Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 110: 261-269, 2017 09.
Article in English | MEDLINE | ID: mdl-28666850

ABSTRACT

Previously published studies strongly suggested that insulin- and exercise-induced skeletal muscle glucose uptake require nitric oxide (NO) production. However, the signal transduction mechanisms by which insulin and contraction regulated NO production and subsequent glucose transport are not known. In the present study, we utilized the myotube cell lines treated with insulin or hydrogen peroxide, the latter to mimic contraction-induced oxidative stress, to characterize these mechanisms. We found that insulin stimulation of neuronal nitric oxide synthase (nNOS) phosphorylation, NO production, and GLUT4 translocation were all significantly reduced by inhibition of either nNOS or Akt2. Hydrogen peroxide (H2O2) induced phosphorylation of nNOS at the same residue as did insulin, and also stimulated NO production and GLUT4 translocation. nNOS inhibition prevented H2O2-induced GLUT4 translocation. AMP activated protein kinase (AMPK) inhibition prevented H2O2 activation and phosphorylation of nNOS, leading to reduced NO production and significantly attenuated GLUT4 translocation. We conclude that nNOS phosphorylation and subsequently increased NO production are required for both insulin- and H2O2-stimulated glucose transport. Although the two stimuli result in phosphorylation of the same residue on nNOS, they do so through distinct protein kinases. Thus, insulin and H2O2-activated signaling pathways converge on nNOS, which is a common mediator of glucose uptake in both pathways. However, the fact that different kinases are utilized provides a basis for the use of exercise to activate glucose transport in the face of insulin resistance.


Subject(s)
Glucose/metabolism , Hydrogen Peroxide/pharmacology , Insulin/pharmacology , Muscle Fibers, Skeletal/drug effects , Nitric Oxide Synthase Type I/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin Resistance , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
2.
Biochem Biophys Res Commun ; 486(4): 937-944, 2017 05 13.
Article in English | MEDLINE | ID: mdl-28351623

ABSTRACT

Individuals born small for gestational age (SGA) are at a higher risk of developing the metabolic syndrome later in life. IGF-1 resistance has been reported in placentae from SGA births and mutations in the Igf1 receptor gene have been reported in several cohorts of SGA subjects. We have used the Igf1r heterozygous (Igf1r+/-) male mouse as a model to investigate the mechanisms by which Igf1r haploinsufficiency leads to insulin resistance. Despite exhibiting IGF-1 resistance, insulin signaling is enhanced in young Igf1r+/- mice but is attenuated in the muscle of old Igf1r+/- mice. Although smaller than WT (wild type) mice, old-aged Igf1r+/- had increased adiposity and exhibit increased lipogenesis. We hypothesize that IGF-1 resistance initially causes a transient increase in insulin signaling thereby promoting a lipogenic phenotype, which subsequently leads to insulin resistance.


Subject(s)
Aging/genetics , Haploinsufficiency/genetics , Infant, Small for Gestational Age , Insulin Resistance/genetics , Metabolic Syndrome/genetics , Receptor, IGF Type 1/genetics , Animals , Female , Genetic Predisposition to Disease/genetics , Humans , Infant, Newborn , Male , Mice , Mice, Inbred C57BL
3.
Endocrinology ; 156(8): 2872-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26076041

ABSTRACT

In the current study, we determined the effects of IGF-1 receptor haploinsufficiency on osteoblast differentiation and bone formation throughout the lifespan. Bone mineral density was significantly decreased in femurs of male and female Igf1r(+/-) mice compared with wild-type mice. mRNA expression of osteoblast differentiation markers was significantly decreased in femurs and calvariae from Igf1r(+/-) mice compared with cells from wild-type mice. Bone morphogenetic protein-7-induced ectopic bone in Igf1r(+/-) mice was significantly smaller with fewer osteoblasts but more lipid droplets and had reduced expression of osteoblast differentiation markers compared with wild-type mice. In bone marrow cells from middle-aged and old wild-type and Igf1r(+/-) male mice, palmitate inhibited osteoblast markers expression. In cells from young wild-type male mice, palmitate did not inhibit marker expression, but in cells from young male Igf1r(+/-) mice, palmitate inhibited bone sialoprotein and osterix but not osteocalcin or type I collagen (TIC). In female wild-type mice, palmitate inhibited osteoblast markers expression in cells from young, middle-aged, and old mice except TIC in cells from middle-aged mice. Palmitate inhibited bone sialoprotein expression in cells from middle-aged and old female Igf1r(+/-) mice and osteocalcin, osterix, and TIC expression in young and middle-aged female Igf1r(+/-) mice but stimulated expression in cells from old female Igf1r(+/-) mice. We conclude that IGF-1 receptor haploinsufficiency results in a prolipid accrual phenotype in bone in association with inhibition of growth factor-induced osteoblast differentiation, a situation which may phenocopy age-related decreases in bone formation.


