Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
JCI Insight ; 9(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38258905

ABSTRACT

Pain of unknown etiology is frequent in individuals with the tumor predisposition syndrome neurofibromatosis 1 (NF1), even when tumors are absent. Nerve Schwann cells (SCs) were recently shown to play roles in nociceptive processing, and we find that chemogenetic activation of SCs is sufficient to induce afferent and behavioral mechanical hypersensitivity in wild-type mice. In mouse models, animals showed afferent and behavioral hypersensitivity when SCs, but not neurons, lacked Nf1. Importantly, hypersensitivity corresponded with SC-specific upregulation of mRNA encoding glial cell line-derived neurotrophic factor (GDNF), independently of the presence of tumors. Neuropathic pain-like behaviors in the NF1 mice were inhibited by either chemogenetic silencing of SC calcium or by systemic delivery of GDNF-targeting antibodies. Together, these findings suggest that alterations in SCs directly modulate mechanical pain and suggest cell-specific treatment strategies to ameliorate pain in individuals with NF1.


Subject(s)
Hypersensitivity , Neuralgia , Neurofibromatosis 1 , Animals , Mice , Neurofibromatosis 1/genetics , Nociception , Glial Cell Line-Derived Neurotrophic Factor/genetics , Schwann Cells
2.
bioRxiv ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38106002

ABSTRACT

Nerve growth factor (NGF) monoclonal antibodies (mAb) are one of the few patient-validated non-opioid treatments for chronic pain, despite failing to gain FDA approval due to worsened joint damage in some osteoarthritis patients. Herein, we demonstrate that neuropilin-1 (NRP1) is a nociceptor-enriched co-receptor for NGF that is necessary for tropomyosin-related kinase A (TrkA) signaling of pain. NGF binds NRP1 with nanomolar affinity. NRP1 and G Alpha Interacting Protein C-terminus 1 (GIPC1), a NRP1/TrkA adaptor, are coexpressed with TrkA in human and mouse nociceptors. NRP1 small molecule inhibitors and blocking mAb prevent NGF-stimulated action potential firing and activation of Na+ and Ca2+ channels in human and mouse nociceptors and abrogate NGF-evoked and inflammatory nociception in mice. NRP1 knockdown blunts NGF-stimulated TrkA phosphorylation, kinase signaling and transcription, whereas NRP1 overexpression enhances NGF and TrkA signaling. As well as interacting with NGF, NRP1 forms a heteromeric complex with TrkA. NRP1 thereby chaperones TrkA from the biosynthetic pathway to the plasma membrane and then to signaling endosomes, which enhances NGF-induced TrkA dimerization, endocytosis and signaling. Knockdown of GIPC1, a PDZ-binding protein that scaffolds NRP1 and TrkA to myosin VI, abrogates NGF-evoked excitation of nociceptors and pain-like behavior in mice. We identify NRP1 as a previously unrecognized co-receptor necessary for NGF/TrkA pain signaling by direct NGF binding and by chaperoning TrkA to the plasma membrane and signaling endosomes via the adaptor protein GIPC1. Antagonism of NRP1 and GIPC1 in nociceptors offers a long-awaited alternative to systemic sequestration of NGF with mAbs for the treatment of pain.

3.
J Diabetes Res ; 2014: 421827, 2014.
Article in English | MEDLINE | ID: mdl-25389530

ABSTRACT

This study determined if blocking ligand occupancy of the αVß3 integrin could inhibit the pathophysiologic changes that occur in the early stages of diabetic nephropathy (DN). Diabetic rats were treated with either vehicle or a monoclonal antibody that binds the ß3 subunit of the αVß3 integrin. After 4 weeks of diabetes the urinary albumin to creatinine ratio (UACR) increased in both diabetic animals that subsequently received vehicle and in the animals that subsequently received the anti-ß3 antibody compared with control nondiabetic rats. After 8 weeks of treatment the UACR continued to rise in the vehicle-treated rats; however it returned to levels comparable to control nondiabetic rats in rats treated with the anti-ß3 antibody. Treatment with the antibody prevented the increase of several profibrotic proteins that have been implicated in the development of DN. Diabetes was associated with an increase in phosphorylation of the ß3 subunit in kidney homogenates from diabetic animals, but this was prevented by the antibody treatment. This study demonstrates that, when administered after establishment of early pathophysiologic changes in renal function, the anti-ß3 antibody reversed the effects of diabetes normalizing albuminuria and profibrotic proteins in the kidney to the levels observed in nondiabetic control animals.


Subject(s)
Albuminuria/prevention & control , Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Integrin alphaVbeta3/antagonists & inhibitors , Kidney/drug effects , Albuminuria/diagnosis , Albuminuria/etiology , Albuminuria/urine , Animals , Biomarkers/urine , Collagen Type IV/metabolism , Creatinine/urine , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/etiology , Diabetic Nephropathies/urine , Fibrosis , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Kidney/metabolism , Kidney/pathology , Ligands , Male , Membrane Proteins/metabolism , Phosphorylation , Protein Binding , Rats, Sprague-Dawley , Streptozocin , Transforming Growth Factor beta/metabolism
4.
Endocrinology ; 155(12): 4665-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25171599

ABSTRACT

Hyperglycemia stimulates secretion of αVß3 ligands from vascular cells, including endothelial cells, resulting in activation of the αVß3 integrin. This study determined whether blocking ligand occupancy of αVß3 would inhibit the development of diabetic nephropathy. Ten diabetic pigs received an F(ab)2 fragment of an antibody directed against the extracellular domain of the ß3-subunit, and 10 received a control IgG F(ab)2 for 18 weeks. Nondiabetic pigs excreted 115 ± 50 µg of protein/mg creatinine compared with control F(ab)2-treated diabetic animals (218 ± 57 µg/mg), whereas diabetic animals treated with the anti-ß3 F(ab)2 excreted 119 ± 55 µg/mg (P < .05). Mesangial volume/glomerular volume increased to 21 ± 2.4% in control-treated diabetic animals compared with 14 ± 2.8% (P < .01) in animals treated with active antibody. Diabetic animals treated with control F(ab)2 had significantly less glomerular podocin staining compared with nondiabetic animals, and this decrease was attenuated by treatment with anti-ß3 F(ab)2. Glomerular basement membrane thickness was increased in the control, F(ab)2-treated diabetic animals (212 ± 14 nm) compared with nondiabetic animals (170 ± 8.8 nm), but it was unchanged (159.9 ± 16.4 nm) in animals receiving anti-ß3 F(ab)2. Podocyte foot process width was greater in control, F(ab)2-treated, animals (502 ± 34 nm) compared with animals treated with the anti-ß3 F(ab)2 (357 ± 47 nm, P < .05). Renal ß3 tyrosine phosphorylation decreased from 13 934 ± 6437 to 6730 ± 1524 (P < .01) scanning units in the anti-ß3-treated group. We conclude that administration of an antibody that inhibits activation of the ß3-subunit of αVß3 that is induced by hyperglycemia attenuates proteinuria and early histologic changes of diabetic nephropathy, suggesting that it may have utility in preventing the progression of this disease complication.


Subject(s)
Diabetic Nephropathies/etiology , Integrin alphaVbeta3/metabolism , Animals , Antibodies, Monoclonal , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Glomerular Basement Membrane/pathology , Integrin alphaVbeta3/antagonists & inhibitors , Male , Mice, Inbred BALB C , Podocytes/pathology , Proteinuria/etiology , Swine
5.
Int J Burns Trauma ; 3(4): 180-9, 2013.
Article in English | MEDLINE | ID: mdl-24273693

ABSTRACT

PURPOSE: Burn injury is associated with early apoptotic death of T cells. Insulin-like growth factor-1 (IGF-I) is able to protect T cells from apoptosis. Association of IGF-I with its IGFBP (Binding Protein)-1 limits its bioavailability and serine phosphorylation of IGFBP-1 lowers this further because of an increased affinity for IGF-I. The level of phosphorylated IGFBP-1 has been shown to increase in pediatric burn patients. Thus we hypothesized that a longitudinal study of burn patients would demonstrate 1) increased IGFBP-1 levels, 2) increased IGFBP-1 phosphorylation and 3) decreased IGF-I levels over time. METHODS: We conducted a prospective observational study in adult burn patients admitted to UNC Jaycee Burn Center. Plasma levels of insulin, insulin-like growth factor 1 (IGF-I) and insulin-like growth factor binding protein 1 (IGGBP-1) were measured on admission up to 10 days post admission. ELISA was used to measure serum levels of insulin, IGF-I and IGFBP-1. Serine phosphorylation of IGFBP-1 was measured by Western blot with and without the incubation of calf intestinal phosphatase (CIP). Significant findings: There was a significant positive correlation of increasing %TBSA burn and increasing levels of serum IGFBP-1 from admittance blood draws. Levels of IGF-I also decreased with increasing Total Body Surface Area (TBSA, p<0.05). In patients studied longitudinally (n=84) we found that IGFBP-1 levels are significantly (p<0.05) increased 1-72 hours post burn (mean±SEM serum concentration; burn=172±23 ng/mL, normal=13±3 ng/mL) and that levels of IGF-I are reduced. IGFBP-1 is serine phosphorylated in burn patients. In patients surviving past 72 hours IGFBP-1 remained phosphorylated over the study period. CONCLUSIONS: IGFBP-1 and its serine phosphorylation regulate and limit IGF-I bioavailability. Our results suggest that increases in IGFBP-1 and persistent serine phosphorylation of IGFBP-1 correlate with the severity of burn injury, and may contribute to burn-associated T cell apoptosis and subsequent immune dysfunction by reducing the bioavailability of this important cell survival factor.

6.
Endocrinology ; 154(11): 4146-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23981772

ABSTRACT

IGF-binding protein (IGFBP)-2 overexpression confers resistance to high-fat feeding and inhibits the differentiation of preadipocytes in vitro. However, whether administration of IGFBP-2 can regulate adipogenesis in vivo and the domains that mediate this response have not been defined. IGFBP-2 contains 2 heparin-binding domains (HBD), which are localized in the linker region (HBD1) and C-terminal region (HBD2) of IGFBP-2. To determine the relative importance of these domains, we used synthetic peptides as well as mutagenesis. Both HBD1 and HBD2 peptides inhibited preadipocyte differentiation, but the HBD2 peptide was more effective. Selective substitution of charged residues in the HBD1 or HBD2 regions attenuated the ability of the full-length protein to inhibit cell differentiation, but the HBD2 mutant had the greatest reduction. To determine their activities in vivo, pegylated forms of each peptide were administered to IGFBP-2(-/-) mice for 12 weeks. Magnetic resonance imaging scanning showed that only the HBD2 peptide significantly reduced (48 ± 9%, P < .05) gain in total fat mass. Both inguinal (32 ± 7%, P < .01) and visceral fat (44 ± 7%, P < .01) were significantly decreased by HBD2 whereas HBD1 reduced only visceral fat accumulation (24 ± 5%, P < .05). The HBD2 peptide was more effective peptide in reducing triglyceride content and serum adiponectin, but only the HBD2 peptide increased serum leptin. These findings demonstrate that the HBD2 domain of IGFBP-2 is the primary region that accounts for its ability to inhibit adipogenesis and that a peptide encompassing this region has activity that is comparable with native IGFBP-2.


Subject(s)
Adipocytes/cytology , Adipose Tissue/physiology , Heparin/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Adipocytes/metabolism , Animals , Cell Differentiation , Cells, Cultured , Epididymis/cytology , Insulin-Like Growth Factor Binding Protein 2/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Protein Structure, Tertiary
7.
Mol Cell Biol ; 32(20): 4116-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22869525

ABSTRACT

Insulin-like growth factor I (IGF-I) is a mitogen for vascular smooth muscle cells (VSMC) and has been implicated in the development and progression of atherosclerosis. IGF binding proteins (IGFBPs) modify IGF-I actions independently of IGF binding, but a receptor-based mechanism by which they function has not been elucidated. We investigated the role of IGFBP-2 and receptor protein tyrosine phosphatase ß (RPTPß) in regulating IGF-I signaling and cellular proliferation. IGFBP-2 bound RPTPß, which led to its dimerization and inactivation. This enhanced PTEN tyrosine phosphorylation and inhibited PTEN activity. Utilization of substrate trapping and phosphatase-dead mutants showed that RPTPß bound specifically to PTEN and dephosphorylated it. IGFBP-2 knockdown led to decreased PTEN tyrosine phosphorylation and decreased AKT Ser473 activation. IGFBP-2 enhanced IGF-I-stimulated VSMC migration and proliferation. Analysis of aortas obtained from IGFBP-2(-/-) mice showed that RPTPß was activated, and this was associated with inhibition of IGF-I stimulated AKT Ser473 phosphorylation and VSMC proliferation. These changes were rescued following administration of IGFBP-2. These findings present a novel mechanism for coordinate regulation of IGFBP-2 and IGF-I signaling functions that lead to stimulation of VSMC proliferation. The results have important implications for understanding how IGFBPs modulate the cellular response to IGF-I.


Subject(s)
Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Signal Transduction/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor I/pharmacology , Mice , Muscle, Smooth, Vascular/drug effects , PTEN Phosphohydrolase/metabolism , Serine/metabolism
8.
Diabetes ; 61(1): 104-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22148072

ABSTRACT

IGF-I-stimulated sarcoma viral oncogene (Src) activation during hyperglycemia is required for propagating downstream signaling. The aim of the current study was to determine the mechanism by which hyperglycemia enhances IGF-I-stimulated Src activation and the role of NADPH oxidase 4 (Nox4) and protein kinase C ζ (PKCζ) in mediating this response in vascular smooth muscle cells (VSMCs). Nox4 expression was analyzed in VSMCs exposed to hyperglycemia. The role of Nox4-derived reactive oxygen species (ROS) in IGF-I-stimulated Src activation was investigated via knockdown of Nox4. Different isoforms of PKC were screened to investigate their role in hyperglycemia-induced Nox4. The oxidation of Src was shown to be a prerequisite for its activation in response to IGF-I during hyperglycemia. Hyperglycemia induced Nox4, but not Nox1, and p22 phagocyte oxidase (p22phox) expression and IGF-I stimulated Nox4/p22phox complex formation, leading to increased ROS generation. Knockdown of Nox4 prevented ROS generation and impaired the oxidation and activation of Src in response to IGF-I, whereas knockdown of Nox1 had no effect. PKCζ was shown to mediate the hyperglycemia-induced increase in Nox4 expression. The key observations in cultured VSMCs were confirmed in the diabetic mice. Nox4-derived ROS is responsible for the enhancing effect of hyperglycemia on IGF-I-stimulated Src activation, which in turn amplifies IGF-I-linked downstream signaling and biological actions.


Subject(s)
Hyperglycemia/metabolism , Insulin-Like Growth Factor I/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NADPH Oxidases/metabolism , Protein Kinase C/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , NADPH Oxidase 4 , Oxidation-Reduction/drug effects , Protein Kinase C/metabolism
9.
J Bone Miner Res ; 27(2): 390-400, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22006816

ABSTRACT

Global deletion of the Igfbp2 gene results in the suppression of bone turnover. To investigate the role of insulin-like growth factor-binding protein-2 (IGFBP-2) in regulating osteoclast differentiation, we cultured Igfbp2(-/-) bone marrow cells and found a reduction in the number of osteoclasts and impaired resorption. Addition of full-length IGFBP-2 restored osteoclast differentiation, fusion, and resorption. To determine the molecular domains of IGFBP-2 that were required for this effect to be manifest, Igfbp2(-/-) bone marrow cells were transfected with constructs in which the heparin-binding (HBD) or the IGF-binding domains of IGFBP-2 were mutated. We found that both domains were necessary for osteoclastogenesis because expression of the mutated forms of either domain failed to support the formation of functionally mature osteoclasts. To discern the mechanism by which IGFBP-2 regulates osteoclast formation, PTEN abundance and phosphorylation status as well as AKT responsiveness to IGF-I were analyzed. Igfbp2(-/-) cells had elevated levels of PTEN and phospho-PTEN compared with controls. Expression of wild-type IGFBP-2 reduced the level of PTEN to that of wild-type cells. Cells expressing the IGF-binding mutant showed suppression of PTEN and phospho-PTEN equivalent to the wild-type protein, whereas those expressing the IGFBP-2 HBD mutant showed no PTEN suppression. When the ability of IGF-I to stimulate AKT activation, measured by Thr³°8 and Ser47³ phosphorylation, was analyzed, stimulation of Ser47³ in response to IGF-I in preosteoclasts required the presence of intact IGFBP-2. This effect was duplicated by the addition of a CK2 inhibitor that prevents the phosphorylation of PTEN. In contrast, in fully differentiated osteoclasts, stimulation of Thr³°8 phosphorylation required the presence of intact IGFBP-2. We conclude that IGFBP-2 is an important regulator of osteoclastogenesis and that both the heparin- and the IGF-binding domains of IGFBP-2 are essential for the formation of fully differentiated and functional osteoclasts.


Subject(s)
Cell Differentiation , Insulin-Like Growth Factor Binding Protein 2/metabolism , Osteoclasts/pathology , Acid Phosphatase/metabolism , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Female , Humans , Insulin-Like Growth Factor Binding Protein 2/chemistry , Insulin-Like Growth Factor I/pharmacology , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Mutant Proteins/metabolism , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteogenesis/drug effects , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/metabolism , Tartrate-Resistant Acid Phosphatase
10.
Mol Endocrinol ; 25(9): 1636-49, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21799000

ABSTRACT

IGF-I plays an important role in smooth muscle cell proliferation and migration. In vascular smooth muscle cells cultured in 25 mm glucose, IGF-I stimulated a significant increase in Src homology 2 domain containing protein tyrosine phosphatase substrate-1 (SHPS-1) phosphorylation compared with 5 mm glucose and this increase was required for smooth muscle cell proliferation. A proteome-wide screen revealed that carboxyl-terminal SRC kinase homologous kinase (CTK) bound directly to phosphotyrosines in the SHPS-1 cytoplasmic domain. Because the kinase(s) that phosphorylates these tyrosines in response to IGF-I is unknown, we determined the roles of IGF-I receptor (IGF-IR) and CTK in mediating SHPS-1 phosphorylation. After IGF-I stimulation, CTK was recruited to IGF-IR and subsequently to phospho-SHPS-1. Expression of an IGF-IR mutant that eliminated CTK binding reduced CTK transfer to SHPS-1, SHPS-1 phosphorylation, and cell proliferation. IGF-IR phosphorylated SHPS-1, which provided a binding site for CTK. CTK recruitment to SHPS-1 resulted in a further enhancement of SHPS-1 phosphorylation. CTK knockdown also impaired IGF-I-stimulated SHPS-1 phosphorylation and downstream signaling. Analysis of specific tyrosines showed that mutation of tyrosines 428/452 in SHPS-1 to phenylalanine reduced SHPS-1 phosphorylation but allowed CTK binding. In contrast, the mutation of tyrosines 469/495 inhibited IGF-IR-mediated the phosphorylation of SHPS-1 and CTK binding, suggesting that IGF-IR phosphorylated Y469/495, allowing CTK binding, and that CTK subsequently phosphorylated Y428/452. Based on the above findings, we conclude that after IGF-I stimulation, CTK is recruited to IGF-IR and its recruitment facilitates CTK's subsequent association with phospho-SHPS-1. This results in the enhanced CTK transfer to SHPS-1, and the two kinases then fully phosphorylate SHPS-1, which is necessary for IGF-I stimulated cellular proliferation.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/enzymology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Immunologic/metabolism , Amino Acid Substitution/genetics , Animals , Gene Silencing/drug effects , Glucose/pharmacology , Humans , Mice , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Rats , Sus scrofa
11.
J Bone Miner Res ; 26(9): 2068-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21638321

ABSTRACT

Integrin-associated protein (IAP/CD47) has been implicated in macrophage-macrophage fusion. To understand the actions of CD47 on skeletal remodeling, we compared Cd47(-/-) mice with Cd47(+/+) controls. Cd47(-/-) mice weighed less and had decreased areal bone mineral density compared with controls. Cd47(-/-) femurs were shorter in length with thinner cortices and exhibited lower trabecular bone volume owing to decreased trabecular number and thickness. Histomorphometry revealed reduced bone-formation and mineral apposition rates, accompanied by decreased osteoblast numbers. No differences in osteoclast number were observed despite a nonsignificant but 40% decrease in eroded surface/bone surface in Cd47(-/-) mice. In vitro, the number of functional osteoclasts formed by differentiating Cd47(-/-) bone marrow cells was significantly decreased compared with wild-type cultures and was associated with a decrease in bone-resorption capacity. Furthermore, by disrupting the CD47-SHPS-1 association, we found that osteoclastogenesis was markedly impaired. Assays for markers of osteoclast maturation suggested that the defect was at the point of fusion and not differentiation and was associated with a lack of SHPS-1 phosphorylation, SHP-1 phosphatase recruitment, and subsequent dephosphorylation of non-muscle cell myosin IIA. We also demonstrated a significant decrease in osteoblastogenesis in bone marrow stromal cells derived from Cd47(-/-) mice. Our finding of cell-autonomous defects in Cd47(-/-) osteoblast and osteoclast differentiation coupled with the pronounced skeletal phenotype of Cd47(-/-) mice support the conclusion that CD47 plays an important role in regulating skeletal acquisition and maintenance through its actions on both bone formation and bone resorption.


Subject(s)
Bone Remodeling , CD47 Antigen/metabolism , Receptors, Immunologic/metabolism , Aging/blood , Aging/drug effects , Animals , Biomarkers/blood , Body Composition/drug effects , Bone Remodeling/drug effects , Cell Differentiation/drug effects , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Humans , Male , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Phenotype , Protein Binding/drug effects , RANK Ligand/pharmacology , Tomography, X-Ray Computed
12.
Curr Diabetes Rev ; 7(4): 235-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21707534

ABSTRACT

IGF-I is structurally related to proinsulin and when administered to human subjects it enhances insulin sensitivity. However because of its growth promoting properties and its relationship to growth hormone, it has been proposed as a etiologic factor in the development of diabetic complications. This review discusses recently published data regarding the ability of hyperglycemia to sensitize cells that are capable of dedifferentiating to the growth promoting effects of IGF-I. Under normoglycemic conditions vascular smooth muscle and endothelial cells are cystostatic and stimulation of the IGF-I receptor activates the adaptor protein IRS-1 which leads to PI-3 kinase pathway activation. Following exposure to hyperglycemia these cell types undergo a signaling switch whereby an entirely different mechanism is utilized to activate both the PI-3 kinase and the MAP pathways. This leads to increased cell proliferation and migration. This molecular mechanism involves the coordinate regulation of signaling molecules and scaffolding proteins. Activation of this alternative signaling mechanism is directly linked to the stimulation of pathophysiologic processes that are involved in the pathogenesis of both diabetic retinopathy and atherosclerosis. Inhibition of activation of these intermediates has been shown to attenuate glucose induced pathophysiologic changes and results in the inhibition of both atherosclerotic lesion progression and diabetic retinopathy. In summary, hyperglycemia induces a signaling switch in vascular endothelial and smooth muscle cells that results in enhanced sensitivity to the growth promoting effects of IGF-I. This may be an important variable for determining the progression of atherosclerosis in poorly controlled diabetes and in the development of retinopathy.


Subject(s)
Diabetes Complications/etiology , Hyperglycemia/complications , Hyperglycemia/metabolism , Insulin-Like Growth Factor I/physiology , Animals , Cell Proliferation/drug effects , Diabetes Complications/genetics , Diabetes Complications/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Hyperglycemia/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Models, Biological , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
13.
Endocrinology ; 151(12): 5851-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20881255

ABSTRACT

The IGF-I pathway and renin-angiotensin-aldosterone axis are both involved in the pathogenesis of hypertension and atherosclerosis, but no information is available about IGF-I and aldosterone interaction or their potential synergistic effects in vascular smooth muscle cells (VSMCs). The aims of this study were to investigate whether aldosterone influences IGF-I signaling and to determine the mechanism(s) by which aldosterone affects IGF-I function. Aldosterone resulted in significant increases in the Akt (1.87 ± 0.24, P < 0.001), MAPK (1.78 ± 0.13, P < 0.001), p70S6kinase (1.92 ± 0.15, P < 0.001), IGF-I receptor (1.69 ± 0.05, P < 0.01), and insulin receptor substrate-1 (1.7 ± 0.04, P < 0.01) (fold increase, mean ± SEM, n = 3) phosphorylation responses to IGF-I compared with IGF-I treatment alone. There were also significant increases in VSMC proliferation, migration, and protein synthesis (1.63 ± 0.03-, 1.56 ± 0.08-, and 1.51 ± 0.04-fold increases compared with IGF-I alone, respectively, n = 3, P < 0.001). Aldosterone induced osteopontin (OPN) mRNA expression and activation of αVß3-integrin as well as an increase in the synthesis of IGF-I receptor. The enhancing effects of aldosterone were inhibited by eplerenone (10 µmol/liter), actinomycin-D (20 nmol/liter), and an anti-αVß3-integrin antibody that blocks OPN binding. The antioxidant N-acetylcysteine (2 mmol/liter) completely inhibited the ability of aldosterone to induce any of these changes. In conclusion, our results show that aldosterone enhances IGF-I signaling and biological actions in VSMCs through induction of OPN followed by its subsequent activation of the αVß3-integrin and by increasing IGF-I receptor. These changes are mediated in part through increased oxidative stress. The findings suggest a new mechanism by which aldosterone could accelerate the development of atherosclerosis.


Subject(s)
Aldosterone/pharmacology , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Signal Transduction/physiology , Animals , Aorta/cytology , Cell Movement , Cell Proliferation , Cells, Cultured , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Swine
14.
Exp Diabetes Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20689700

ABSTRACT

Smooth muscle cells (SMC) maintained in high glucose are more responsive to IGF-I than those in normal glucose. There is significantly more thrombospondin-1 (TSP-1) in extracellular matrix surrounding SMC grown in 25 mM glucose. In this study we investigated 1) the mechanism by which glucose regulates TSP-1 levels and 2) the mechanism by which TS-1 enhances IGF-I signaling. The addition of TSP-1 to primary SMC was sufficient to enhance IGF-I responsiveness in normal glucose. Reducing TSP-1 protein levels inhibited IGF-I signaling in SMC maintained in high glucose. We determined that TSP-1 protected IAP/CD47 from cleavage and thereby facilitated its association with SHP substrate-1 (SHPS-1). We have shown previously that the hyperglycemia induced protection of IAP from cleavage is an important component of the ability of hyperglycemia to enhance IGF-I signaling. Furthermore we determined that TSP-1 also enhanced phosphorylation of the beta3 subunit of the alphaVbeta3 integrin, another molecular event that we have shown are critical for SMC response to IGF-I in high glucose. Our studies also revealed that the difference in the amount of TSP-1 in the two different glucose conditions was due, at least in part, to a difference in the cellular uptake and degradation of TSP-1.


Subject(s)
Cell Proliferation , Glucose/metabolism , Hyperglycemia/metabolism , Insulin-Like Growth Factor I/metabolism , Myocytes, Smooth Muscle/physiology , Thrombospondin 1/metabolism , Animals , CD47 Antigen/metabolism , Glucose/pharmacology , Integrin alphaVbeta3/metabolism , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Swine , Thrombospondin 1/pharmacology
15.
Endocrinology ; 151(8): 3611-23, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20534722

ABSTRACT

Hyperglycemia has been shown to induce the p66shc expression leading to increased reactive oxygen species (ROS) generation and apoptosis. In the present study, we demonstrated that hyperglycemia induced p66shc expression in vascular smooth muscle cells. This induction was associated with an increase in apoptosis as assessed by the increase of capspase-3 enzymatic activity, cleaved caspase-3 protein, and the number of dead cells. The ability of IGF-I to inhibit apoptosis was also attenuated. Further studies showed that hyperglycemia-induced p66shc inhibited IGF-I-stimulated phosphoinositide (PI)-3 kinase and AKT activation. Mechanistic studies showed that knockdown of p66shc enhanced IGF-I-stimulated SHPS-1/p85, p85/SHP-2, and p85/Grb2 association, all of which are required for PI-3 kinase/AKT activation. These responses were attenuated by overexpression of p66shc. IGF-I-stimulated p85 and AKT recruitment to the cell membrane fraction was altered in the same manner. Disruption of p66shc-Src interaction using either a blocking peptide or by expressing a p66shc mutant that did not bind to Src rescued IGF-I-stimulated PI-3 kinase/AKT activation as well as IGF-I-dependent cell survival. Although the highest absolute level of ROS was detected in p66shc-overexpressing cells, the relative increase in ROS induced by hyperglycemia was independent of p66shc expression. Taken together, our data suggest that the increase in p66shc that occurs in response to hyperglycemia is functioning to inhibit IGF-I-stimulated signaling and that the incremental increase in SMC sensitivity to IGF-I stimulation that occurs in response to p66shc induction of ROS is not sufficient to overcome the inhibitory effect of p66shc on Src kinase activation.


Subject(s)
Hyperglycemia/metabolism , Insulin-Like Growth Factor I/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Shc Signaling Adaptor Proteins/physiology , src-Family Kinases/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Down-Regulation , Enzyme Activation/physiology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Humans , Hyperglycemia/pathology , Insulin-Like Growth Factor I/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , RNA, Small Interfering/pharmacology , Shc Signaling Adaptor Proteins/antagonists & inhibitors , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Swine , src-Family Kinases/physiology
16.
Endocrinology ; 151(8): 3752-63, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20555032

ABSTRACT

Obesity morbidity is associated with excess visceral adiposity, whereas sc adipose tissue is much less metabolically hazardous. Human abdominal sc preadipocytes have greater capacity for proliferation, differentiation, and survival than omental preadipocytes. IGF-I is a critical mediator of preadipocyte proliferation, differentiation, and survival through multiple signaling pathways. We investigated IGF-I action in primary cultures of human preadipocytes isolated from sc and omental adipose tissue of obese subjects. IGF-I-stimulated DNA synthesis was significantly lower in omental compared with sc preadipocytes. IGF-I phosphorylation of the IGF-I receptor and the ERK pathway was comparable in sc and omental cells. However, omental preadipocytes had decreased insulin receptor substrate (IRS)-1 protein associated with increased IRS-1-serine(636/639) phosphorylation and degradation. IGF-I-stimulated phosphorylation of AKT on serine(473) but not threonine(308) was decreased in omental cells, and activation of downstream targets, including S6Kinase, glycogen synthase kinase-3, and Forkhead box O1 was also impaired. CyclinD1 abundance was decreased in omental cells due to increased degradation. Over-expression of IRS-1 by lentivirus in omental preadipocytes increased IGF-I-stimulated AKT-serine(473) phosphorylation. The mammalian target of rapamycin (mTOR)-Rictor complex regulates phosphorylation of AKT-serine(473) in 3T3-L1 adipocytes, but knockdown of Rictor by lentivirus-delivered short hairpin RNA in sc preadipocytes did not affect AKT-serine(473) phosphorylation by IGF-I. These data reveal an intrinsic defect in IGF-I activation of the AKT pathway in omental preadipocytes from obese subjects that involves IRS-1 but probably not mTOR-Rictor complex. We conclude that impaired cell cycle regulation by AKT contributes to the distinct growth phenotype of preadipocytes in visceral fat of obese subjects.


Subject(s)
Adipocytes, White/drug effects , Insulin-Like Growth Factor I/pharmacology , Intra-Abdominal Fat/pathology , Obesity/pathology , Proto-Oncogene Proteins c-akt/metabolism , Subcutaneous Fat/pathology , 3T3-L1 Cells , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adipocytes, White/physiology , Adult , Animals , Cell Culture Techniques , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Down-Regulation/drug effects , Enzyme Activation/drug effects , Female , Humans , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Male , Mice , Middle Aged , Obesity/metabolism , Signal Transduction/drug effects , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism
17.
Sci Transl Med ; 2(18): 18ra11, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20371482

ABSTRACT

Atherosclerotic lesions develop and progress more rapidly in diabetic patients than in nondiabetic individuals. This may be caused by accelerated lesion formation in the high-glucose environment of diabetes. Smooth muscle cells (SMCs) cultured in high glucose are more responsive to growth factors such as insulin-like growth factor-1 (IGF-1). This enhanced response to IGF-1 is due in part to increased activation of the alpha(V)beta(3) integrin. We tested whether alpha(V)beta(3) integrin activation was increased in diabetic animals and whether an antibody to beta(3) would inhibit IGF-1 action and development of atherosclerosis. Eight male pigs were made diabetic with streptozotocin and fed a high-fat diet. A F(ab)(2) antibody fragment directed at beta(3) was infused into one femoral artery, whereas the other artery received control F(ab)(2) for 3.5 months. There was a 65 +/- 8% reduction in atherosclerotic lesion area in the arteries treated with F(ab)(2) antibody to beta(3). Phosphorylation of beta(3) was reduced by 75 +/- 18% in vessels treated with the antibody. Shc and mitogen-activated protein kinase phosphorylation, which are required for IGF-1-stimulated SMC proliferation, were also significantly reduced. We conclude that activation of IGF-1 receptor and alpha(V)beta(3)-linked signaling pathways accelerates atherosclerosis in diabetes and that administration of an antibody to beta(3) to diabetic pigs inhibits alpha(V)beta(3) activation, IGF-1-stimulated signaling, and atherosclerotic lesion development. This approach offers a potential therapeutic approach to the treatment of this disorder.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Atherosclerosis/complications , Atherosclerosis/prevention & control , Diabetes Mellitus, Experimental/complications , Integrin alphaVbeta3/immunology , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Femoral Artery/metabolism , Femoral Artery/pathology , Immunoglobulin Fab Fragments , Insulin-Like Growth Factor I/metabolism , Male , Mice , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Signal Transduction , Swine , Time Factors
18.
J Biol Chem ; 285(21): 15682-95, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20207740

ABSTRACT

Vascular smooth muscle cells maintained in normal (5.6 mm) glucose respond to insulin-like growth factor-I (IGF-I) with increased protein synthesis but do not proliferate. In contrast, hyperglycemia alters responsiveness to IGF-I, resulting in increased SHPS-1 phosphorylation and assembly of a signaling complex that enhances MAPK and phosphatidylinositol 3-kinase pathways. Hyperglycemia also reduces the basal IRS-1 concentration and IGF-I-stimulated IRS-1-linked signaling. To determine if failure to down-regulate IRS-1 alters vascular smooth muscle cell (VSMC) responses to IGF-I, we overexpressed IRS-1 in VSMCs maintained in high glucose. These cultures showed reduced SHPS-1 phosphorylation, transfer of SHP-2 to SHPS-1, and impaired Shc and MAPK phosphorylation and cell proliferation in response to IGF-I. In vitro studies demonstrated that SHPS-1 was a substrate for type I IGF receptor (IGF-IR) and that IRS-1 competitively inhibited SHPS-1 phosphorylation. Exposure of VSMC cultures to a peptide that inhibited IRS-1/IGF-IR interaction showed that IRS-1 binding to IGF-IR impairs SHPS-1 phosphorylation in vivo. IRS-1 also sequestered SHP-2. Expression of an IRS-1 mutant (Y1179F/Y1229F) reduced IRS-1/SHP-2 association, and exposure of cells expressing the mutant to the inhibitory peptide enhanced SHPS-1 phosphorylation and SHP-2 transfer. This result was confirmed by expressing an IRS-1 mutant that had both impaired binding to IGF-IR and to SHP-2 IGF-I increased SHPS-1 phosphorylation, SHP-2 association with SHPS-1, Shc MAPK phosphorylation, and proliferation in cells expressing the mutant. We conclude that IRS-1 is an important factor for maintaining VSMCs in the non-proliferative state and that its down-regulation is a component of the VSMC response to hyperglycemic stress that results in an enhanced response to IGF-I.


Subject(s)
Antigens, Differentiation/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Immunologic/metabolism , Animals , Antigens, Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Glucose/pharmacology , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin-Like Growth Factor I/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mutation , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptors, Immunologic/genetics , Shc Signaling Adaptor Proteins , Stress, Physiological/drug effects , Stress, Physiological/physiology , Sweetening Agents/pharmacology , Swine
19.
Endocrinology ; 150(3): 1321-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18974270

ABSTRACT

Prior published reports have demonstrated that glucose-oxidized low-density lipoproteins (g-OxLDL) enhance the proliferative response of vascular smooth muscle cells (SMC) to IGF-I. Our previous studies have determined that the regulation of cleavage of integrin-associated protein (IAP) by matrix-metalloprotease-2 (MMP-2) in diabetic mice in response to hyperglycemia is a key regulator of the response of SMC to IGF-I. Because chronic hyperglycemia enhances glucose-induced LDL oxidation, these studies were conducted to determine whether g-OxLDL modulates the response of SMC to IGF-I by regulating MMP-2-mediated cleavage of IAP. We determined that exposure of SMC to g-OxLDL, but not native LDL, was sufficient to facilitate an increase in cell proliferation in response to IGF-I. Exposure to an anti-CD36 antibody, which has been shown to inhibit g-OxLDL-mediated signaling, inhibited the effects of g-OxLDL on IGF-I-stimulated SMC proliferation. The effect of g-OxLDL could be attributed, in part, to an associated decrease in proteolytic cleavage of IAP leading to increase in the basal association between IAP and Src homology 2 domain-containing protein tyrosine phosphatase substrate-1, which is required for IGF-I-stimulated proliferation. The inhibitory effect of g-OxLDL on IAP cleavage appeared to be due to its ability to decrease the amount of activated MMP-2, the protease responsible for IAP cleavage. In conclusion, these data provide a molecular mechanism to explain previous studies that have reported an enhancing effect of g-OxLDL on IGF-I-stimulated SMC proliferation.


Subject(s)
CD47 Antigen/metabolism , Cell Proliferation/drug effects , Insulin-Like Growth Factor I/pharmacology , Lipoproteins, LDL/pharmacology , Myocytes, Smooth Muscle/drug effects , Protein Processing, Post-Translational/drug effects , Animals , CD36 Antigens/physiology , Cells, Cultured , Down-Regulation/drug effects , Glucose/pharmacology , Matrix Metalloproteinase 2/metabolism , Myocytes, Smooth Muscle/physiology , Phosphorylation , Protein Binding/drug effects , Receptors, Immunologic/metabolism , Swine
20.
Exp Diabetes Res ; 2009: 267107, 2009.
Article in English | MEDLINE | ID: mdl-20111736

ABSTRACT

Increased responsiveness of vascular cells to the growth factor IGF-I has been implicated in complications associated with diabetes. Here we describe the development of an assay and screening of a library of compounds for their ability to accelerate cleavage of the transmembrane protein integrin-associated protein (IAP) thereby disrupting the association between IAP and SHPS-1 which we have shown as critical for the enhanced response of vascular cells to IGF-I. The cell-based ELISA utilizes an antibody that specifically detects cleaved, but not intact, IAP. Of the 1040 compounds tested, 14 were considered active by virtue of their ability to stimulate an increase in antibody-binding indicative of IAP cleavage. In experiments with smooth muscle and retinal endothelial cell cultures in hyperglycemic conditions, each active compound was shown to accelerate the cleavage of IAP, and this was associated with a decrease in IAP association with SHPS-1 as determined by coimmunoprecipitation of the proteins from cell lysates. As a consequence of the acceleration in IAP cleavage, the compounds were shown to inhibit IGF-I-stimulated phosphorylation of key signaling molecules including Shc and ERK1/2, and this in turn was associated with a decrease in IGF-I-stimulated cell proliferation. Identification of these compounds that utilize this mechanism has the potential to yield novel therapeutic approaches for the prevention and treatment of vascular complications associated with diabetes.


Subject(s)
Hyperglycemia/physiopathology , Insulin-Like Growth Factor I/antagonists & inhibitors , Signal Transduction/physiology , Animals , CD47 Antigen/physiology , Cattle , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Glucose/pharmacology , Insulin-Like Growth Factor I/physiology , Signal Transduction/drug effects , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...