terminal kinase (JNK) and enhanced bone morphogenetic protein (BMP)-2 and 7 compared to control. Further, in vivo, Ch-GNP/IGFBP-3 coated implants were associated with inhibition of implant induced osteoclastogenesis molecules, receptor activator of nuclear factor kappa-B ligand (RANKL) and enhanced expression of osteogenic molecules including BMP2/7 and osteopontin (OPN). The µCT analysis demonstrated that IGFBP-3 increased the volume of newly formed bone surrounding the implants compared to control (n=5; p<0.05). These results support the view that IGFBP-3 overexpression diminishes osteoclastogenesis and enhances osteogenesis of Ti implants, and can serve as a potent molecule for the development of good implantation.


Subject(s)
Dental Implants , Insulin-Like Growth Factor Binding Protein 3/physiology , Mandible/surgery , Osteogenesis , 3T3 Cells , Animals , Male , Mice , Rats , Rats, Sprague-Dawley
13.
Diabetes ; 64(7): 2289-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26106186

ABSTRACT

Cellular senescence is a fundamental aging mechanism that has been implicated in many age-related diseases and is a significant cause of tissue dysfunction. Accumulation of senescent cells occurs during aging and is also seen in the context of obesity and diabetes. Senescent cells may play a role in type 2 diabetes pathogenesis through direct impact on pancreatic ß-cell function, senescence-associated secretory phenotype (SASP)-mediated tissue damage, and involvement in adipose tissue dysfunction. In turn, metabolic and signaling changes seen in diabetes, such as high circulating glucose, altered lipid metabolism, and growth hormone axis perturbations, can promote senescent cell formation. Thus, senescent cells might be part of a pathogenic loop in diabetes, as both a cause and consequence of metabolic changes and tissue damage. Therapeutic targeting of a basic aging mechanism such as cellular senescence may have a large impact on disease pathogenesis and could be more effective in preventing the progression of diabetes complications than currently available therapies that have limited impact on already existing tissue damage. Therefore, senescent cells and the SASP represent significant opportunities for advancement in the prevention and treatment of type 2 diabetes and its complications.


Subject(s)
Cellular Senescence , Diabetes Mellitus, Type 2/etiology , Adipose Tissue/physiology , Animals , Cellular Microenvironment , Ceramides/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/therapy , Humans , Insulin Resistance , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Insulin-Secreting Cells/pathology
14.
Pediatr Endocrinol Rev ; 13(2): 521-30, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26841640

ABSTRACT

The insulin-like growth factor (IGF) system is essential for normal growth and development, and its perturbation is implicated in a number of diseases. IGF activity is finely regulated by a family of six high-affinity IGF binding proteins (IGFBPs). 1GFBPs usually inhibit IGF actions but may enhance them under certain conditions. Additionally, IGFBPs bind non-IGF ligands in the extracellular space, cell membrane, cytoplasm and nucleus, thereby modulating cell proliferation, survival and migration in an IGF-independent manner. IGFBP activity is regulated by transcriptional mechanisms as well as by post-translational modifications and proteolysis. Understanding the balance between the various actions of IGFBPs in vivo may lead to novel insights into disease processes and possible IGFBP-based therapeutics.


Subject(s)
Insulin-Like Growth Factor Binding Proteins , Animals , Cell Physiological Phenomena/physiology , Humans , Insulin-Like Growth Factor Binding Protein 1/chemistry , Insulin-Like Growth Factor Binding Protein 1/physiology , Insulin-Like Growth Factor Binding Protein 2/chemistry , Insulin-Like Growth Factor Binding Protein 2/physiology , Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor Binding Protein 4/chemistry , Insulin-Like Growth Factor Binding Protein 4/physiology , Insulin-Like Growth Factor Binding Protein 5/chemistry , Insulin-Like Growth Factor Binding Protein 5/physiology , Insulin-Like Growth Factor Binding Protein 6/chemistry , Insulin-Like Growth Factor Binding Protein 6/physiology , Insulin-Like Growth Factor Binding Proteins/chemistry , Insulin-Like Growth Factor Binding Proteins/physiology , Molecular Structure , Somatomedins/physiology
15.
Ann Rheum Dis ; 74(10): 1861-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24928840