Subject(s)
Aging/physiology , Cell Differentiation/genetics , Haploinsufficiency , Osteoblasts/physiology , Receptor, IGF Type 1/genetics , Aging/genetics , Animals , Bone Density/genetics , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteogenesis/genetics
4.
Biochem Biophys Res Commun ; 450(1): 777-81, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24955854

ABSTRACT

Aging is associated with the accumulation of ectopic lipid resulting in the inhibition of normal organ function, a phenomenon known as lipotoxicity. Within the bone marrow microenvironment, elevation in fatty acid levels may produce an increase in osteoclast activity and a decrease in osteoblast number and function, thus contributing to age-related osteoporosis. However, little is known about lipotoxic mechanisms in intramembraneous bone. Previously we reported that the long chain saturated fatty acid palmitate inhibited the expression of the osteogenic markers RUNX2 and osteocalcin in fetal rat calvarial cell (FRC) cultures. Moreover, the acetyl CoA carboxylase inhibitor TOFA blocked the inhibitory effect of palmitate on expression of these two markers. In the current study we have extended these observations to show that palmitate inhibits spontaneous mineralized bone formation in FRC cultures in association with reduced mRNA expression of RUNX2, alkaline phosphatase, osteocalcin, and bone sialoprotein and reduced alkaline phosphatase activity. The effects of palmitate on osteogenic marker expression were inhibited by TOFA. Palmitate also inhibited the mRNA expression of fatty acid synthase and PPARγ in FRC cultures, and as with osteogenic markers, this effect was inhibited by TOFA. Palmitate had no effect on FRC cell proliferation or apoptosis, but inhibited BMP-7-induced alkaline phosphatase activity. We conclude that palmitate accumulation may lead to lipotoxic effects on osteoblast differentiation and mineralization and that increases in fatty acid oxidation may help to prevent these lipotoxic effects.


Subject(s)
Embryonic Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis/physiology , Palmitates/administration & dosage , Skull/cytology , Skull/embryology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/drug effects , Rats , Skull/drug effects
5.
Aging Cell ; 13(3): 408-18, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24341939

ABSTRACT

In lower or simple species, such as worms and flies, disruption of the insulin-like growth factor (IGF)-1 and the insulin signaling pathways has been shown to increase lifespan. In rodents, however, growth hormone (GH) regulates IGF-1 levels in serum and tissues and can modulate lifespan via/or independent of IGF-1. Rodent models, where the GH/IGF-1 axis was ablated congenitally, show increased lifespan. However, in contrast to rodents where serum IGF-1 levels are high throughout life, in humans, serum IGF-1 peaks during puberty and declines thereafter during aging. Thus, animal models with congenital disruption of the GH/IGF-1 axis are unable to clearly distinguish between developmental and age-related effects of GH/IGF-1 on health. To overcome this caveat, we developed an inducible liver IGF-1-deficient (iLID) mouse that allows temporal control of serum IGF-1. Deletion of liver Igf-1 gene at one year of age reduced serum IGF-1 by 70% and dramatically impaired health span of the iLID mice. Reductions in serum IGF-1 were coupled with increased GH levels and increased basal STAT5B phosphorylation in livers of iLID mice. These changes were associated with increased liver weight, increased liver inflammation, increased oxidative stress in liver and muscle, and increased incidence of hepatic tumors. Lastly, despite elevations in serum GH, low levels of serum IGF-1 from 1 year of age compromised skeletal integrity and accelerated bone loss. We conclude that an intact GH/IGF-1 axis is essential to maintain health span and that elevated GH, even late in life, associates with increased pathology.


