Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 247
1.
Eur J Hum Genet ; 30(10): 1159-1166, 2022 10.
Article En | MEDLINE | ID: mdl-35688891

Insulin-like growth factor binding protein 4 (IGFBP4) is involved in adipogenesis, and IGFBP4 null mice have decreased body fat through decreased PPAR-γ expression. In the current study, we assessed whether variation in the IGFBP4 coding region influences body mass index (BMI) in American Indians who are disproportionately affected by obesity. Whole exome sequence data from a population-based sample of 6779 American Indians with longitudinal measures of BMI were used to identify variation in IGFBP4 that associated with BMI. A novel variant that predicts a p.Ser76Thr in IGFBP4 (Thr-allele frequency = 0.02) was identified which associated with the maximum BMI measured during adulthood (BMI 39.8 kg/m2 for Thr-allele homozygotes combined with heterozygotes vs. 36.2 kg/m2 for Ser-allele homozygotes, ß = 6.7% per Thr-allele, p = 8.0 × 10-5, adjusted for age, sex, birth-year and the first five genetic principal components) and the maximum age- and sex-adjusted BMI z-score measured during childhood/adolescence (z-score 0.70 SD for Thr-allele heterozygotes vs. 0.32 SD for Ser-allele homozygotes, ß = 0.37 SD per Thr-allele, p = 8.8 × 10-6). In vitro functional studies showed that IGFBP4 with the Thr-allele (BMI-increasing) had a 55% decrease (p = 0.0007) in FOXO-induced transcriptional activity, reflecting increased activation of the PI3K/AKT pathway mediated through increased IGF signaling. Over-expression and knock-down of IGFBP4 in OP9 cells during differentiation showed that IGFBP4 upregulates adipogenesis through PPARγ, CEBPα, AGPAT2 and SREBP1 expression. We propose that this American Indian specific variant in IGFBP4 affects obesity via an increase of IGF signaling.


Indians, North American , Insulin-Like Growth Factor Binding Protein 4 , Animals , Body Mass Index , Humans , Indians, North American/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Mice , Obesity/genetics , PPAR gamma/genetics , Phosphatidylinositol 3-Kinases/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-akt/genetics , American Indian or Alaska Native
2.
PLoS One ; 16(6): e0251732, 2021.
Article En | MEDLINE | ID: mdl-34143773

Prior studies demonstrated that deletion of the protein phosphatase Phlpp1 in Ctsk-Cre expressing cells enhances bone mass, characterized by diminished osteoclast activity and increased coupling to bone formation. Due to non-specific expression of Ctsk-Cre, the definitive mechanism for this observation was unclear. To further define the role of bone resorbing osteoclasts, we performed ovariectomy (Ovx) and Sham surgeries on Phlpp1 cKOCtsk and WT mice. Micro-CT analyses confirmed enhanced bone mass of Phlpp1 cKOCtsk Sham females. In contrast, Ovx induced bone loss in both groups, with no difference between Phlpp1 cKOCtsk and WT mice. Histomorphometry demonstrated that Ovx mice lacked differences in osteoclasts per bone surface, suggesting that estradiol (E2) is required for Phlpp1 deficiency to have an effect. We performed high throughput unbiased transcriptional profiling of Phlpp1 cKOCtsk osteoclasts and identified 290 differentially expressed genes. By cross-referencing these differentially expressed genes with all estrogen response element (ERE) containing genes, we identified IGFBP4 as potential estrogen-dependent target of Phlpp1. E2 induced PHLPP1 expression, but reduced IGFBP4 levels. Moreover, genetic deletion or chemical inhibition of Phlpp1 was correlated with IGFBP4 levels. We then assessed IGFBP4 expression by osteoclasts in vivo within intact 12-week-old females. Modest IGFBP4 immunohistochemical staining of TRAP+ osteoclasts within WT females was observed. In contrast, TRAP+ bone lining cells within intact Phlpp1 cKOCtsk females robustly expressed IGFBP4, but levels were diminished within TRAP+ bone lining cells following Ovx. These results demonstrate that effects of Phlpp1 conditional deficiency are lost following Ovx, potentially due to estrogen-dependent regulation of IGFBP4.


Bone Resorption/pathology , Cathepsin K/metabolism , Estrogens/pharmacology , Insulin-Like Growth Factor Binding Protein 4/metabolism , Osteoclasts/metabolism , Osteoporosis/pathology , Phosphoprotein Phosphatases/physiology , Animals , Bone Resorption/etiology , Bone Resorption/metabolism , Cathepsin K/genetics , Cell Differentiation , Female , Insulin-Like Growth Factor Binding Protein 4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/drug effects , Osteoporosis/etiology , Osteoporosis/metabolism , Ovariectomy
3.
Hum Mol Genet ; 30(3-4): 277-293, 2021 04 26.
Article En | MEDLINE | ID: mdl-33355648

Ankylosing spondylitis (AS) is a rheumatic disease with pathological osteogenesis that causes bony ankylosis and even deformity over time. Mesenchymal stem cells (MSCs) are multipotent stem cells that are the main source of osteoblasts. We previously demonstrated that enhanced osteogenic differentiation of MSCs from AS patients (ASMSCs) is related to pathological osteogenesis in AS. However, the more concrete mechanism needs further exploration. Super enhancers (SEs) are dense clusters of stitched enhancers that control cell identity determination and disease development. Single-nucleotide polymorphisms (SNPs) regulate the formation and interaction of SEs and denote genes accounting for AS susceptibility. Via integrative analysis of multiomic data, including histone 3 lysine 27 acetylation (H3K27ac), chromatin immunoprecipitation sequencing (ChIP-seq), SNPs and RNA sequencing (RNA-seq) data, we discovered a transcription network mediated by AS SNP-adjacent SEs (SASEs) in ASMSCs and identified key genes, such as Toll-like receptor 4 (TLR4), interleukin 18 receptor 1 (IL18R1), insulin-like growth factor binding protein 4 (IGFBP4), transportin 1 (TNPO1) and proprotein convertase subtilisin/kexin type 5 (PCSK5), which are pivotal in osteogenesis and AS pathogenesis. The SASE-regulated network modulates the enhanced osteogenic differentiation of ASMSCs by synergistically activating the PI3K-Akt, NF-kappaB and Hippo signaling pathways. Our results emphasize the crucial role of the SASE-regulated network in pathological osteogenesis in AS, and the preferential inhibition of ASMSC osteogenic differentiation by JQ1 indicates that SEs may be attractive targets in future treatment for new bone formation in AS.


Gene Regulatory Networks , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Signal Transduction , Spondylitis, Ankylosing/genetics , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation Sequencing , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Interleukin-18 Receptor alpha Subunit/genetics , Mesenchymal Stem Cells/physiology , Polymorphism, Single Nucleotide , Proprotein Convertase 5/genetics , Sequence Analysis, RNA , Spondylitis, Ankylosing/physiopathology , Toll-Like Receptor 4/genetics , beta Karyopherins/genetics
4.
Int J Biol Sci ; 16(13): 2271-2282, 2020.
Article En | MEDLINE | ID: mdl-32760196

Insulin-like growth factor binding protein 4-1 (IGFBP4-1), a new long noncoding RNA (lncRNA), has been reported to contribute to tumorigenesis and has been suggested to be a poor prognostic marker in human lung cancer. However, there still lacks basic studies that investigated the biological role of IGFBP4-1 in bladder urothelial carcinoma to date. In this study, we investigated the relationship between IGFBP4-1 expression and prognosis in patients with bladder cancer. Cell proliferation, cell cycle and cell apoptosis assays were performed to assess IGFBP4-1 function by up-regulating or down-regulating IGFBP4-1 in bladder cancer cells. A xenograft mice model was used to validate the in vitro results. Blockade of Janus kinase-signal transducer and activator of transcription pathway (JAK/STAT) was used to evaluate JAK/STAT signaling activity. The results showed that IGFBP4-1 was overexpressed in bladder cancer tissues compared with that in normal bladder tissues, and its expression level was positively correlated with poor prognosis in bladder cancer patients. Overexpression of IGFBP4-1 markedly promoted cell proliferation and cell cycle progression, and inhibited cell apoptosis, while knockdown of IGFBP4-1 notably suppressed the proliferation, promoted cell apoptosis, and induced cell cycle arrest at the G0/G1 phase. Mechanistically, we revealed that IGFBP4-1 promotes the activation of the JAK/STAT pathway in bladder cancer cells. Moreover, the JAK/STAT inhibitor dramatically blocked the tumor-promoting activity of IGFBP4-1. Tumor growth in vivo was also suppressed by knocking down of IGFBP4-1. In conclusion, IGFBP4-1 promoted bladder cancer progression by activating the JAK/STAT signaling pathway. These findings suggest that IGFBP4-1 exhibits an oncogenic role in the development of human bladder cancer.


Carcinoma/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Apoptosis , Carcinoma/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Databases, Genetic , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Janus Kinases/genetics , Male , Mice , Mice, Nude , Middle Aged , Neoplasms, Experimental , RNA, Long Noncoding , STAT Transcription Factors/genetics , Up-Regulation , Urinary Bladder Neoplasms/genetics
5.
J Cell Mol Med ; 24(16): 9466-9471, 2020 08.
Article En | MEDLINE | ID: mdl-32597006

Vascular endothelial growth factor (VEGF) is a well-known angiogenic factor, however its ability in promoting therapeutic angiogenesis following myocardial infarction (MI) is limited. Here, we aimed to investigate whether dual treatment with insulin-like growth factor binding protein-4 (IGFBP-4), an agent that protects against early oxidative damage, can be effective in enhancing the therapeutic effect of VEGF following MI. Combined treatment with IGFBP-4 enhanced VEGF-induced angiogenesis and prevented cell damage via enhancing the expression of a key angiogenic factor angiopoietin-1. Dual treatment with the two agents synergistically decreased cardiac fibrosis markers collagen-I and collagen-III following MI. Importantly, while the protective action of IGFBP-4 occurs at an early stage of ischemic injury, the action of VEGF occurs at a later stage, at the onset angiogenesis. Our findings demonstrate that VEGF treatment alone is often not enough to protect against oxidative stress and promote post-ischemic angiogenesis, whereas the combined treatment with IGFBP4 and VEGF can utilize the dual roles of these agents to effectively protect against ischemic and oxidative injury, and promote angiogenesis. These findings provide important insights into the roles of these agents in the clinical setting, and suggest new strategies in the treatment of ischemic heart disease.


Disease Models, Animal , Insulin-Like Growth Factor Binding Protein 4/metabolism , Myocardial Infarction/complications , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Insulin-Like Growth Factor Binding Protein 4/genetics , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/genetics
6.
Elife ; 92020 03 30.
Article En | MEDLINE | ID: mdl-32223893

Senescent cells secrete several molecules, collectively named senescence-associated secretory phenotype (SASP). In the SASP of cells that became senescent following several in vitro chemical and physical stress, we identified the IGFBP-4 protein that can be considered a general stress mediator. This factor appeared to play a key role in senescence-paracrine signaling. We provided evidences showing that genotoxic injury, such as low dose irradiation, may promote an IGFBP-4 release in bloodstream both in mice irradiated with 100 mGy X-ray and in human subjects that received Computer Tomography. Increased level of circulating IGFBP-4 may be responsible of pro-aging effect. We found a significant increase of senescent cells in the lungs, heart, and kidneys of mice that were intraperitoneally injected with IGFBP-4 twice a week for two months. We then analyzed how genotoxic stressors may promote the release of IGFBP-4 and the molecular pathways associated with the induction of senescence by this protein.


Aging , Cellular Senescence/genetics , DNA Damage , Insulin-Like Growth Factor Binding Protein 4/blood , Insulin-Like Growth Factor Binding Protein 4/genetics , Adolescent , Adult , Aged , Animals , Cell Proliferation , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype , Signal Transduction , Tomography, X-Ray Computed , Young Adult
7.
Sci Rep ; 9(1): 13231, 2019 09 13.
Article En | MEDLINE | ID: mdl-31519945

Pregnancy-associated plasma protein-A (PAPP-A) is a key regulator of insulin-like growth factor (IGF) bioactivity, by releasing the IGFs from their corresponding IGF-binding proteins (IGFBPs). The minor allele of the single nucleotide polymorphism (SNP), rs7020782 (serine < tyrosine), in PAPPA has previously been associated with recurrent pregnancy loss as well as with significant reduced levels of PAPP-A protein in human ovarian follicles. The aim of the present study was to reveal a possible functional effect of the rs7020782 SNP in PAPPA by comparing recombinant PAPP-A proteins from transfected human embryonic kidney 293 T cells. The proteolytic cleavage of IGFBP-4 was shown to be affected by the rs7020782 SNP in PAPPA, showing a significantly reduced cleavage rate for the serine variant compared to the tyrosine variant (p-value < 0.001). The serine variant also showed a trend towards reduced cleavage rates, that was not significant, towards IGFBP-2 and IGFBP-5 compared to the tyrosine variant. No differences were found when analysing cell surface binding, complex formation between PAPP-A and STC2 or proMBP, nor when analysing STC1 inhibition of PAPP-A-mediated IGFBP-4 cleavage. Regulation of IGF bioactivity in reproductive tissues is important and the rs7020782 SNP in PAPPA may disturb this regulation by altering the specific activity of PAPP-A.


Glycoproteins/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Polymorphism, Single Nucleotide , Pregnancy-Associated Plasma Protein-A/genetics , Female , Glycoproteins/genetics , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 5/genetics , Intercellular Signaling Peptides and Proteins/genetics , Pregnancy , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Binding , Proteolysis
8.
Exp Eye Res ; 185: 107650, 2019 08.
Article En | MEDLINE | ID: mdl-31075223

THE AIM OF THE STUDY: was to investigate the molecular genetic mechanisms of the influence of laser radiation with 577 nm wavelength in a microimpulse mode on the retina in the experimental conditions after the intravitreal injection of VEGF. MATERIALS AND METHODS: The study was performed on 4-5 week-old male mice of the line C57BL/6J. The animals were divided into 4 groups of 5 mice in each group, one eye was excremental, the contralateral eye remained intact. In the first group, intravitreal injection of PBS was performed; in the second group, intravitreal injection of 50 ng/ml of recombinant VEGF165 in 2 µL of phosphate-buffered saline (PBS) was performed; in the third and fourth groups, a day after the intravitreal injection of recombinant VEGF165, laser radiation with wavelength 577 nm was applied in the micropulse and continuous modes, respectively. Tissue samples (neuroepithelium, pigment epithelium) for the microarray transcription analysis in the animals from group 1 and 2 were taken 2 days after the injection of PBS and VEGF, in the animals from group 3 and 4 - a day after the retina was exposed to laser radiation. RESULTS AND CONCLUSION: Molecular genetic mechanisms of the influence of laser radiation with wavelength 577 nm in a microimpulse mode on the retina in experimental conditions were studied and the genes that significantly changed the level of expression (the genes that take part in the regulation of neoangiogenesis, structural cell functions, processes of cells proliferation, transcription, differentiation, transmembrane transport, signaling, synaptic transmission, etc.) were identified.


Eye Proteins/genetics , Gene Expression Regulation/physiology , Laser Therapy , Retina/radiation effects , Animals , GPI-Linked Proteins/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Intravitreal Injections , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Molecular Biology , Recombinant Proteins/administration & dosage , Retina/metabolism , Uridine Phosphorylase/genetics , Vascular Endothelial Growth Factor A/administration & dosage
9.
Cell Biochem Funct ; 36(6): 331-341, 2018 Aug.
Article En | MEDLINE | ID: mdl-30028031

Insulin-like growth factor binding protein 4 (IGFBP-4) was reported to trigger cellular senescence and reduce cell growth of bone marrow mesenchymal stem cells (BMSCs), but its contribution to neurogenic differentiation of BMSCs remains unknown. In the present study, BMSCs were isolated from the femur and tibia of young rats to investigate effects of IGFBP-4 on BMSC proliferation and growth of neurospheres derived from BMSCs. Bone marrow mesenchymal stem cell proliferation was assessed using CCK-8 after treatment with IGFBP-4 or blockers of IGF-IR and ß-catenin. Phosphorylation levels of Akt, Erk, and p38 in BMSCs were analysed by Western blotting. Bone marrow mesenchymal stem cells were induced into neural lineages in NeuroCult medium; the number and the size of BMSC-derived neurospheres were counted after treatment with IGFBP-4 or the blockers. It was shown that addition of IGFBP-4 inhibited BMSC proliferation and immunodepletion of IGFBP-4 increased the proliferation. The blockade of IGF-IR with AG1024 increased BMSC proliferation and reversed IGFBP-4-induced proliferation inhibition; however, blocking of ß-catenin with FH535 did not. p-Erk was significantly decreased in IGFBP-4-treated BMSCs. IGFBP-4 promoted the growth of neurospheres derived from BMSCs, as manifested by the increases in the number and the size of the derived neurospheres. Both AG1024 and FH535 inhibited the formation of NeuroCult-induced neurospheres, but FH535 significantly inhibited the growth of neurospheres in NeuroCult medium with EGF, bFGF, and IGFBP-4. The data suggested that IGFBP-4 inhibits BMSC proliferation through IGF-IR pathway and promotes growth of BMSC-derived neurospheres via stabilizing ß-catenin.


Cell Differentiation/drug effects , Cell Proliferation/drug effects , Insulin-Like Growth Factor Binding Protein 4/pharmacology , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sulfonamides/pharmacology , Tyrphostins/pharmacology , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Nucleic Acids Res ; 46(17): 8832-8847, 2018 09 28.
Article En | MEDLINE | ID: mdl-29992318

Genomic sequencing of hepatocellular carcinoma (HCC) uncovers a paucity of actionable mutations, underscoring the necessity to exploit epigenetic vulnerabilities for therapeutics. In HCC, EZH2-mediated H3K27me3 represents a major oncogenic chromatin modification, but how it modulates the therapeutic vulnerability of signaling pathways remains unknown. Here, we show EZH2 acts antagonistically to AKT signaling in maintaining H3K27 methylome through epigenetic silencing of IGFBP4. ChIP-seq revealed enrichment of Ezh2/H3K27me3 at silenced loci in HBx-transgenic mouse-derived HCCs, including Igfbp4 whose down-regulation significantly correlated with EZH2 overexpression and poor survivals of HCC patients. Functional characterizations demonstrated potent growth- and invasion-suppressive functions of IGFBP4, which was associated with transcriptomic alterations leading to deregulation of multiple signaling pathways. Mechanistically, IGFBP4 stimulated AKT/EZH2 phosphorylation to abrogate H3K27me3-mediated silencing, forming a reciprocal feedback loop that suppressed core transcription factor networks (FOXA1/HNF1A/HNF4A/KLF9/NR1H4) for normal liver homeostasis. Consequently, the in vivo tumorigenicity of IGFBP4-silenced HCC cells was vulnerable to pharmacological inhibition of EZH2, but not AKT. Our study unveils chromatin regulation of a novel liver tumor suppressor IGFBP4, which constitutes an AKT-EZH2 reciprocal loop in driving H3K27me3-mediated epigenetic reprogramming. Defining the aberrant chromatin landscape of HCC sheds light into the mechanistic basis of effective EZH2-targeted inhibition.


Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone Code/genetics , Histones/metabolism , Insulin-Like Growth Factor Binding Protein 4/deficiency , Liver Neoplasms/genetics , Tumor Suppressor Proteins/deficiency , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/physiology , Liver Neoplasms, Experimental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy , Prognosis , Protein Interaction Mapping , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/genetics , Sequence Analysis, RNA , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Xenograft Model Antitumor Assays
11.
J Pathol ; 245(4): 491-501, 2018 08.
Article En | MEDLINE | ID: mdl-29774544

Peritoneal fibrosis is a common complication of abdominal and pelvic surgery, and can also be triggered by peritoneal dialysis, resulting in treatment failure. In these settings, fibrosis is driven by activated myofibroblasts that are considered to be partly derived by mesothelial-to-mesenchymal transition (MMT). We hypothesized that, if the molecular signature of MMT could be better defined, these insights could be exploited to block this pathological cellular transition. Rat peritoneal mesothelial cells were purified by the use of an antibody against HBME1, a protein present on mesothelial cell microvilli, and streptavidin nanobead technology. After exposure of sorted cells to a well-known mediator of MMT, transforming growth factor (TGF)-ß1, RNA sequencing was undertaken to define the transcriptomes of mesothelial cells before and during early-phase MMT. MMT was associated with dysregulation of transcripts encoding molecules involved in insulin-like growth factor (IGF) and bone morphogenetic protein (BMP) signalling. The application of either recombinant BMP4 or IGF-binding protein 4 (IGFBP4) ameliorated TGF-ß1-induced MMT in culture, as judged from the retention of epithelial morphological and molecular phenotypes, and reduced migration. Furthermore, peritoneal tissue from peritoneal dialysis patients showed less prominent immunostaining than control tissue for IGFBP4 and BMP4 on the peritoneal surface. In a mouse model of TGF-ß1-induced peritoneal thickening, BMP4 immunostaining on the peritoneal surface was attenuated as compared with healthy controls. Finally, genetic lineage tracing of mesothelial cells was used in mice with peritoneal injury. In this model, administration of BMP4 ameliorated the injury-induced shape change and migration of mesothelial cells. Our findings demonstrate a distinctive MMT signature, and highlight the therapeutic potential for BMP4, and possibly IGFBP4, to reduce MMT. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis , Peritoneal Fibrosis/genetics , Peritoneum/metabolism , Transcriptome , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Movement , Cell Shape , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice, Inbred C57BL , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Peritoneum/drug effects , Peritoneum/pathology , Rats, Wistar , Transforming Growth Factor beta1/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Growth Horm IGF Res ; 39: 54-61, 2018 04.
Article En | MEDLINE | ID: mdl-29398370

INTRODUCTION: Pregnancy-associated plasma protein-A (PAPP-A) stimulates insulin-like growth factor (IGF)-I action through proteolytic cleavage of IGF binding protein-4 (IGFBP-4). Recently, stanniocalcin-2 (STC2) was discovered as an inhibitor of PAPP-A. Most members of the IGF system are expressed in adipose tissue (AT), but there is a relative paucity of information on the distribution of IGFBP-4, PAPP-A, and STC2 in different AT depots. Since IGF-I expression in AT is highly GH-dependent, we used bovine GH transgenic (bGH) and GH receptor knockout (GHR-/-) mice to investigate AT depot-specific expression patterns of IGFBP-4, PAPP-A, and STC2, and whether the regulation is GH-dependent. METHODS: Seven-month-old male bGH, GHR-/- and wild type (WT) control mice were used. Body composition was determined, and subcutaneous, epididymal, retroperitoneal, mesenteric and brown adipose tissue (BAT) depots were collected. RNA expression of Igfbp4, Pappa, and Stc2 was assessed by reverse transcription quantitative PCR and IGFBP-4 protein by Western blotting. RESULTS: Igfbp4, Pappa, and Stc2 RNA levels were differentially expressed in an AT depot-dependent manner in WT mice. Igfbp4 RNA levels were significantly higher in all white AT depots than in BAT. Pappa was most highly expressed in the mesenteric depot: levels were 7.5-fold higher in mesenteric than in subcutaneous AT (p < .001). Although intraabdominal in origin, epididymal and retroperitoneal Pappa expression levels were 69% and 68% lower, respectively, as compared to mesenteric levels (p < .001). Stc2 RNA expression was significantly higher in all intraabdominal white AT as compared to subcutaneous AT and BAT; levels in epididymal, retroperitoneal, and mesenteric were all more than three-fold higher than in subcutaneous AT (p < .001) and 12-fold higher than in BAT (p < .001). Gene expression patterns in bGH and GHR-/- mice mimicked those in WT mice, suggesting that GH does not affect the transcription of the STC2-PAPP-A-IGFBP-4-axis in AT. However, proteins levels of intact IGFBP-4 were significantly increased in bGH mice and decreased in GHR-/- mice, whereas the PAPP-A-generated IGFBP-4 fragment level was unaltered. CONCLUSION: Expression of Igfbp4, Pappa, and Stc2 differ between AT depots and is generally higher in white AT than in BAT. The transcription appears to occur in a GH-independent manner, whereas IGFBP-4 protein levels are highly influenced by altered GH activity.


Adipose Tissue/metabolism , Carrier Proteins/physiology , Glycoproteins/metabolism , Growth Hormone/physiology , Human Growth Hormone/administration & dosage , Insulin-Like Growth Factor Binding Protein 4/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Animals , Body Composition , Cattle , Cells, Cultured , Female , Gene Expression Regulation , Glycoproteins/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy , Pregnancy-Associated Plasma Protein-A/genetics
13.
Int J Mol Sci ; 18(11)2017 Nov 07.
Article En | MEDLINE | ID: mdl-29112142

Adipose tissue has traditionally only been considered as an energy storage organ. Nevertheless, the importance of this tissue in systemic physiology and, especially, in systemic inflammation has been highlighted in recent years. Adipose tissue expresses proteins related to vitamin D (VD) metabolism, and it has been proposed that it can act as a VD storage tissue. The active form of VD, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), is able to modify adipocyte and adipose tissue physiology via the VD receptor (VDR), decreasing the expression of pro-inflammatory cytokines in adipose tissue. Moreover, VD deficiency and VDR has been reported to be associated with obesity and diabetes. However, the results of the different studies are not conclusive. Insulin growth binding proteins (IGFBPs) have been identified in adipose tissue, but their roles are poorly understood. Therefore, the objective of this study was to analyze the plasma levels of VD and the gene expression of VDR in the adipose tissue of subjects with morbid obesity (MO) and with different degrees of insulin resistance (IR), as well as the functionality of direct interaction between IGFBP-3 and VDR, which could explain its inhibitory role in adipogenesis. Our results show a novel role of the VD system in the regulation and activation of IGFBP-3 in visceral adipose tissue (VAT) of patients with MO, as a new and alternative mechanism proposed in the insulin signaling associated with obesity.


Adipose Tissue/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Obesity/metabolism , Receptors, Calcitriol/metabolism , Adult , Female , Humans , Insulin Resistance/genetics , Insulin Resistance/physiology , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/metabolism , Intra-Abdominal Fat/metabolism , Male , Middle Aged , Obesity/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Regression Analysis , Signal Transduction/genetics , Signal Transduction/physiology
14.
Mol Cancer ; 16(1): 154, 2017 09 25.
Article En | MEDLINE | ID: mdl-28946875

BACKGROUND: Reprogrammed energy metabolism as an emerging hallmark of cancer has recently drawn special attention since it facilitate cell growth and proliferation. Recently, long noncoding RNAs (lncRNAs) have been served as key regulators implicated in tumor development and progression by promoting proliferation, invasion and metastasis. However, the associations of lncRNAs with cellular energy metabolism in lung cancer (LC) need to be clarified. METHODS: Here, we conducted bioinformatics analysis and found insulin-like growth factor binding protein 4-1 (IGFBP4-1) as a new candidate lncRNA located in the upstream region of IGFBP4 gene. The expression levels of lnc-IGFBP4-1, mRNA levels of IGFBP4 in 159 paired lung cancer samples and adjacent, histological normal tissues by qRT-PCR. Over-expression and RNA interference (RNAi) approaches were adopted to investigate the biological functions of lnc-IGFBP4-1. The intracellular ATP level was measured using the Cell Titer-Glo Luminescent Cell Viability Assay kit, and changes in metabolic enzymes were examined in cancer cells and normal pulmonary epithelial cells with qRT-PCR. RESULTS: Our results showed that lnc-IGFBP4-1 was significantly up-regulated in LC tissues compared with corresponding non-tumor tissues (P < 0.01), and its expression level was significantly correlated with TNM stage (P < 0.01) and lymph node metastasis (P < 0.05). Further investigation showed that overexpression of lnc-IGFBP4-1 significantly promoted LC cell proliferation in vitro and in vivo, while downregulation of endogenous lnc-IGFBP4-1 could inhibited cell proliferation and induce apoptosis. Moreover, we found lnc-IGFBP4-1 could influences ATP production levels and expression of enzymes including HK2, PDK1 and LDHA, in addition, decline in both ATP production and these enzymes in response to 2-DG and 2-DG-combined Rho123, respectively, was observed in lnc-IGFBP4-1-overespressing LC cells, indicative of an enhanced aerobic glycolysis rate. Finally, lnc-IGFBP4-1 was observed to negatively correlate with gene IGFBP4, and lower expression level of IGFPB4 was found after lnc-IGFBP4-1-overexpression was transfected into PC9 cells, higher expression level of IGFPB4 was also found after lnc-IGFBP4-1-downregulation was transfected into GLC-82 cells, which indicates that IGFBP4 may exert its targeting function regulated by lnc-IGFBP4-1. CONCLUSIONS: Taken together, these findings provide the first evidence that lnc-IGFBP4-1 is significantly up-regulated in LC tissues and plays a positive role in cell proliferation and metastasis through possible mechanism of reprogramming tumor cell energy metabolism, which suggests that lnc-IGFBP4-1 may be a promising biomarker in LC development and progression and as a potential therapeutic target for LC intervention.


Energy Metabolism/genetics , Gene Expression , Insulin-Like Growth Factor Binding Protein 4/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , RNA, Long Noncoding/genetics , Adenosine Triphosphate/metabolism , Adult , Aged , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Neoplasm Staging
15.
Endocrinology ; 158(10): 3488-3500, 2017 10 01.
Article En | MEDLINE | ID: mdl-28938423

Insulinlike growth factor (IGF) I induces adipogenesis in vitro. IGF-binding protein 4 (IGFBP4) is highly expressed in adipocytes and osteoblasts and is inhibitory of IGFs in vitro. We previously reported that Igfbp4 null mice (Igfbp4-/-) had decreased fat proportions at 8 and 16 weeks of age. However, the mechanism leading to the reduced adiposity remains unknown. The purpose of this study was to elucidate how IGFBP4 mediates adipose tissue development in vivo. Our results showed that inguinal and gonadal white adipose tissue (gWAT) from Igfbp4-/- mice had decreased weights and Pparγ expression. Cultures of primary bone marrow stromal cells (BMSCs) and ear mesenchymal stem cells (eMSCs) from mutant mice showed reduced adipogenesis. Both BMSCs and eMSC had a strong induction of Igfbp4 expression during adipogenesis. Furthermore, the increase in phosphorylated Akt (p-Akt), a downstream target of IGF-I signaling, in wild-type cells, was blunted in mutant eMSCs. On a high-fat diet (HFD) there were sexual differences in adipocyte expansion of Igfbp4-/- mice. Mutant males gained weight by expanding their white fat depots. However, Igfbp4-/- female mice were protected against diet-induced obesity. Ovariectomized Igfbp4-/- female mice gained weight in a manner similar to that seen in ovariectomized controls. Thus, Igfbp4 is required for inguinal fat expansion in female mice but not in male mice. However, gWAT expansion, which is prevented by estrogen during HFD, does not require Igfbp4.


Adipogenesis/genetics , Adipose Tissue, White/metabolism , Body Fat Distribution , Insulin-Like Growth Factor Binding Protein 4/genetics , Absorptiometry, Photon , Adipocytes , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue, White/pathology , Animals , Blotting, Western , Cells, Cultured , Diet, High-Fat , Female , Gene Expression Profiling , Male , Mesenchymal Stem Cells , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Ovariectomy , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
16.
Cell Physiol Biochem ; 42(2): 640-650, 2017.
Article En | MEDLINE | ID: mdl-28595186

BACKGROUND/AIMS: Insulin-like growth factor binding proteins (IGFBP) play important roles in bone metabolism. IGFBP4 is involved in senescent-associated phenomena in mesenchymal stem cells (MSCs). The goal of the present study was to determine whether age-related IGFBP4 overexpression is associated with the impaired osteogenic differentiation potential of aged bone marrow derived MSCs. METHODS: MSCs were isolated from Sprague-Dawley rats aged 3-26 months. The bone morphogenetic protein (BMP)-2-induced osteogenic differentiation of rat MSCs was assessed by analyzing the expression levels of osteoblast marker genes [runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), and osteocalcin (OC)], ALP activity and calcification. RESULTS: Our study showed that IGFBP4 mRNA and protein expression increased with age in parallel with impaired osteogenic differentiation of MSCs cultured in BMP2-containing osteogenic medium, as evidenced by the downregulation of osteoblast marker genes, and decreased ALP activity and calcium deposits. IGFBP4 overexpression impaired BMP2-induced osteogenic differentiation potential of young MSCs, whereas IGFBP4 knockdown restored the osteogenic potency of aged MSCs. Moreover, IGFBP4 knockdown stimulated the activation of Erk and Smad by increasing phosphorylation. CONCLUSION: Collectively, our results demonstrate that IGFBP4 overexpression plays a role in the impairment of MSC differentiation potential via the Erk and Smad pathways, suggesting potential targets to improve MSC function for cell therapy applications.


Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/genetics , Insulin-Like Growth Factor Binding Protein 4/biosynthesis , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Animals , Bone Marrow Cells/drug effects , Cells, Cultured , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor Binding Protein 4/genetics , MAP Kinase Signaling System/genetics , Mesenchymal Stem Cells/cytology , Osteoblasts/metabolism , Phosphorylation , RNA, Messenger/biosynthesis , Rats , Smad Proteins/genetics
17.
Endocr Regul ; 51(2): 84-95, 2017 Apr 25.
Article En | MEDLINE | ID: mdl-28609285

OBJECTIVE: The aim of the present study was to examine the effect of chromium disilicide and titanium nitride nanoparticles on the expression level of genes encoding important regulatory factors (IGFBP1, IGFBP2, IGFBP3, IGFBP4, IGFBP5, SNARK/NUAK2, CD36, and PECAM1/CD31) in mouse liver for evaluation of possible toxic effects of these nanoparticles. METHODS: Male mice received 20 mg chromium disilicide nanoparticles (45 nm) and titanium nitride nanoparticles (20 nm) with food every working day for 2 months. The expression of IGFBP1, IGFBP2, IGFBP3, IGFBP4, IGFBP5, SNARK, CD36, and PECAM1 genes in mouse liver was studied by quantitative polymerase chain reaction. RESULTS: Treatment of mice with chromium disilicide nanoparticles led to down-regulation of the expression of IGFBP2, IGFBP5, PECAM1, and SNARK genes in the liver in comparison with control mice, with more prominent changes for SNARK gene. At the same time, the expression of IGFBP3 and CD36 genes was increased in mouse liver upon treatment with chromium disilicide nanoparticles. We have also shown that treatment with titanium nitride nanoparticles resulted in down-regulation of the expression of IGFBP2 and SNARK genes in the liver with more prominent changes for SNARK gene. At the same time, the expression of IGFBP3, IGFBP4, and CD36 genes was increased in the liver of mice treated with titanium nitride nanoparticles. Furthermore, the effect of chromium disilicide nanoparticles on IGFBP2 and CD36 genes expression was significantly stronger as compared to titanium nitride nanoparticles. CONCLUSIONS: The results of this study demonstrate that chromium disilicide and titanium nitride nanoparticles have variable effects on the expression of IGFBP2, IGFBP3, IGFBP4, IGFBP5, SNARK, CD36, and PECAM1 genes in mouse liver, which may reflect the genotoxic activities of the studied nanoparticles.


Gene Expression/drug effects , Liver/drug effects , Nanoparticles , RNA, Messenger/drug effects , Titanium/pharmacology , Animals , CD36 Antigens/drug effects , CD36 Antigens/genetics , Chromium Compounds/pharmacology , Down-Regulation , Insulin-Like Growth Factor Binding Protein 1/drug effects , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 2/drug effects , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 3/drug effects , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 4/drug effects , Insulin-Like Growth Factor Binding Protein 4/genetics , Liver/metabolism , Male , Mice , Platelet Endothelial Cell Adhesion Molecule-1/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Silicon Compounds/pharmacology , Up-Regulation
18.
J Endocrinol ; 233(1): 131-144, 2017 04.
Article En | MEDLINE | ID: mdl-28184001

Insulin-like growth factor-1 (IGF-1) and its binding proteins are critical mediators of skeletal growth. Insulin-like growth factor-binding protein 4 (IGFBP-4) is highly expressed in osteoblasts and inhibits IGF-1 actions in vitro Yet, in vivo studies suggest that it could potentiate IGF-1 and IGF-2 actions. In this study, we hypothesized that IGFBP-4 might potentiate the actions of IGF-1 on the skeleton. To test this, we comprehensively studied 8- and 16-week-old Igfbp4-/- mice. Both male and female adult Igfbp4-/- mice had marked growth retardation with reductions in body weight, body and femur lengths, fat proportion and lean mass at 8 and 16 weeks. Marked reductions in aBMD and aBMC were observed in 16-week-old Igfbp4-/- females, but not in males. Femoral trabecular BV/TV and thickness, cortical fraction and thickness in 16-week-old Igfbp4-/- females were significantly reduced. However, surprisingly, males had significantly more trabeculae with higher connectivity density than controls. Concordantly, histomorphometry revealed higher bone resorption and lower bone formation in Igfbp4-/- females. In contrast, Igfbp4-/- males had lower mineralized surface/bone surface. Femoral expression of Sost and circulating levels of sclerostin were reduced but only in Igfbp4-/- males. Bone marrow stromal cultures from mutants showed increased osteogenesis, whereas osteoclastogenesis was markedly increased in cells from Igfbp4-/- females but decreased in males. In sum, our results indicate that loss of Igfbp4 affects mesenchymal stromal cell differentiation, regulates osteoclastogenesis and influences both skeletal development and adult bone maintenance. Thus, IGFBP-4 modulates the skeleton in a gender-specific manner, acting as both a cell autonomous and cell non-autonomous factor.


Bone Development/physiology , Bone and Bones/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Osteogenesis/physiology , Sex Characteristics , Animals , Body Weight/physiology , Female , Insulin-Like Growth Factor Binding Protein 4/genetics , Male , Mice , Mice, Knockout , Osteoclasts/metabolism
19.
Clin Gastroenterol Hepatol ; 15(6): 937-944.e5, 2017 Jun.
Article En | MEDLINE | ID: mdl-28065765

BACKGROUND & AIMS: There is a close relationship between hepatitis C virus (HCV) infection and lichen planus, a chronic inflammatory mucocutaneous disease. We performed a genome-wide association study (GWAS) to identify genetic variants associated with HCV-related lichen planus. METHODS: We conducted a GWAS of 261 patients with HCV infection treated at a tertiary medical center in Japan from October 2007 through January 2013; a total of 71 had lichen planus and 190 had normal oral mucosa. We validated our findings in a GWAS of 38 patients with HCV-associated lichen planus and 7 HCV-infected patients with normal oral mucosa treated at a medical center in Italy. RESULTS: Single-nucleotide polymorphisms in NRP2 (rs884000) and IGFBP4 (rs538399) were associated with risk of HCV-associated lichen planus (P < 1 × 10-4). We also found an association between a single-nucleotide polymorphism in the HLA-DR/DQ genes (rs9461799) and susceptibility to HCV-associated lichen planus. The odds ratios for the minor alleles of rs884000, rs538399, and rs9461799 were 3.25 (95% confidence interval, 1.95-5.41), 0.40 (95% confidence interval, 0.25-0.63), and 2.15 (95% confidence interval, 1.41-3.28), respectively. CONCLUSIONS: In a GWAS of Japanese patients with HCV infection, we replicated associations between previously reported polymorphisms in HLA class II genes and risk for lichen planus. We also identified single-nucleotide polymorphisms in NRP2 and IGFBP4 loci that increase and reduce risk of lichen planus, respectively. These genetic variants might be used to identify patients with HCV infection who are at risk for lichen planus.


Genetic Predisposition to Disease , Hepatitis C/complications , Insulin-Like Growth Factor Binding Protein 4/genetics , Lichen Planus/genetics , Neuropilin-2/genetics , Polymorphism, Single Nucleotide , Aged , Female , Genome-Wide Association Study , Humans , Japan , Male , Middle Aged , Tertiary Care Centers
20.
Circulation ; 134(24): 1991-2007, 2016 Dec 13.
Article En | MEDLINE | ID: mdl-27803037

BACKGROUND: Myocardial infarction is one of the leading causes of morbidity and mortality worldwide, triggering irreversible myocardial cell damage and heart failure. The role of low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) as coreceptors of the Wnt/ß-catenin pathway in the adult heart remain unknown. Insulin-like growth factor binding protein 4 and dickkopf-related protein 1 (Dkk1) are 2 secreted LRP5/6 binding proteins that play a crucial role in heart development through preventing Wnt/ß-catenin pathway activation. However, their roles in the adult heart remain unexplored. METHODS: To understand the role of LRP5/6 and ß-catenin in the adult heart, we constructed conditional cardiomyocyte-specific LRP5/6 and ß-catenin knockout mice and induced surgical myocardial infarction. We also directly injected recombinant proteins of insulin-like growth factor binding protein 4 and Dkk1 into the heart immediately following myocardial infarction to further examine the mechanisms through which these proteins regulate LRP5/6 and ß-catenin. RESULTS: Deletion of LRP5/6 promoted cardiac ischemic insults. Conversely, deficiency of ß-catenin, a downstream target of LRP5/6, was beneficial in ischemic injury. It is interesting to note that although both insulin-like growth factor binding protein 4 and Dkk1 are secreted Wnt/ß-catenin pathway inhibitors, insulin-like growth factor binding protein 4 protected the ischemic heart by inhibiting ß-catenin, whereas Dkk1 enhanced the injury response mainly through inducing LRP5/6 endocytosis and degradation. CONCLUSIONS: Our findings reveal previously unidentified dual roles of LRP5/6 involved in the cardiomyocyte response to ischemic injury. These findings suggest new therapeutic strategies in ischemic heart disease by fine-tuning LRP5/6 and ß-catenin signaling within the Wnt/ß-catenin pathway.


Insulin-Like Growth Factor Binding Protein 4/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Myocardial Ischemia/pathology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , DNA Damage/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Histones/metabolism , Hydrogen Peroxide/toxicity , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/therapeutic use , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/therapeutic use , Low Density Lipoprotein Receptor-Related Protein-5/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Ischemia/metabolism , Myocardial Ischemia/prevention & control , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
...