ABSTRACT

OBJECTIVES: To identify genetic associations with hip osteoarthritis (HOA), we performed a meta-analysis of genome-wide association studies (GWAS) of HOA. METHODS: The GWAS meta-analysis included approximately 2.5 million imputed HapMap single nucleotide polymorphisms (SNPs). HOA cases and controls defined radiographically and by total hip replacement were selected from the Osteoporotic Fractures in Men (MrOS) Study and the Study of Osteoporotic Fractures (SOF) (654 cases and 4697 controls, combined). Replication of genome-wide significant SNP associations (p ≤5×10(-8)) was examined in five studies (3243 cases and 6891 controls, combined). Functional studies were performed using in vitro models of chondrogenesis and osteogenesis. RESULTS: The A allele of rs788748, located 65 kb upstream of the IGFBP3 gene, was associated with lower HOA odds at the genome-wide significance level in the discovery stage (OR 0.71, p=2×10(-8)). The association replicated in five studies (OR 0.92, p=0.020), but the joint analysis of discovery and replication results was not genome-wide significant (p=1×10(-6)). In separate study populations, the rs788748 A allele was also associated with lower circulating IGFBP3 protein levels (p=4×10(-13)), suggesting that this SNP or a variant in linkage disequilibrium could be an IGFBP3 regulatory variant. Results from functional studies were consistent with association results. Chondrocyte hypertrophy, a deleterious event in OA pathogenesis, was largely prevented upon IGFBP3 knockdown in chondrocytes. Furthermore, IGFBP3 overexpression induced cartilage catabolism and osteogenic differentiation. CONCLUSIONS: Results from GWAS and functional studies provided suggestive links between IGFBP3 and HOA.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/genetics , Osteoarthritis, Hip/genetics , Aged , Aged, 80 and over , Case-Control Studies , Chondrogenesis/genetics , Chondrogenesis/physiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Middle Aged , Osteoarthritis, Hip/physiopathology , Osteogenesis/genetics , Osteogenesis/physiology , Polymorphism, Single Nucleotide
16.
Discov Med ; 18(98): 105-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25227751

ABSTRACT

The earliest and most significant change in diabetic retinopathy (DR) is blood-retinal barrier (BRB) dysfunction, followed by two main pathologies that may cause severe visual impairment: Diabetic Macular Edema (DME) and Proliferative Diabetic Retinopathy (PDR). The pathological hallmarks of BRB dysfunction include loss of tight junction integrity, VEGF- and AGE-induced damage, oxidative stress, and inflammatory changes. Recently, several BRB protective factors have been reported. Our aim is to give a review of those protective factors and discuss new potential therapeutic targets for DR.


Subject(s)
Blood-Retinal Barrier/physiopathology , Diabetic Retinopathy/prevention & control , Diabetic Retinopathy/physiopathology , Animals , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/pathology , Diabetic Retinopathy/etiology , Erythropoietin/pharmacology , Fenofibrate/analogs & derivatives , Fenofibrate/pharmacology , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Macular Edema/complications , Macular Edema/pathology , Macular Edema/physiopathology , Protective Factors , Pyrazines/pharmacology , Sitagliptin Phosphate , Triazoles/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
17.
Growth Horm IGF Res ; 24(5): 164-73, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24953254

ABSTRACT

One of the hallmarks in the advancement of cancer cells is an ability to overcome and acquire resistance to adverse conditions. There has been a large amount of cancer research on IGFBP-3 as a pro-apoptotic molecule in vitro. These pro-apoptotic properties, however, do not correlate with several studies linking high IGFBP-3 levels in breast cancer tissue to rapid growth and poor prognosis. Evidence is emerging that IGFBP-3 also exhibits pro-survival and growth-promoting properties in vitro. How IGFBP-3 pivots cell fate to either death or survival, it seems, comes down to a complex interplay between cells' microenvironments and the presence of cellular IGFBP-3 binding partners and growth factor receptors. The cytoprotective actions of IGFBP-3 are not restricted to cancer but are also observed in other disease states, such as retinopathy and brain ischaemia. Here we review the literature on this paradoxical nature of IGFBP-3, its pro-apoptotic and growth-inhibitory actions versus its cytoprotective and growth-potentiating properties, and discuss the implications of targeting IGFBP-3 for treatment of disease.


Subject(s)
Disease/genetics , Genes, Switch , Insulin-Like Growth Factor Binding Protein 3/physiology , Neoplasms/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Endoplasmic Reticulum/metabolism , Humans
18.
J Neurosurg ; 121(2): 374-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24878287

ABSTRACT

OBJECT: Insulin-like growth factor binding proteins (IGFBPs) have been implicated in the pathogenesis of glioma. In a previous study the authors demonstrated that IGFBP-3 is a novel glioblastoma biomarker associated with poor survival. Since signal transducer and activator of transcription 1 (STAT-1) has been shown to be regulated by IGFBP-3 during chondrogenesis and is a prosurvival and radioresistant molecule in different tumors, the aim in the present study was to explore the functional significance of IGFBP-3 in malignant glioma cells, to determine if STAT-1 is indeed regulated by IGFBP-3, and to study the potential of STAT-1 as a biomarker in glioblastoma. METHODS: The functional significance of IGFBP-3 was investigated using the short hairpin (sh)RNA gene knockdown approach on U251MG cells. STAT-1 regulation by IGFBP-3 was tested on U251MG and U87MG cells by shRNA gene knockdown and exogenous treatment with recombinant IGFBP-3 protein. Subsequently, the expression of STAT-1 was analyzed with real-time reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC) in glioblastoma and control brain tissues. Survival analyses were done on a uniformly treated prospective cohort of adults with newly diagnosed glioblastoma (136 patients) using Kaplan-Meier and Cox regression models. RESULTS: IGFBP-3 knockdown significantly impaired proliferation, motility, migration, and invasive capacity of U251MG cells in vitro (p < 0.005). Exogenous overexpression of IGFBP-3 in U251MG and U87MG cells demonstrated STAT-1 regulation. The mean transcript levels (by real-time RT-PCR) and the mean labeling index of STAT-1 (by IHC) were significantly higher in glioblastoma than in control brain tissues (p = 0.0239 and p < 0.001, respectively). Multivariate survival analysis revealed that STAT-1 protein expression (HR 1.015, p = 0.033, 95% CI 1.001-1.029) along with patient age (HR 1.025, p = 0.005, 95% CI 1.008-1.042) were significant predictors of shorter survival in patients with glioblastoma. CONCLUSIONS: IGFBP-3 influences tumor cell proliferation, migration, and invasion and regulates STAT-1 expression in malignant glioma cells. STAT-1 is overexpressed in human glioblastoma tissues and emerges as a novel prognostic biomarker.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioma/genetics , Glioma/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , STAT1 Transcription Factor/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Glioblastoma/mortality , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Predictive Value of Tests , STAT1 Transcription Factor/biosynthesis , Survival Analysis
19.
Hypertension ; 63(5): 1070-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24566078