Subject(s)
Aging/metabolism , Insulin-Like Growth Factor I/deficiency , Aging/blood , Animals , Female , Insulin-Like Growth Factor I/metabolism , Male , Mice , Models, Animal , Oxidative Stress/physiology
6.
Endocrinology ; 154(11): 4182-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23970783

ABSTRACT

Bone formation and maintenance represents the summation of the balance of local and endocrine hormonal stimuli within a complex organ. Protein kinase D (PKD) is a member of the Ca(2+)/calmodulin-dependent kinase superfamily of serine/threonine kinases and has been described as the crossroads for the bone morphogenetic protein (BMP)-IGF-I signaling axis, which plays a major role in bone formation. The current study exploits the PKD1-deficient mouse model to examine the role of PKD in vivo in the skeleton. Dual-energy x-ray absorptiometry scan analysis of male and female pubescent mice demonstrated significantly decreased bone mineral density in the whole body and femoral bone compartments of PKD1 (+/-) mice, compared with their wild-type littermates. The body weight, nasal-anal length, and percentage body fat of the mice were not significantly different from their wild-type littermates. Cultured bone marrow stromal cells from PKD1 (+/-) mice demonstrated lower alkaline phosphatase activity in early differentiating osteoblasts and decreased mineralized nodule formation in mature osteoblasts. Quantitative RT-PCR analysis of osteoblast differentiation markers and osteoclast markers exhibited lower levels of expression in PKD1 (+/-) male mice than wild type. In female mice, however, only markers of osteoblast differentiation were reduced. PKD1 (+/-) mice also demonstrated a profound reduction in mRNA expression levels of BMP type II receptor and IGF-I receptor and in BMP-7 responsiveness in vitro. Together these data suggest that in mice, PKD1 action contributes to the regulation of osteoblastogenesis by altering gene expression with gender-specific effects on osteoclastogenesis, subsequently affecting skeletal matrix acquisition during puberty.


Subject(s)
Bone Development/physiology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Protein Kinase C/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Biomarkers , Bone Density , Cell Differentiation , Female , Male , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/metabolism , Protein Kinase C/genetics , Sex Factors , Sexual Maturation/physiology , Signal Transduction
7.
Biochem Biophys Res Commun ; 435(3): 501-5, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23680665

ABSTRACT

Type 2 Diabetes (T2DM) is the seventh leading cause of death in the United States, and is quickly becoming a global pandemic. T2DM results from reduced insulin sensitivity coupled with a relative failure of insulin secretion. Reduced insulin sensitivity has been associated with reduced nitric oxide synthase (NOS) activity and impaired glucose uptake in T2DM skeletal muscle. Upon insulin stimulation, NO synthesis increases in normal adult skeletal muscle, whereas no such increase is observed in T2DM adults. Endothelial NOS is activated by phosphorylation in the C-terminal tail in response to insulin. Neuronal NOS (nNOS), the primary NOS isoform in skeletal muscle, contains a homologous phosphorylation site, raising the possibility that nNOS, too, may undergo an activating phosphorylation event upon insulin treatment. Yet it remains unknown if or how nNOS is regulated by insulin in skeletal muscle. Data shown herein indicate that nNOS is phosphorylated in response to insulin in skeletal muscle and that this phosphorylation event occurs rapidly in C2C12 myotubes, resulting in increased NO production. In vivo phosphorylation of nNOS was also observed in response to insulin in mouse skeletal muscle. These results indicate, for the first time, that nNOS is phosphorylated in skeletal muscle in response to insulin and in association with increased NO production.


Subject(s)
Insulin/metabolism , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type I/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Insulin/pharmacology , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/chemistry , Phosphorylation
8.
PLoS One ; 8(5): e63838, 2013.
Article in English | MEDLINE | ID: mdl-23675509

ABSTRACT

IGF-1 receptor (IGF-1R) signaling regulates cell growth, transformation and survival. Haploinsufficiency of the IGF-1R is reported to paradoxically confer resistance to oxidative stress in vivo and in cells cultured from Igf1r(+/-) mice. In order to determine whether IGF-1R deficiency directly confers resistance to oxidative stress in specific cell types, an siRNA-mediated approach was applied to reduce IGF-1R in C2C12 myoblasts, NIH3T3 fibroblasts and MC3T3-E1 osteoblasts. Treating the IGF-1R deficient myoblasts with H2O2 resulted in significantly higher phosphorylation of Akt as compared to cells having normal expression of IGF-1R. Similar results were obtained with UV treatment, another inducer of oxidative stress. This enhanced activation of Akt was associated with reduced level of cleaved caspase-3 and PARP. Moreover, in the IGF-1R knockdown myoblasts, phosphorylation of the Akt substrate Bad was enhanced after peroxide treatment. However, in NIH-3T3 fibroblasts and MC3T3-E1 osteoblasts, the loss of IGF-1R by siRNA directed knockdown was associated with reduced levels of phosphorylated Akt on treatment with H2O2 or UV as compared to control cells and these cells showed more apoptosis. These results suggest a novel mechanism of cell type specific differential regulation of resistance to oxidative stress induced apoptosis by reduced levels of IGF-1R.


Subject(s)
Myoblasts/metabolism , Oxidative Stress/genetics , Receptor, IGF Type 1/deficiency , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Proliferation , Gene Knockdown Techniques , Hydrogen Peroxide/pharmacology , Mice , Myoblasts/drug effects , Myoblasts/radiation effects , NIH 3T3 Cells , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Ultraviolet Rays/adverse effects , bcl-Associated Death Protein/metabolism
9.
J Cell Biochem ; 114(8): 1760-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23444145

ABSTRACT

Bone morphogenetic proteins (BMPs) promote osteoblast differentiation and bone formation in vitro and in vivo. BMPs canonically signal through Smad transcription factors, but BMPs may activate signaling pathways traditionally stimulated by growth factor tyrosine kinase receptors. Of these, the mTOR pathway has received considerable attention because BMPs activate P70S6K, a downstream effector of mTOR, suggesting that BMP-induced osteogenesis is mediated by mTOR activation. However, contradictory effects of the mTOR inhibitor rapamycin (RAPA) on bone formation have been reported. Since bone formation is thought to be inversely related to lipid accumulation and mTOR is also important for lipid synthesis, we postulated that BMP-7 may stimulate lipogenic enzyme expression in a RAPA-sensitive mechanism. To test this hypothesis, we determined the effects of RAPA on BMP-7-stimulated expression of osteogenic and lipogenic markers in cultured fetal rat calvarial cells. Our study showed that BMP-7 promoted the expression of osteogenic and lipogenic markers. The effect of BMP-7 on osteogenic markers was greater in magnitude than on lipogenic markers and was temporally more sustained. RAPA inhibited basal and BMP-7-stimulated osteogenic and lipogenic marker expression and bone nodule mineralization. The acetyl CoA carboxylase inhibitor TOFA stimulated the expression of osteoblast differentiation markers, whereas palmitate suppressed their expression. We speculate that the BMP-7-stimulated adipogenesis is part of the normal anabolic response to BMPs, but that inappropriate activation of the lipid biosynthetic pathway by mTOR could have deleterious effects on bone formation and could explain paradoxical effects of RAPA to promote bone formation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antigens, Differentiation/biosynthesis , Bone Morphogenetic Protein 7/biosynthesis , Gene Expression Regulation/drug effects , Lipids/biosynthesis , Osteogenesis/drug effects , Sirolimus/pharmacology , Skull/metabolism , Animals , Calcification, Physiologic/drug effects , Cells, Cultured , Rats , Skull/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
10.
Endocrinol Metab Clin North Am ; 41(2): 231-47, v, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22682628

ABSTRACT

Insulin-like growth factor 1 (IGF-1) is a pleiotropic polypeptide. Its expression is tightly regulated and it plays significant roles during early development, maturation, and adulthood. This article discusses the roles of IGF-1 in determination of body size, skeletal acquisition, muscle growth, carbohydrate metabolism, and longevity, as learned from mouse models.


Subject(s)
Insulin-Like Growth Factor I/physiology , Models, Animal , Animals , Body Size/physiology , Female , Humans , Insulin-Like Growth Factor II/physiology , Lipid Metabolism , Longevity/physiology , Male , Mice , Muscle, Skeletal/physiology , Reproduction/physiology , Signal Transduction/physiology
11.
PLoS One ; 6(11): e26891, 2011.
Article in English | MEDLINE | ID: mdl-22132081

ABSTRACT

Mutations in insulin/IGF-1 signaling pathway have been shown to lead to increased longevity in various invertebrate models. Therefore, the effect of the haplo-insufficiency of the IGF-1 receptor (Igf1r(+/-)) on longevity/aging was evaluated in C57Bl/6 mice using rigorous criteria where lifespan and end-of-life pathology were measured under optimal husbandry conditions using large sample sizes. Igf1r(+/-) mice exhibited reductions in IGF-1 receptor levels and the activation of Akt by IGF-1, with no compensatory increases in serum IGF-1 or tissue IGF-1 mRNA levels, indicating that the Igf1r(+/-) mice show reduced IGF-1 signaling. Aged male, but not female Igf1r(+/-) mice were glucose intolerant, and both genders developed insulin resistance as they aged. Female, but not male Igf1r(+/-) mice survived longer than wild type mice after lethal paraquat and diquat exposure, and female Igf1r(+/-) mice also exhibited less diquat-induced liver damage. However, no significant difference between the lifespans of the male Igf1r(+/-) and wild type mice was observed; and the mean lifespan of the Igf1r(+/-) females was increased only slightly (less than 5%) compared to wild type mice. A comprehensive pathological analysis showed no significant difference in end-of-life pathological lesions between the Igf1r(+/-) and wild type mice. These data show that the Igf1r(+/-) mouse is not a model of increased longevity and delayed aging as predicted by invertebrate models with mutations in the insulin/IGF-1 signaling pathway.


Subject(s)
Aging/physiology , Receptor, IGF Type 1/metabolism , Signal Transduction , Aging/drug effects , Animals , Female , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Insulin/pharmacology , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Insulin-Like Growth Factor I/pharmacology , Longevity , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
12.
Biochem Biophys Res Commun ; 413(3): 476-80, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21910970

ABSTRACT

Interactions between genes and environment play a critical role in the pathogenesis of type 2 diabetes. Low birth weight, due to genetic and environmental variables affecting fetal growth, is associated with increased susceptibility to the development of type 2 diabetes and metabolic disorders in adulthood. Clinical studies have shown that polymorphisms in the Insulin-like growth factor 1 (IGF-1) gene or heterozygous mutations in IGF-1 and IGF-1 receptor (IGF-1R) genes, resulting in reduced IGF-1 action, are associated with low birth weight and post-natal growth. Mice lacking one of the IGF-1R alleles (Igf1r(+/-)) exhibit a 10% reduction in post-natal growth, and develop glucose intolerance (males) and insulin resistance (males and females) as they age. To investigate whether adverse environmental factors could accelerate the onset of the metabolic syndrome, we conducted a short duration intervention of high fat diet (HFD) feeding in male and female Igf1r(+/-) and wild-type (WT) control mice. The HFD resulted in insulin resistance, hyperglycemia, and impaired glucose tolerance in males of both genotypes whereas in females exacerbated diabetes was observed only in the Igf1r(+/-) genotype, thus suggesting a sexual dimorphism in the influence of obesity on the genetic predisposition to diabetes caused by reduced IGF-1 action.


Subject(s)
Diet, High-Fat/adverse effects , Genetic Predisposition to Disease , Glucose Intolerance/genetics , Insulin Resistance/genetics , Receptor, IGF Type 1/genetics , Sex Characteristics , Animals , Blood Glucose/genetics , Body Weight/genetics , CD36 Antigens/genetics , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Female , Gene Expression , Glucose Tolerance Test , Heterozygote , Insulin/blood , Male , Mice , Mice, Mutant Strains , Sex Factors
13.
Aging Cell ; 10(3): 547-50, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21418509

ABSTRACT

Although the literature suggests a protective (anabolic) effect of insulin-like growth factor-1 (IGF-1) on the musculoskeletal system during growth and aging, there is evidence that reductions in IGF-1 signaling are advantageous for promoting an increase in life span through reduction in oxidative stress-induced tissue damage. To better understand this paradox, we utilized the hepatocyte-specific IGF-1 transgenic (HIT) mice, which exhibit 3-fold increases in serum IGF-1, with normal IGF-1 expression in other tissues, and mice with an IGF-1 null background that exclusively express IGF-1 in the liver, which thereby deliver IGF-1 by the endocrine route only (KO-HIT mice). We found that in the total absence of tissue igf1 gene expression (KO-HIT), increases in serum IGF-1 levels were associated with increased levels of lipid peroxidation products in serum and increased mortality rate at 18 months of age in both genders. Surprisingly, however, we found that in female mice, tissue IGF-1 plays an important role in preserving trabecular bone architecture as KO-HIT mice show bone loss in the femoral distal metaphysis. Additionally, in male KO-HIT mice, increases in serum IGF-1 levels were insufficient to protect against age-related muscle loss.


Subject(s)
Aging , Insulin-Like Growth Factor I , Liver/metabolism , Longevity , Musculoskeletal System/metabolism , Animals , Bone Development/genetics , Female , Gene Expression , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Isoprostanes/analysis , Isoprostanes/biosynthesis , Lipid Peroxidation , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Mice , Mice, Transgenic , Models, Animal , Musculoskeletal System/pathology , Oxidative Stress , Sex Factors , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality
14.
Exp Gerontol ; 46(4): 265-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21094246

ABSTRACT

With the advancement of age, skeletal muscle undergoes a progressive decline in mass, function, and regenerative capacity. Previously, our laboratory has reported an age-reduction in recovery and local induction of IGF-I gene expression with age following tourniquet (TK)-induced skeletal muscle ischemia/reperfusion (I/R). In this study, young (6 mo) and old (24-28 mo) mice were subjected to 2h of TK-induced ischemia of the hindlimb followed by 1, 3, 5, or 7 days of reperfusion. Real time-PCR analysis revealed clear age-related reductions and temporal alterations in the expression of IGF-I and individual IGF-I Ea and Eb splice variants. ELISA verified a reduction of IGF-I peptide with age following 7 day recovery from TK. Western blotting showed that the phosphorylation of Akt, mTOR, and FoxO3, all indicators of anabolic activity, were reduced in the muscles of old mice. These data indicate that an age-related impairment of IGF-I expression and intracellular signaling does exist following injury, and potentially has a role in the impaired recovery of skeletal muscle with age.


Subject(s)
Aging/genetics , Insulin-Like Growth Factor I/genetics , Muscle, Skeletal/injuries , Reperfusion Injury/genetics , Aging/metabolism , Aging/pathology , Alternative Splicing , Animals , Base Sequence , DNA Primers/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sarcopenia/genetics , Sarcopenia/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
15.
Endocrinology ; 151(10): 4861-70, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20685873

ABSTRACT

IGF-I is an anabolic factor that mediates GH and PTH actions in bone. Expression of skeletal Igf1 differs for inbred strains of mice, and Igf expression levels correlate directly with bone mass. Previously we reported that peroxisome proliferator-activated receptor-γ2 activation in bone marrow suppressed Igf1 expression and that peroxisome proliferator-activated receptor-γ2 activation-induced Nocturnin (Noc) expression, a circadian gene with peak expression at light offset, which functions as a deadenylase. In 24-h studies we found that Igf1 mRNA exhibited a circadian rhythm in femur with the lowest Igf1 transcript levels at night when Noc transcripts were highest. Immunoprecipitation/RT-PCR analysis revealed a physical interaction between Noc protein and Igf1 transcripts. To clarify which portions of the Igf1 3' untranslated region (UTR) were necessary for regulation by Noc, we generated luciferase constructs containing various lengths of the Igf1 3'UTR. Noc did not affect the 170-bp short-form 3'UTR, but suppressed luciferase activity in constructs bearing the longer-form 3'UTR, which contains a number of potential regulatory motifs involved in mRNA degradation. C57BL/6J mice have low skeletal Igf1 mRNA compared with C3H/HeJ mice, and the Igf1 3' UTR is polymorphic between these strains. Interestingly, the activity of luciferase constructs bearing the long-form 3'UTR from C57BL/6J mice were repressed by Noc overexpression, whereas those bearing the corresponding region from C3H/HeJ were not. In summary, Noc interacts with Igf1 in a strain- and tissue-specific manner and reduces Igf1 expression by targeting the longer form of the Igf1 3'UTR. Posttranscriptional regulation of Igf1 may be critically important during skeletal acquisition and maintenance.


Subject(s)
3' Untranslated Regions , Bone and Bones/metabolism , Insulin-Like Growth Factor I/genetics , Nuclear Proteins/physiology , Transcription Factors/physiology , 3' Untranslated Regions/genetics , Animals , Animals, Newborn , Cells, Cultured , Circadian Rhythm/genetics , Down-Regulation/genetics , Female , Gene Targeting , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Proc Natl Acad Sci U S A ; 107(23): 10508-13, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20498072

ABSTRACT

Nocturnin (NOC) is a circadian-regulated protein related to the yeast family of transcription factors involved in the cellular response to nutrient status. In mammals, NOC functions as a deadenylase but lacks a transcriptional activation domain. It is highly expressed in bone-marrow stromal cells (BMSCs), hepatocytes, and adipocytes. In BMSCs exposed to the PPAR-gamma (peroxisome proliferator-activated receptor-gamma) agonist rosiglitazone, Noc expression was enhanced 30-fold. Previously, we reported that Noc(-/-) mice had low body temperature, were protected from diet-induced obesity, and most importantly exhibited absence of Pparg circadian rhythmicity on a high-fat diet. Consistent with its role in influencing BMSCs allocation, Noc(-/-) mice have reduced bone marrow adiposity and high bone mass. In that same vein, NOC overexpression enhances adipogenesis in 3T3-L1 cells but negatively regulates osteogenesis in MC3T3-E1 cells. NOC and a mutated form, which lacks deadenylase activity, bind to PPAR-gamma and markedly enhance PPAR-gamma transcriptional activity. Both WT and mutant NOC facilitate nuclear translocation of PPAR-gamma. Importantly, NOC-mediated nuclear translocation of PPAR-gamma is blocked by a short peptide fragment of NOC that inhibits its physical interaction with PPAR-gamma. The inhibitory effect of this NOC-peptide was partially reversed by rosiglitazone, suggesting that effect of NOC on PPAR-gamma nuclear translocation may be independent of ligand-mediated PPAR-gamma activation. In sum, Noc plays a unique role in the regulation of mesenchymal stem-cell lineage allocation by modulating PPAR-gamma activity through nuclear translocation. These data illustrate a unique mechanism whereby a nutrient-responsive gene influences BMSCs differentiation, adipogenesis, and ultimately body composition.


Subject(s)
Adipogenesis , Nuclear Proteins/metabolism , PPAR gamma/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , Body Composition , Cell Line , Cell Lineage , Circadian Rhythm , Humans , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factors/deficiency
17.
Growth Factors ; 28(5): 318-28, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20380591

ABSTRACT

We previously showed that exogenous insulin-like growth factor-I (IGF-I) and bone morphogenetic protein-7 (BMP-7) synergistically stimulated osteoblast differentiation in fetal rat calvaria (FRC) cells. We have now shown that BMP-7 alone and the BMP-7 and IGF-I combination synergistically stimulated protein kinase D (PKD) phosphorylation at Ser744/748 and Ser916. Transfection of FRC cells with a constitutively active PKD stimulated marker expression, while transfection with a catalytically inactive PKD did not. Moreover, Gö6976, which inhibits protein kinase C (PKC) α and β1, blocked PKD phosphorylation and the synergistic action of the BMP-7 and IGF-I combination on osteoblast differentiation, whereas Gö6983, which inhibits PKCα, β, γ, δ, and ζ, did not. Our results suggest that the FRC cell differentiation induced by BMP-7 and the BMP-7 and IGF-I combination requires stimulation of PKD activity. Our results are consistent with a novel mechanism in which combined BMP-7 and IGF-I signaling activates upstream novel PKC(s), which then phosphorylates and activates PKD, leading to enhanced osteoblast differentiation.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Cell Differentiation/drug effects , Insulin-Like Growth Factor I/pharmacology , Osteoblasts/drug effects , Protein Kinase C/metabolism , Animals , Carbazoles/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Osteoblasts/enzymology , Osteoblasts/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Rats
18.
Endocrinology ; 151(3): 865-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20130113

ABSTRACT

The discovery that IGF-I mRNAs encoding isoforms of the pro-IGF-I molecule are differentially regulated in response to mechanical stress in skeletal muscle has been the impetus for a number of studies designed to demonstrate that alternative splicing of IGF-I pre-mRNA involving exons 4, 5, and 6 gives rise to a unique peptide derived from pro-IGF-I that plays a novel role in myoblast proliferation. Research suggests that after injury to skeletal muscle, the IGF-IEb mRNA splice variant is up-regulated initially, followed by up-regulation of the IGF-IEa splice variant at later time points. Up-regulation of IGF-IEb mRNA correlates with markers of satellite cell and myoblast proliferation, whereas up-regulation of IGF-IEa mRNA is correlated with differentiation to mature myofibers. Due to the apparent role of IGF-IEb up-regulation in muscle remodeling, IGF-IEb mRNA was also named mechano-growth factor (MGF). A synthetically manufactured peptide (also termed MGF) corresponding to the 24 most C-terminal residues of IGF-IEb has been shown to promote cellular proliferation and survival. However, no analogous peptide product of the Igf1 gene has been identified in or isolated from cultured cells, their conditioned medium, or in vivo animal tissues or biological fluids. This review will discuss the relationship of the Igf1 gene to MGF and will differentiate actions of synthetic MGF from any known product of Igf1. Additionally, the role of MGF in satellite cell activation, aging, neuroprotection, and signaling will be discussed. A survey of outstanding questions relating to MGF will also be provided.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Regeneration , Aging/physiology , Animals , Gene Expression , Humans , Insulin-Like Growth Factor I/genetics , Mitogens/metabolism , Neuroprotective Agents/metabolism , Satellite Cells, Skeletal Muscle/physiology , Signal Transduction
19.
Biochem Biophys Res Commun ; 390(2): 252-7, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19799871

ABSTRACT

The PI3K/Akt/mTOR signaling pathway is critical for cellular growth and survival in skeletal muscle, and is activated in response to growth factors such as insulin-like growth factor-I (IGF-I). We found that in C2C12 myoblasts, deficiency of PI3K p110 catalytic subunits or Akt isoforms had distinct effects on phosphorylation of mTOR and p70S6K. siRNA-mediated knockdown of PI3K p110alpha, p110beta, and simultaneous knockdown of p110alpha and p110beta resulted in increased basal and IGF-I-stimulated phosphorylation of mTOR S2448 and p70S6K T389; however, phosphorylation of S6 was reduced in p110beta-deficient cells, possibly due to reductions in total S6 protein. We found that IGF-I-stimulated Akt1 activity was enhanced in Akt2- or Akt3-deficient cells, and that knockdown of individual Akt isoforms increased mTOR/p70S6K activation in an isoform-specific fashion. Conversely, levels of IGF-I-stimulated p70S6K phosphorylation in cells simultaneously deficient in both Akt1 and Akt3 were increased beyond those seen with loss of any single Akt isoform, suggesting an alternate, Akt-independent mechanism that activates mTOR/p70S6K. Our results collectively suggest that mTOR/p70S6K is activated in a PI3K/Akt-dependent manner, but that in the absence of p110alpha or Akt, alternate pathway(s) may mediate activation of mTOR/p70S6K in C2C12 myoblasts.


Subject(s)
Carrier Proteins/metabolism , Myoblasts/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Catalytic Domain , Enzyme Activation , Gene Knockdown Techniques , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Myoblasts/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases
20.
Biochem Biophys Res Commun ; 389(1): 117-21, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19703413

ABSTRACT

Oxidative stress has been shown to induce apoptosis in a variety of tissues, while insulin-like growth factor-I (IGF-I) can oppose this effect. We found that H(2)O(2) promoted cell death and apoptosis in C2C12 myoblasts, an effect that was completely prevented by exogenous IGF-I. One downstream mediator of IGF-I survival signaling is the serine/threonine kinase Akt, of which three isoforms have been identified in mammals. We found that Akt1 and Akt3 act on pro-apoptotic target molecules in an isoform-specific manner. Both Akt1 and Akt3 were responsible for phosphorylating FoxO3a at S253 and FoxO1 at T24, while Akt1 alone phosphorylated Bad at S136 and FoxO3a at T32. Our results provide evidence for IGF-I-stimulated isoform-specific actions of Akt on molecules involved in promoting apoptosis.


Subject(s)
Apoptosis , Insulin-Like Growth Factor I/metabolism , Myoblasts/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Cell Survival , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Hydrogen Peroxide/toxicity , Insulin-Like Growth Factor I/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Myoblasts/enzymology , Oxidative Stress , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , Signal Transduction , bcl-Associated Death Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL