Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Stem Cells ; 41(4): 400-414, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36682027

ABSTRACT

Chordin like-1 (CHRDL1) is an antagonist of bone morphogenetic proteins (BMPs) that acts through binding BMPs and blocking their interaction with BMP receptors. CHRDL1 plays a role in osteoblast differentiation but controversial effects were reported. On the other hand, the role of CHRDL1 in adipogenesis is unknown. In the present study, we investigated the function of CHRDL1 in regulating differentiation of osteoblasts and adipocytes and elucidated the underlying mechanism. CHRDL1 expression was downregulated during osteogenesis while it was upregulated during adipogenesis in primary cultured and established mesenchymal progenitor cell lines. Functional experiments revealed that CHRDL1 suppressed osteoblast differentiation and promoted adipocyte differentiation. Mechanistic explorations revealed that CHRDL1 is directly bound to insulin-like growth factor binding protein 3 (IGFBP3) and attenuated the degradation of the latter. Furthermore, CHRDL1 and IGFBP3 suppressed the activity of insulin receptor substrate 1 (IRS1)/AKT serine/threonine kinase (AKT)/mechanistic target of rapamycin kinase complex 1 (mTORC1) signaling in progenitor cells undergoing osteogenic differentiation. By contrast, they activated AKT/mTORC1 signaling independently of IRS1 during adipogenic differentiation. CHRDL1 enhanced the interaction of nuclear IGFBP3 and retinoid X receptor α (RXRα) during adipogenesis, and inhibition of RXR inactivated AKT and attenuated the stimulation of adipogenic differentiation by CHRDL1. Overexpression of IGFBP3 relieved the perturbation of osteogenic and adipogenic differentiation of progenitor cells induced by CHRDL1 silencing. Finally, CHRDL1 and IGFBP3 were upregulated in the trabecular bone of aged mice. Our study provides evidence that CHRDL1 reciprocally regulates osteoblast and adipocyte differentiation through stabilizing IGFBP3 and differentially modulating AKT/mTORC1 signaling.


Subject(s)
Osteogenesis , Proto-Oncogene Proteins c-akt , Animals , Mice , Adipocytes/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Eye Proteins/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Nerve Tissue Proteins/metabolism , Osteoblasts/metabolism , Proto-Oncogene Proteins c-akt/metabolism
2.
Clin Endocrinol (Oxf) ; 94(4): 551-562, 2021 04.
Article in English | MEDLINE | ID: mdl-33249593

ABSTRACT

OBJECTIVE: Previous studies suggested that recombinant human IGF-1 (rhIGF-1) administration affects carbohydrate and lipid metabolism in healthy people and in people with diabetes. This study aimed to determine the effects of rhIGF-1/rhIGF binding protein-3 (rhIGFBP-3) administration on glucose homeostasis and lipid metabolism in healthy recreational athletes. DESIGN AND SETTING: Randomized, double-blind, placebo-controlled rhIGF-1/rhIGFBP-3 administration study at Southampton General Hospital, UK. PARTICIPANTS: 56 recreational athletes (30 men, 26 women). METHODS: Participants were randomly assigned to receive placebo, low-dose rhIGF-1/rhIGFBP-3 (30 mg/day) or high-dose rhIGF-1/rhIGFBP-3 (60 mg/day) for 28 days. The following variables were measured before and immediately after the treatment period: fasting lipids, glucose, insulin, C-peptide and glycated haemoglobin. The homeostatic model assessment (HOMA-IR) was used to estimate insulin sensitivity and indirect calorimetry to assess substrate oxidation rates. The general linear model approach was used to compare treatment group changes with the placebo group. RESULTS: Compared with the placebo group, there was a significant reduction in fasting triglycerides in participants treated with high-dose rhIGF-1/rhIGFBP-3 (p = .030), but not in the low-dose group (p = .390). In women, but not in men, there were significant increases in total cholesterol (p = .003), HDL cholesterol (p = .001) and LDL cholesterol (p = .008). These lipid changes were associated with reduced fasting insulin (p = .010), C-peptide (p = .001) and HOMA-IR (p = .018) in women and reduced C-peptide (p = .046) in men. CONCLUSIONS: rhIGF-1/rhIGFBP-3 administration for 28 days reduced insulin concentration, improved insulin sensitivity and had significant effects on lipid profile including decreased fasting triglycerides.