ABSTRACT

Conflicting evidence exists on the relationship between blood pressure (BP) and insulin-like growth factor-1 (IGF-1). We reviewed available articles and pooled extrapolated regression coefficients for the association between BP and total IGF-1 as reported in the literature and included additional data from 912 individuals from the general population. We identified 20 studies including 11 704 subjects. We also measured total IGF-1, insulin-like binding protein-3, and BP in 912 black and white men and women from South Africa (aged 20-70 years). When plotting positive and negative weighed regression coefficients (29 data points) against IGF-1, we found a significant positive relationship (r=0.31; P<0.001; n=11 704) intercepting the 0 point at 191 ng/mL IGF-1, suggesting an inverse BP/IGF-1 relationship in low IGF-1 conditions, and a positive relationship in overtly high IGF-1 conditions. In conclusion, our findings suggest that the relationship between BP and IGF-1 is dependent on, or related to, IGF-1 concentrations, as an expression of direct or reverse causality. Low IGF-1 bioavailability (associated with aging and vascular deterioration), resistance to IGF-1, and the complex interplay between IGF-1 and other vasoactive hormones could mask the vasoprotective functions of IGF-1 in cross-sectional studies or could modify their functions in prospective studies.


Subject(s)
Blood Pressure/physiology , Insulin-Like Growth Factor I/physiology , Signal Transduction/physiology , Adult , Aged , Female , Humans , Hypertension/physiopathology , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Middle Aged , Phosphatidylinositol 3-Kinases/physiology
20.
Oncogene ; 33(1): 85-96, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-23178489

ABSTRACT

Following exposure to radiation and chemotherapeutic agents, the epidermal growth factor receptor (EGFR) can modulate the repair of DNA double-strand breaks (DSB) by forming protein complexes that include the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs). This is one of the key mechanism by which tumors become resistant to DNA-damaging therapies. Our previous studies have shown that insulin-like growth factor binding protein-3 (IGFBP-3) is a substrate for DNA-PKcs, and can transactivate EGFR. We therefore questioned whether IGFBP-3 might interact with the EGFR-DNA-PK complex that regulates the DNA damage response. The aim of this study was to delineate the role of IGFBP-3 in the response of breast cancer cells to DSB-inducing chemotherapeutic agents. In the estrogen receptor-negative breast cancer cell lines MDA-MB-468 and Hs578T, which express IGFBP-3 highly, nuclear localization of EGFR and IGFBP-3 was enhanced by treatment with cytotoxic drugs etoposide or doxorubicin and reduced by the EGFR kinase inhibitor gefitinib. Enhanced association among IGFBP-3, EGFR and DNA-PKcs, following the exposure to DNA-damaging drugs was supported by both co-immunoprecipitation analysis and direct visualization by proximity ligation assay. The activation of DNA-PKcs at Ser2056, DNA repair as measured by a nonhomologous end-joining assay, and the increase in EGFR and DNA-PKcs interaction induced by DNA-damaging agents, were all decreased by IGFBP-3 silencing, suggesting that IGFBP-3 has an obligatory role in the DNA repair response to DNA-damaging therapy. In conclusion, IGFBP-3 co-translocation to the nucleus of breast cancer cells and its formation of a complex with DNA-PKcs and EGFR in response to DNA damage shows its potential involvement in the regulation of DNA repair. This suggests the possibility of a therapeutic approach for sensitizing breast cancer to chemo- or radiotherapy by targeting the DNA repair function of IGFBP-3.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , Etoposide/pharmacology , Insulin-Like Growth Factor Binding Protein 3/physiology , Active Transport, Cell Nucleus , Breast Neoplasms , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , DNA Repair , DNA-Activated Protein Kinase/metabolism , Doxorubicin/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Membrane Microdomains/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Quinazolines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...