Subject(s)
Athletes , Carrier Proteins , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor I , Carbohydrate Metabolism , Double-Blind Method , Female , Humans , Insulin , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Lipid Metabolism , Male , Recombinant Proteins/pharmacology
3.
Hum Mol Genet ; 22(21): 4306-17, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23773998

ABSTRACT

Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is an autosomal recessive disorder caused by mutations in the laminin-α2 gene (OMIM: 607855). Currently, no treatment other than palliative care exists for this disease. In our previous work, genetic interventions in the Lama2(Dy-w) mouse model for MDC1A demonstrated that limited regeneration and uncontrolled apoptosis are important drivers of this disease. However, targeting one of these disease drivers without addressing the other results in only partial rescue of the phenotype. The present study was designed to determine whether utilizing a combinatorial treatment approach can lead to a more profound amelioration of the disease pathology. To accomplish this task, we generated Bax-null Lama2(Dy-w)mice that overexpressed muscle-specific IGF-1 (Lama2(Dy-w)Bax(-/-)+IGF-1tg). Further to test the translational potential of IGF-1 administration in combination with Bax inhibition, we treated Lama2(Dy-w)Bax(-/-) mice postnatally with systemic recombinant human IGF-1 (IPLEX™). These two combinatorial treatments lead to similar, promising outcomes. In addition to increased body and muscle weights, both transgenic overexpression and systemic administration of IGF-1 combined with Bax-inhibition resulted in improved muscle phenotype and locomotory function that were nearly indistinguishable from wild-type mice. These results provide a fundamental proof of concept that justifies the use of a combination therapy as an effective treatment for MDC1A and highlights a compelling argument toward shifting the paradigm in treating multifaceted neuromuscular diseases.


Subject(s)
Apoptosis/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Muscular Dystrophies/physiopathology , Muscular Dystrophies/therapy , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics , Animals , Apoptosis/genetics , Body Weight/drug effects , Combined Modality Therapy , Gene Expression Regulation , Humans , Insulin-Like Growth Factor Binding Protein 3/administration & dosage , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Laminin/genetics , Laminin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/physiology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Phenotype , Regeneration , Treatment Outcome , bcl-2-Associated X Protein/metabolism
4.
Mol Vis ; 21: 604-11, 2015.
Article in English | MEDLINE | ID: mdl-26015772

ABSTRACT

PURPOSE: In models of diabetic retinopathy, insulin-like growth factor binding protein-3 (IGFBP-3) is protective to the retina, especially retinal microvascular endothelial cells (RECs), but the underlying mechanisms are unclear. For this study, we hypothesized that IGFBP-3 may reduce vascular endothelial growth factor (VEGF) levels through reduced endothelial nitric oxide synthase (eNOS) activity, which may be protective against macular edema. METHODS: To test this hypothesis, we grew primary human retinal endothelial cells in normal glucose (5 mM) or high glucose (25 mM) for three days, treated with IGFBP-3 NB plasmid (a plasmid of IGFBP-3 that cannot bind IGF-1), followed by western blotting for eNOS, protein kinase C zeta (PKCzeta), and VEGF. Additionally, we treated some cells with recombinant eNOS or PKCzeta, after IGFBP-3 NB plasmid transfection to validate that these pathways regulate VEGF levels. Immunoprecipitation experiments were done with the eNOS antibody, followed by western blotting for PKCzeta, to determine if eNOS and PKCzeta interact directly. RESULTS: Our results suggest that 1) IGFBP-3 inhibits the endothelial nitric oxide synthase (eNOS) and protein kinase C zeta (PKCzeta) pathway, which in turn inhibits VEGF production, and 2) that eNOS plays a role in activating PKCzeta to increase VEGF levels in diabetic retinopathy. CONCLUSIONS: In conclusion, IGFBP-3 may be a novel treatment for macular edema through the inhibition of eNOS and PKCzeta activation, leading to reduced VEGF levels.


Subject(s)
Diabetic Retinopathy/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein Kinase C/metabolism , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cells, Cultured , Diabetic Retinopathy/drug therapy , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucose/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Retinal Vessels/drug effects , Signal Transduction
5.
Eur J Pharmacol ; 969: 176421, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38423242

ABSTRACT

Fluoroquinolones (FQs), commonly known for their antibiotic properties, exhibit additional pharmacological potential with anti-proliferative effects on various malignant cell types and immunomodulatory responses. Despite these observed effects, the precise mechanisms of action remain elusive. This study elucidates the biological impact of FQs on insulin-like growth factor-binding protein 3 (IGFBP-3) productions in a p53-dependent manner. Cultured cells and mouse models treated with FQs demonstrated increased IGFBP-3 mRNA expression and protein secretion. The FQ-induced IGFBP-3 was identified to impede cell growth by inhibiting IGF-I signaling and exerting effects through an IGF-independent pathway. Notably, FQ-mediated suppression of cell proliferation was reversed in p53-null and p53 knockdown cells, suggesting the pivotal role of p53 in FQ-induced IGFBP-3 production and IGFBP-3-mediated growth inhibition. Additionally, ciprofloxacin, a clinically used FQ, exhibited the induction of tumor cell apoptosis and attenuation of tumor growth in a syngeneic mouse hepatocellular carcinoma (HCC) model. These findings unveil a novel mechanism through which FQs act as anti-proliferative agents, prompting further exploration of their potential utility or derivative compounds in cancer treatment and prevention.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Fluoroquinolones/pharmacology , Insulin-Like Peptides , Tumor Suppressor Protein p53 , Insulin-Like Growth Factor I/metabolism , Cell Proliferation
6.
Apoptosis ; 18(3): 361-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291901

ABSTRACT

The goal of this study was to investigate whether insulin-like growth factor binding protein-3 receptor (IGFBP-3 receptor) is required for IGFBP-3 to inhibit retinal endothelial cell (REC) apoptosis. REC were grown in normal glucose (5 mM) or high glucose medium (25 mM) for 3 days. Once cells reached confluence, they were transfected with an endothelial- specific IGFBP-3 plasmid DNA (non-IGF binding; IGFBP-3 NB) at 1 µg/ml for 24 h. Cell proteins were extracted and analyzed for IGFBP-3 receptor expression by Western blotting or use in coimmunoprecipitation or co-localization experiments for detection of IGFBP-3 and IGFBP-3 receptor binding. REC were also transfected with or without IGFBP-3 receptor siRNA before IGFBP-3NB plasmid DNA transfection. Cell lysates were processed for a cell death ELISA, a cleaved caspase 3 ELISA, and Western blotting to measure key pro- and anti-apoptotic markers: Bcl-xL, Bax, Cytochrome C and Akt. The IGFBP-3 receptor is present on REC. Overexpression of IGFBP-3 in REC significantly increased protein levels of IGFBP-3 receptor (p < 0.05). Significant increases in cell death were found in cells transfected with IGFBP-3 receptor siRNA versus not treated samples (p < 0.05). Data suggest that IGFBP-3 inhibits retinal endothelial cell death through activation of an IGFBP-3 receptor in a hyperglycemic environment. This is the first demonstration of the involvement of IGFBP-3 receptor in inhibition of REC cell death. Future studies will investigate the mechanism by which IGFBP-3 receptor may inhibit retinal endothelial cell death.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/pathology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Receptors, Cell Surface/metabolism , Retina/cytology , Cells, Cultured , Glucose/administration & dosage , Humans , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Osmolar Concentration , Receptors, Cell Surface/biosynthesis
7.
Blood ; 118(9): 2622-31, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21551235

ABSTRACT

Most antiangiogenic therapies currently being evaluated in clinical trials target the vascular endothelial growth factor pathway; however, the tumor vasculature can acquire resistance to vascular endothelial growth factor-targeted therapy by shifting to other angiogenesis mechanisms. Insulin-like growth factor binding protein-3 (IGFBP-3) has been reported to suppress tumor growth and angiogenesis by both IGF-dependent and IGF-independent mechanisms; however, understanding of its IGF-independent mechanisms is limited. We observed that IGFBP-3 blocked tumor angiogenesis and growth in non-small cell lung cancer and head and neck squamous cell carcinoma. Conditioned media from an IGFBP-3-treated non-small cell lung cancer cell line displayed a significantly decreased capacity to induce HUVEC proliferation and aortic sprouting. In cancer cells, IGFBP-3 directly interacted with Erk1/2, leading to inactivation of Erk1/2 and Elk-1, and suppressed transcription of early growth response protein 1 and its target genes, basic fibroblast growth factor and platelet-derived growth factor. These data suggest that IGF-independent Erk1/2 inactivation and decreased IGFBP-3-induced Egr-1 expression block the autocrine and paracrine loops of angiogenic factors in vascular endothelial and cancer cells. Together, these findings provide a molecular framework of IGFBP-3's IGF-independent antiangiogenic antitumor activities. Future studies are needed for development of IGFBP-3 as a new line of antiangiogengic cancer drug.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma/drug therapy , Early Growth Response Protein 1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/drug therapy , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Promoter Regions, Genetic/drug effects , Transcription, Genetic/drug effects , ets-Domain Protein Elk-1/metabolism , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Carcinoma/blood supply , Carcinoma/pathology , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cells, Cultured/cytology , Cells, Cultured/drug effects , Chick Embryo , Early Growth Response Protein 1/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Female , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/pathology , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/therapeutic use , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Specific Pathogen-Free Organisms , Xenograft Model Antitumor Assays
8.
Arthritis Rheum ; 64(1): 272-80, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21898349

ABSTRACT

OBJECTIVE: To determine the role of insulin-like growth factor binding protein 3 (IGFBP-3) in mediating the effects of transforming growth factor ß (TGFß) on tenascin-C (TN-C) production and to assess the levels of TN-C in vivo in patients with systemic sclerosis (SSc)-associated pulmonary fibrosis. METHODS: Human primary lung fibroblasts were stimulated with TGFß or IGFBP-3 in the presence or absence of specific small interfering RNAs and chemical inhibitors of the signaling cascade. TN-C levels in lung tissue specimens obtained from patients with SSc-associated pulmonary fibrosis were assessed using immunohistochemical analysis and were compared with the levels in specimens obtained from normal donors. TN-C levels were quantified in sera from normal donors and patients with SSc with or without pulmonary fibrosis, using an enzyme-linked immunosorbent assay. RESULTS: IGFBP-3 mediated the induction of TN-C by TGFß. Direct induction of TN-C by IGFBP-3 occurred in a p38 MAP kinase-dependent manner. TN-C levels were abundant in lung tissues from patients with SSc and were localized to subepithelial layers of the distal airways. No TN-C was detectable around the proximal airways. Patients with SSc-associated pulmonary fibrosis had significantly higher levels of circulating TN-C compared with SSc patients without pulmonary fibrosis. Longitudinal samples obtained from patients with SSc before and after the onset of pulmonary fibrosis showed increased levels of TN-C after the onset of pulmonary fibrosis. CONCLUSION: IGFBP-3, which is overexpressed in fibrotic lungs, induces production of TN-C by subepithelial fibroblasts. The increased lung tissue levels of TN-C parallel the levels detected in the sera of SSc patients with pulmonary fibrosis, suggesting that TN-C may be a useful biomarker for SSc-related pulmonary fibrosis.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/pharmacology , Pulmonary Fibrosis/metabolism , Scleroderma, Systemic/metabolism , Tenascin/metabolism , Cells, Cultured , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Lung/drug effects , Lung/metabolism , Lung/pathology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology , RNA Interference , RNA, Small Interfering/administration & dosage , Recombinant Proteins/pharmacology , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology , Tenascin/analysis , Transforming Growth Factor beta/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Fluids Barriers CNS ; 20(1): 59, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37582792

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is essential for normal brain development and regulates essential processes of vascular maturation and stabilization. Importantly, preterm birth is associated with reduced serum levels of IGF-1 as compared to in utero levels. Using a preterm rabbit pup model, we investigated the uptake of systemic recombinant human (rh) IGF-1 in complex with its main binding protein IGF-binding protein 3 (BP-3) to the brain parenchyma via the choroid plexus. Five hours after subcutaneous administration, labeled rhIGF-1/rhIGFBP-3 displayed a widespread presence in the choroid plexus of the lateral and third ventricle, however, to a less degree in the fourth, as well as in the perivascular and subarachnoid space. We found a time-dependent uptake of IGF-1 in cerebrospinal fluid, decreasing with postnatal age, and a translocation of IGF-1 through the choroid plexus. The impact of systemic rhIGF-1/rhIGFBP-3 on IGF-1 receptor activation in the choroid plexus decreased with postnatal age, correlating with IGF-1 uptake in cerebrospinal fluid. In addition, choroid plexus gene expression was observed to increase with postnatal age. Moreover, using choroid plexus in vitro cell cultures, gene expression and protein synthesis were further investigated upon rhIGF-1/rhIGFBP-3 stimulation as compared to rhIGF-1 alone, and found not to be differently altered. Here, we characterize the uptake of systemic rhIGF-1/rhIGFBP-3 to the preterm brain, and show that the interaction between systemic rhIGF-1/rhIGFBP-3 and choroid plexus varies over time.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor I , Animals , Female , Humans , Infant, Newborn , Rabbits , Brain/metabolism , Choroid Plexus/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Recombinant Proteins/metabolism , Animals, Newborn
10.
J Microbiol Biotechnol ; 33(4): 449-462, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-36864505

ABSTRACT

Previously, we confirmed that Mychonastes sp. 246 methanolic extract (ME) markedly reduced the viability of BxPC-3 human pancreatic cancer cells. However, the underlying mechanism ME remained unclear. Hence, we attempted to elucidate the anticancer effect of ME on BxPC-3 human pancreatic cancer cells. First, we investigated the components of ME and their cytotoxicity in normal cells. Then, we confirmed the G1 phase arrest mediated growth inhibitory effect of ME using a cell counting assay and cell cycle analysis. Moreover, we found that the migration-inhibitory effect of ME using a Transwell migration assay. Through RNA sequencing, Gene Ontology-based network analysis, and western blotting, we explored the intracellular mechanisms of ME in BxPC-3 cells. ME modulated the intracellular energy metabolism-related pathway by altering the mRNA levels of IGFBP3 and PPARGC1A in BxPC-3 cells and reduced PI3K and mTOR phosphorylation by upregulating IGFBP3 and 4E-BP1 expression. Finally, we verified that ME reduced the growth of three-dimensional (3D) pancreatic cancer spheroids. Our study demonstrates that ME suppresses pancreatic cancer proliferation through the IGFBP3-PI3K-mTOR signaling pathway. This is the first study on the anticancer effect of the ME against pancreatic cancer, suggesting therapeutic possibilities and the underlying mechanism of ME action.


Subject(s)
Pancreatic Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Proliferation , Cell Line, Tumor , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Apoptosis , Cell Movement/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Pancreatic Neoplasms
11.
Int J Cancer ; 131(10): 2253-63, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22362554

ABSTRACT

Overexpression of insulin-like growth factor binding protein (IGFBP)-3 induces apoptosis of cancer cells. However, preexisting resistance to IGFBP-3 could limit its antitumor activities. This study characterizes the efficacy and mechanism of the combination of recombinant IGFBP-3 (rIGFBP-3) and HDAC inhibitors to overcome IGFBP-3 resistance in a subset of non-small cell lung cancer (NSCLC) and head and neck squamous cell carcinoma (HNSCC) cells. The effects of the combination of rIGFBP-3 and a number of HDAC inhibitors on cell proliferation and apoptosis were assessed in vitro and in vivo by using the MTT assay, a flow cytometry-based TUNEL assay, Western blot analyses and the NSCLC xenograft tumor model. Combined treatment with HDAC inhibitors and rIGFBP-3 had synergistic antiproliferative effects accompanied by increased apoptosis rates in a subset of NSCLC and HNSCC cell lines in vitro. Moreover, combined treatment with depsipeptide and rIGFBP-3 completely suppressed tumor growth and increased the apoptosis rate in vivo in H1299 NSCLC xenografts. Evidence suggests that HDAC inhibitors increased the half-life of rIGFBP-3 protein by blocking protein kinase C (PKC)-mediated phosphorylation and degradation of rIGFBP-3. In addition, combined treatment of IGFBP-3 with an HDAC inhibitor facilitates apoptosis through upregulation of rIGFBP-3 stability and Akt signaling inhibition. The ability of HDAC inhibitors to decrease PKC activation may enhance apoptotic activities of rIGFBP-3 in NSCLC cells in vitro and in vivo. These results indicated that combined treatment with HDAC inhibitor and rIGFBP-3 could be an effective treatment strategy for NSCLC and HNSCC with highly activated PKC.


Subject(s)
Apoptosis/drug effects , Histone Deacetylase Inhibitors/pharmacology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Protein Kinase C/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mice , Mice, Nude , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Stability/drug effects , Proteolysis/drug effects , Squamous Cell Carcinoma of Head and Neck , Transcription, Genetic/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Mol Med ; 18: 1076-85, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22669476

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited neurodegenerative disorder and the first genetic cause of death in childhood. SMA is caused by low levels of survival motor neuron (SMN) protein that induce selective loss of α-motor neurons (MNs) in the spinal cord, resulting in progressive muscle atrophy and consequent respiratory failure. To date, no effective treatment is available to counteract the course of the disease. Among the different therapeutic strategies with potential clinical applications, the evaluation of trophic and/or protective agents able to antagonize MNs degeneration represents an attractive opportunity to develop valid therapies. Here we investigated the effects of IPLEX (recombinant human insulinlike growth factor 1 [rhIGF-1] complexed with recombinant human IGF-1 binding protein 3 [rhIGFBP-3]) on a severe mouse model of SMA. Interestingly, molecular and biochemical analyses of IGF-1 carried out in SMA mice before drug administration revealed marked reductions of IGF-1 circulating levels and hepatic mRNA expression. In this study, we found that perinatal administration of IPLEX, even if does not influence survival and body weight of mice, results in reduced degeneration of MNs, increased muscle fiber size and in amelioration of motor functions in SMA mice. Additionally, we show that phenotypic changes observed are not SMN-dependent, since no significant SMN modification was addressed in treated mice. Collectively, our data indicate IPLEX as a good therapeutic candidate to hinder the progression of the neurodegenerative process in SMA.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/administration & dosage , Insulin-Like Growth Factor Binding Protein 3/therapeutic use , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/therapeutic use , Motor Activity/drug effects , Motor Neurons/pathology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/physiopathology , Animals , Cell Survival/drug effects , Disease Models, Animal , Humans , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Motor Neurons/drug effects , Muscles/drug effects , Muscles/pathology , Muscles/physiopathology , Muscular Atrophy, Spinal/blood , Muscular Atrophy, Spinal/pathology , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Phenotype , Survival of Motor Neuron 2 Protein/genetics , Transcription, Genetic/drug effects
13.
Allergy ; 67(7): 869-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22563687

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) plays a pro-inflammatory mediator as well as a vascular permeability factor in bronchial asthma. Insulin-like growth factor (IGF)-I is also involved in the inflammatory process associated with bronchial asthma and stimulates VEGF expression. The IGF-binding proteins (IGFBPs), especially IGFBP-3, display distinctive properties and can interfere with various biological processes. METHODS: In this study, an ovalbumin (OVA)-induced murine model of allergic airway disease was used to investigate which mechanism is implicated in the preventive and therapeutic actions of IGFBP-3 administered exogenously on allergen-induced bronchial inflammation and airway hyper-responsiveness, in particular focusing on the regulation of VEGF expression. RESULTS: Administration of recombinant human IGFBP-3 to OVA-inhaled mice substantially attenuated the increases in hypoxia-inducible factor (HIF)-α activity, IGF-I production, and VEGF protein levels in the lung. In addition, the blockade of IGF-I action decreased the OVA-induced VEGF expression, airway inflammation, and bronchial hyper-responsiveness. The administration of recombinant human IGFBP-3 or CBO-P11 also reduced significantly increases in inflammatory cells, airway hyper-responsiveness, levels of IL-4, IL-5, IL-13, and vascular permeability in the lung of OVA-inhaled mice. Moreover, when recombinant human IGFBP-3 was administered after the completion of OVA inhalation, these therapeutic effects of IGFBP-3 were also observed. CONCLUSIONS: These results indicate that IGFBP-3 administered exogenously may attenuate antigen-induced airway inflammation and hyper-responsiveness through the modulation of vascular leakage and VEGF expression mediated by HIF-1α/HIF-2α signaling as well as IGF-I action in allergic airway disease of mice.


Subject(s)
Asthma/immunology , Capillary Permeability/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Pneumonia/immunology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Asthma/drug therapy , Asthma/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Endothelial Growth Factors/administration & dosage , Endothelial Growth Factors/pharmacology , Exudates and Transudates/immunology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoglobulin E/blood , Immunoglobulin E/immunology , Insulin-Like Growth Factor Binding Protein 3/administration & dosage , Insulin-Like Growth Factor I/immunology , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pneumonia/drug therapy , Pneumonia/pathology , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Th2 Cells/immunology
14.
Autophagy ; 18(6): 1297-1317, 2022 06.
Article in English | MEDLINE | ID: mdl-34520334

ABSTRACT

Cerebral ischemia is a pathology involving a cascade of cellular mechanisms, leading to the deregulation of proteostasis, including macroautophagy/autophagy, and finally to neuronal death. If it is now accepted that cerebral ischemia induces autophagy, the effect of thrombolysis/energy recovery on proteostasis remains unknown. Here, we investigated the effect of thrombolysis by PLAT/tPA (plasminogen activator, tissue) on autophagy and neuronal death. In two in vitro models of hypoxia reperfusion and an in vivo model of thromboembolic stroke with thrombolysis by PLAT/tPA, we found that ischemia enhances neuronal deleterious autophagy. Interestingly, PLAT/tPA decreases autophagy to mediate neuroprotection by modulating the PI3K-AKT-MTOR pathways both in vitro and in vivo. We identified IGF1R (insulin-like growth factor I receptor; a tyrosine kinase receptor) as the effective receptor and showed in vitro, in vivo and in human stroke patients and that PLAT/tPA is able to degrade IGFBP3 (insulin-like growth factor binding protein 3) to increase IGF1 (insulin-like growth factor 1) bioavailability and thus IGF1R activation.Abbreviations: AKT/protein kinase B: thymoma viral proto-oncogene 1; EGFR: epidermal growth factor receptor; Hx: hypoxia; IGF1: insulin-like growth factor 1; IGF1R: insulin-like growth factor I receptor; IGFBP3: insulin-like growth factor binding protein 3; Ka: Kainate; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MAPK/ERK: mitogen-activated protein kinase; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; OGD: oxygen and glucose deprivation; OGDreox: oxygen and glucose deprivation + reoxygentation; PepA: pepstatin A1; PI3K: phosphoinositide 3-kinase; PLAT/tPA: plasminogen activator, tissue; PPP: picropodophyllin; SCH77: SCH772984; ULK1: unc-51 like kinase 1; Wort: wortmannin.


Subject(s)
Brain Ischemia , Stroke , Autophagy , Brain Ischemia/drug therapy , Glucose/pharmacology , Humans , Hypoxia , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Oxygen/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stroke/drug therapy , TOR Serine-Threonine Kinases/metabolism , Thrombolytic Therapy , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology
15.
Am J Physiol Endocrinol Metab ; 300(6): E1158-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21467302

ABSTRACT

To test the postulate that sex difference, sex steroids, and peptidyl secretagogues control GH autofeedback, 11 healthy postmenopausal women and 14 older men were each given 1) a single iv pulse of GH to enforce negative feedback and 2) continuous iv infusion of saline vs. combined GHRH/GHRP-2 to drive feedback escape during pharmacological estradiol (E(2); women) or testosterone (T; men) supplementation vs. placebo in a double-blind, prospectively randomized crossover design. By three-way ANCOVA, sex difference, sex hormone treatment, peptide stimulation, and placebo/saline responses (covariate) controlled total (integrated) GH recovery during feedback (each P < 0.001). Both sex steroid milieu (P = 0.019) and dual-peptide stimulation (P < 0.001) determined nadir (maximally feedback-suppressed) GH concentrations. E(2)/T exposure elevated nadir GH concentrations during saline infusion (P = 0.003), whereas dual-peptide infusion did so independently of T/E(2) and sex difference (P = 0.001). All three of sex difference (P = 0.001), sex steroid treatment (P = 0.005), and double-peptide stimulation (P < 0.001) augmented recovery of peak (maximally feedback-escaped) GH concentrations. Peak GH responses to dual-peptidyl agonists were greater in women than in men (P = 0.016). E(2)/T augmented peak GH recovery during saline infusion (P < 0.001). Approximate entropy analysis corroborated independent effects of sex steroid treatment (P = 0.012) and peptide infusion (P < 0.001) on GH regularity. In summary, sex difference, sex steroid supplementation, and combined peptide drive influence nadir, peak, and entropic measurements of GH release under controlled negative feedback. To the degree that the pharmacological sex steroid, GH, and dual-peptide clamps provide prephysiological regulatory insights, these outcomes suggest major determinants of pulsatile GH secretion in the feedback domain.


Subject(s)
Feedback, Physiological/physiology , Gonadal Steroid Hormones/pharmacology , Human Growth Hormone/physiology , Aged , Body Mass Index , Double-Blind Method , Estradiol/blood , Female , Gonadotropin-Releasing Hormone/pharmacology , Humans , Insulin-Like Growth Factor Binding Protein 1/pharmacology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Linear Models , Male , Middle Aged , Oligopeptides/pharmacology , Sex Characteristics , Sex Hormone-Binding Globulin/pharmacology , Tandem Mass Spectrometry , Testosterone/blood , Testosterone/pharmacology
16.
Neurochem Res ; 36(3): 406-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21136151

ABSTRACT

Insulin like growth factor-1 (IGF-1) plays an important role in the proliferation and differentiation of neural progenitor cells. The effects of IGF-1 can be regulated by insulin like growth factor binding protein-3 (IGFBP-3) which can either inhibit or stimulate the proliferation of cells depending on the expression of proteases that can release IGF-1 from IGF1-IGFBP3 complex. Although IGF-1 is essential for the development of brain, both IGFBP-3 and IGF-1 are elevated in the brains of children younger than 6 months of age. Likewise, IGFBP-3 is also upregulated following cerebral ischemia and hypoxia. However, the role of IGFBP-3 in neurogenesis is not clear. Using an in vitro culture system of rat neural progenitor cells, we demonstrate that IGFBP-3 specifically regulates the IGF-1 mediated neural progenitor cell proliferation via down regulation of phospho-Akt, and cyclin D1. In addition, IGFBP-3 also decreased the content of nestin in the neural progenitor cells indicating its potential role in neurogenesis.


Subject(s)
Cell Proliferation/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Animals , Cells, Cultured , Insulin-Like Growth Factor I/pharmacology , Neural Stem Cells/cytology , Rats
17.
Carcinogenesis ; 31(8): 1344-53, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20513670

ABSTRACT

Insulin-like growth factor-binding protein (IGFBP)-3 is overexpressed frequently in esophageal squamous cell carcinoma. Yet, the role of IGFBP3 in esophageal tumor biology remains to be elucidated. We find that IGFBP3 facilitates transforming growth factor (TGF)-beta1-mediated epithelial-to-mesenchymal transition (EMT) in transformed human esophageal epithelial cells, EPC2-hTERT-EGFR-p53(R175H). In organotypic 3D culture, a form of human tissue engineering, laser-capture microdissection revealed concurrent upregulation of TGF-beta target genes, IGFBP3 and EMT-related genes in the cells invading into the stromal compartment. IGFBP3 enhanced TGF-beta1-mediated EMT as well as transcription factors essential in EMT by allowing persistent SMAD2 and SMAD3 phosphorylation. TGF-beta1-mediated EMT and cell invasion were enhanced by ectopically expressed IGFBP3 and suppressed by RNA interference directed against IGFBP3. The IGFBP3 knockdown effect was rescued by IGFBP3(I56G/L80G/L81G), a mutant IGFBP3 lacking an insulin-like growth factor (IGF)-binding capacity. Thus, IGFBP3 can regulate TGF-beta1-mediated EMT and cell invasion in an IGF or insulin-like growth factor 1 receptor-independent manner. IGFBP3(I56G/L80G/L81G) also promoted EMT in vivo in a Ras-transformed human esophageal cell line T-TeRas upon xenograft transplantation in nude mice. In aggregate, IGFBP3 may have a novel IGF-binding independent biological function in regulation of TGF-beta1-mediated EMT and cell invasion.


Subject(s)
Epithelial Cells/cytology , Esophagus/cytology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Mesoderm/cytology , Transforming Growth Factor beta1/pharmacology , Animals , Carcinoma, Squamous Cell/pathology , Cell Movement/drug effects , Epithelial Cells/drug effects , Esophageal Neoplasms/pathology , Esophagus/drug effects , Esophagus/pathology , Gene Knockdown Techniques , Gene Transfer Techniques , Humans , Insulin-Like Growth Factor Binding Protein 3/deficiency , Insulin-Like Growth Factor Binding Protein 3/genetics , Lentivirus/genetics , Luciferases/genetics , Mesoderm/drug effects , Mice , Neoplasm Invasiveness , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
18.
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
19.
Growth Factors ; 27(6): 394-408, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19919528

ABSTRACT

High tissue insulin-like growth factor binding protein-3 (IGFBP-3) expression in breast cancers is associated in some studies with rapid growth and poor outcome. This study examined the effects of endogenous IGFBP-3 in Hs578T breast cancer cells, which are IGF-unresponsive and grow aggressively despite relatively high IGFBP-3 expression. IGFBP-3 downregulation using siRNA was associated with increases in DNA synthesis, the percentage of cells in S phase and viable cell numbers, accompanied by increases in cyclins A and D1, a decrease in p27 expression, and increased phosphorylation of retinoblastoma (Rb) on Ser795. Downregulation of IGFBP-3 inhibited extracellular signal-regulated kinase (ERK) activation and cell migration in a monolayer wound healing assay. These results indicate that endogenous IGFBP-3 is anti-proliferative and pro-migratory in Hs578T cells and that these effects are IGF-independent. Poor outcome in breast tumours expressing high levels of IGFBP-3 may be due to the effects of IGFBP-3 on cell migration rather than cell growth.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Movement/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , RNA Interference
20.
J Cell Biochem ; 103(2): 624-35, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17562544

ABSTRACT

We have reported that nordihydroguaiaretic acid (NDGA) inhibits the tyrosine kinase activities of the IGF-1 receptor (IGF-1R) and the HER2 receptor in breast cancer cells. Herein, we studied the effects of NDGA on the growth of estrogen receptor (ER) positive MCF-7 cells engineered to overexpress HER2 (MCF-7/HER2-18). These cells are an in vitro model of HER2-driven, ER positive, tamoxifen resistant breast cancer. NDGA was equally effective at inhibiting the growth of both parental MCF-7 and MCF-7/HER2-18 cells. Half maximal effects for both cell lines were in the 10-15 microM range. The growth inhibitory effects of NDGA were associated with an S phase arrest in the cell cycle and the induction of apoptosis. NDGA inhibited both IGF-1R and HER2 kinase activities in these breast cancer cells. In contrast, Gefitinib, an epidermal growth factor receptor inhibitor but not an IGF-1R inhibitor, was more effective in MCF-7/HER2-18 cells than in the parental MCF-7 cells and IGF binding protein-3 (IGFBP-3) was more effective against MCF-7 cells compared to MCF-7/HER2-18. MCF-7/HER2-18 cells are known to be resistant to the effects of the estrogen receptor inhibitor, tamoxifen. Interestingly, NDGA not only inhibited the growth of MCF-7/HER2-18 on its own, but it also demonstrated additive growth inhibitory effects when combined with tamoxifen. These studies suggest that NDGA may have therapeutic benefits in HER2-positive, tamoxifen resistant, breast cancers in humans.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Masoprocol/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Hormone-Dependent/pathology , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , Adenocarcinoma/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Drug Screening Assays, Antitumor , Drug Synergism , Female , Gefitinib , Humans , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Quinazolines/pharmacology , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL