Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 8(4): 796-809, 2016 04.
Article in English | MEDLINE | ID: mdl-27070352

ABSTRACT

Mitochondria are key players in aging and in the pathogenesis of age-related diseases. Recent mitochondrial transcriptome analyses revealed the existence of multiple small mRNAs transcribed from mitochondrial DNA (mtDNA). Humanin (HN), a peptide encoded in the mtDNA 16S ribosomal RNA region, is a neuroprotective factor. An in silico search revealed six additional peptides in the same region of mtDNA as humanin; we named these peptides small humanin-like peptides (SHLPs). We identified the functional roles for these peptides and the potential mechanisms of action. The SHLPs differed in their ability to regulate cell viability in vitro. We focused on SHLP2 and SHLP3 because they shared similar protective effects with HN. Specifically, they significantly reduced apoptosis and the generation of reactive oxygen species, and improved mitochondrial metabolism in vitro. SHLP2 and SHLP3 also enhanced 3T3-L1 pre-adipocyte differentiation. Systemic hyperinsulinemic-euglycemic clamp studies showed that intracerebrally infused SHLP2 increased glucose uptake and suppressed hepatic glucose production, suggesting that it functions as an insulin sensitizer both peripherally and centrally. Similar to HN, the levels of circulating SHLP2 were found to decrease with age. These results suggest that mitochondria play critical roles in metabolism and survival through the synthesis of mitochondrial peptides, and provide new insights into mitochondrial biology with relevance to aging and human biology.


Subject(s)
Apoptosis/physiology , Insulin Resistance/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Peptides/metabolism , 3T3-L1 Cells , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , DNA, Mitochondrial/metabolism , Humans , Intracellular Signaling Peptides and Proteins/pharmacology , Mice , Peptides/pharmacology
2.
Endocrinology ; 154(10): 3739-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836030

ABSTRACT

Humanin (HN) is a novel 24-amino acid mitochondrial-derived peptide that has demonstrated diverse cytoprotective effects, including an emerging role in diabetes. The purpose of this study was to examine the pharmacokinetics of humanin analogues, which show great potential as therapeutic agents (HNG and the non-IGFBP-3 binding, HNGF6A). 11-week-old male IGFBP-3(-/-) and wild type (WT) mice were divided into 3 groups: WT mice treated with HNG, WT mice treated with HNGF6A, and IGFBP-3(-/-) mice treated with HNG. Plasma was obtained from mice following ip injection with HN analogues, and HN levels were measured with ELISA. WT mice treated with HNGF6A and IGFBP-3(-/-) mice treated with HNG displayed a longer half-life of HN compared with WT mice treated with HNG. Following HNG injection, both IGF-1 and IGFBP-3 levels decreased over time. Adult male Sprague Dawley rats were also ip injected with HNG, and HN levels were measured in various tissues (plasma, liver, heart, and brain) by ELISA. The half-life of HN was found to be longer in rats compared with mice. In rats, HN levels were found to be highest in plasma, present in liver, and undetectable in brain or heart. The current study provides evidence of HN and IGFBP-3 association in the circulation and suggests that native HN may modulate the distribution of IGF-1 and IGFBP-3. The results also demonstrate varying kinetic profiles of HN analogues and interspecies variation in rodents. Sustainable levels of circulating HN measured in plasma underline the potential value of HN analogues as a new therapeutic intervention in the treatment of diabetes.


Subject(s)
Drugs, Investigational/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/pharmacokinetics , Animals , Biotransformation , Drugs, Investigational/administration & dosage , Drugs, Investigational/chemistry , Drugs, Investigational/metabolism , Enzyme-Linked Immunosorbent Assay , Half-Life , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Injections, Intraperitoneal , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution
3.
Atherosclerosis ; 219(1): 65-73, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21763658

ABSTRACT

OBJECTIVE: Humanin (HN) is a cytoprotective peptide derived from endogenous mitochondria, expressed in the endothelial layer of human vessels, but its role in atherogenesis in vivo is not known. In vitro study, however, HN reduced oxidized low-density lipoprotein induced formation of reactive oxygen species and apoptosis. The present study tested the hypothesis that long term treatment with HN will have a protective role against endothelial dysfunction and progression of atherosclerosis in vivo. METHODS AND RESULTS: Daily intraperitonial injection of the HN analogue HNGF6A for 16 weeks prevented endothelial dysfunction and decreased atherosclerotic plaque size in the proximal aorta of ApoE-deficient mice fed on a high cholesterol diet, without showing direct vasoactive effects or cholesterol-reducing effects. HN was expressed in the endothelial layer on the aortic plaques. HNGF6A treatment reduced apoptosis and nitrotyrosine immunoreactivity in the aortic plaques without affecting the systemic cytokine profile. HNGF6A also preserved expression of endothelial nitric oxide synthase in aorta. CONCLUSIONS: HN may have a protective effect on endothelial function and progression of atherosclerosis by modulating oxidative stress and apoptosis in the developing plaque.


Subject(s)
Hypercholesterolemia/physiopathology , Intracellular Signaling Peptides and Proteins/pharmacology , Plaque, Atherosclerotic/prevention & control , Animals , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Cholesterol, Dietary/administration & dosage , Disease Progression , Female , Mice
4.
Cancer Res ; 71(15): 5154-63, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21697285

ABSTRACT

The insulin-like growth factor binding protein IGFBP-3 is a proapoptotic and antiangiogenic protein in prostate cancer (CaP). Epidemiologic studies suggest that low IGFBP-3 is associated with greater risk of aggressive, metastatic prostate cancers, but in vivo functional data are lacking. Here we show that mice that are genetically deficient in IGFBP-3 exhibit weaker growth of primary prostate tumors but higher incidence of metastatic disease. Prostates in IGFBP-3 knockout mice (IGFBP-3KO mice) failed to undergo apoptosis after castration. Spontaneous prostate tumors did not develop in IGFBP-3KO mice, but splenic lymphomas occurred in 23% of female IGFBP-3KO mice by 80 weeks of age. To assess the effects of IGFBP-3 deficiency on prostate cancer development, we crossed IGFBP-3KO mice with a c-Myc-driven model of CaP that develops slow-growing, nonmetastatic tumors. By 24 weeks of age, well-differentiated prostate cancers were observed in all mice regardless of IGFBP-3 status. However, by 80 weeks of age IGFBP-3KO mice tended to exhibit larger prostate tumors than control mice. More strikingly, lung metastases were observed at this time in 55% of the IGFBP-3KO mice but none in the control animals. Cell lines established from IGFBP-3KO:Myc tumors displayed more aggressive phenotypes in proliferation, invasion, and colony formation assays, relative to control Myc tumor cell lines. In addition, Myc:IGFBP-3KO cells exhibited evidence of epithelial-mesenchymal transition. Our findings established a function for IGFBP-3 in suppressing metastasis in prostate cancer, and they also offered the first reported transgenic model of spontaneous metastatic prostate cancer for studies of this advanced stage of disease.


Subject(s)
Adenocarcinoma/secondary , Androgens , Insulin-Like Growth Factor Binding Protein 3/physiology , Neoplasm Invasiveness/genetics , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/secondary , Prostatic Neoplasms/pathology , Adenocarcinoma/genetics , Animals , Apoptosis , Cell Line, Tumor/cytology , Crosses, Genetic , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Genes, myc , Humans , Insulin-Like Growth Factor Binding Protein 3/deficiency , Insulin-Like Growth Factor Binding Protein 3/genetics , Lung Neoplasms/secondary , Lymphoma, Non-Hodgkin/genetics , Male , Mice , Mice, Knockout , Neoplasms, Hormone-Dependent/genetics , Orchiectomy , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Splenic Neoplasms/genetics , Tumor Burden , Tumor Stem Cell Assay
5.
Cardiovasc Res ; 88(2): 360-6, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20562421

ABSTRACT

AIMS: Humanin (HN) is a 24-amino acid peptide that has been shown to have an anti-apoptotic function against neuronal cell death caused by Alzheimer's disease. Increased oxidative stress, one of the major factors contributing to this cell death, also plays an important role in the inflammatory process of atherosclerosis. The current study was designed to test the hypothesis that HN is expressed in the human vascular wall and may protect against oxidative stress. METHODS AND RESULTS: HN expression in the vascular wall was detected by immunostaining in the endothelial cell layer of human internal mammary arteries (n = 5), atherosclerotic coronary arteries (n = 17), and sections of the greater saphenous vein (n = 3). HN mRNA was expressed in the human aortic endothelial cells (HAECs). Cytoprotective effects of HN against oxidative stress were tested in vitro in HAECs. Pre-treatment with 0.1 µM HN reduced oxidized LDL (Ox-LDL)-induced (i) formation of reactive oxygen species by 50%, (ii) apoptosis by ∼50% as determined by TUNEL staining, and (iii) formation of ceramide, a lipid second messenger involved in the apoptosis signalling cascade, by ∼20%. CONCLUSION: The current study demonstrates for the first time the expression of HN in the endothelial cell layer of human blood vessels. Exogenous addition of HN to endothelial cell cultures was shown to be effective against Ox-LDL-induced apoptosis. These findings suggest that HN may play a role and may have a protective effect in early atherosclerosis in humans.


Subject(s)
Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipoproteins, LDL/metabolism , Oxidative Stress , Adult , Aged , Apoptosis , Cells, Cultured , Ceramides/metabolism , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Cytoprotection , Endothelial Cells/pathology , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/genetics , Mammary Arteries/metabolism , Mammary Arteries/pathology , Middle Aged , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Saphenous Vein/metabolism , Saphenous Vein/pathology
6.
Endocrinology ; 151(1): 350-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19952275

ABSTRACT

Modulating germ cell death and survival have significant therapeutic potential for male infertility and contraception. We have shown previously that IGF binding protein 3 (IGFBP3) gene expression is up-regulated in human testis when germ cell apoptosis is induced by intratesticular hormonal deprivation created by testosterone administration. Humanin (HN) is a binding partner of IGFBP3, and both are expressed in rat testes. We therefore hypothesized that IGFBP3, a proapoptotic factor, and HN, an antiapoptotic factor, are important regulators of male germ cell apoptosis. Whereas baseline apoptosis in the testis was equivalent between Igfbp3 knockout and wild-type mice, treatment with GnRH antagonist (GnRH-A) for 2 wk induced germ cell apoptosis in wild type, which was dramatically reduced in Igfbp3 knockout mice. To investigate the direct effects of IGFBP3 and HN on germ cell apoptosis, intratesticular administration of IGFBP3 for 5 d in rats induced a 4.2- and 3.8-fold increase in apoptosis at stages VII-VIII and XIV-I of the seminiferous epithelium cycle, respectively. GnRH-A treatment for 5 d increased apoptosis, mainly at stages VII-VIII. Addition of IGFBP3 to GnRH-A treatment enhanced apoptosis to 39.3-fold at stages VII-VIII, which was higher than either treatment alone. Intratesticular injection of HN significantly decreased GnRH-A-induced apoptosis at stages XIV-I but not stages VII-VIII. We conclude that IGFBP3 and HN play key roles in the coordinated regulation of testicular germ cell homeostasis. Perturbation of this interaction is important in enhancing or preventing germ cell death, providing new targets for future therapies.


Subject(s)
Apoptosis , Germ Cells/physiology , Insulin-Like Growth Factor Binding Protein 3/physiology , Intracellular Signaling Peptides and Proteins/physiology , Spermatozoa/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Drug Antagonism , Female , Germ Cells/drug effects , Germ Cells/metabolism , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Injections, Subcutaneous , Insulin-Like Growth Factor Binding Protein 3/administration & dosage , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/anatomy & histology , Testis/drug effects , Testis/physiology
7.
Growth Horm IGF Res ; 20(1): 55-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19853487

ABSTRACT

The IGF axis is critical for the regulation of apoptosis in many human cancer cell lines. Recently, potent anti-tumorigenic effects of pomegranate juice and extracts have been reported. Consequently, pomegranate has potential not only as a treatment but also as a preventative measure against certain types of cancer, including prostate. In this study, we investigated the relationship between pomegranate-induced apoptosis in human prostate cancer cells and the IGF/IGFBP system. Treatment of LAPC4 prostate cancer cells with 10microg/ml POMx, a highly potent pomegranate extract prepared from skin and arils minus seeds and standardized to ellagitannin content (37% punicalagins by HPLC), resulted in inhibition of cell proliferation and induction of apoptosis. Interestingly, co-treatment with POMx and IGFBP-3 revealed synergistic stimulation of apoptosis and additive inhibition of cell growth. Western blot analysis revealed that treatment with POMx or POMx/IGFBP-3 combination resulted in increased JNK phosphorylation, and decreased Akt and mTOR activation, consistent with a growth inhibitory, pro-apoptotic function. We also investigated the relationship between IGF-1 and pomegranate-induced apoptosis in 22RV1 prostate cancer cells. Co-treatment with 100ng/ml IGF-1 completely blocked apoptosis induction by POMx. In contrast, IGF-I failed to inhibit POMx-induced apoptosis in R(-) cells, suggesting the importance of IGF-IR. POMx-treatment decreased Igf1 mRNA expression in a dose-dependent manner indicating that its actions also involve tumor-specific suppression of IGF-1. These studies revealed novel interactions between the IGF system and pomegranate-induced apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Lythraceae/chemistry , Plant Extracts/pharmacology , Prostatic Neoplasms/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hydrolyzable Tannins/analysis , Hydrolyzable Tannins/pharmacology , Intracellular Signaling Peptides and Proteins/drug effects , JNK Mitogen-Activated Protein Kinases/drug effects , Male , Plant Extracts/chemistry , Protein Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases
8.
J Biol Chem ; 285(3): 1726-32, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19887447

ABSTRACT

Germ cell apoptosis is crucial for spermatogenesis and can be triggered by various stimuli, including intratesticular hormone deprivation. This study proposes a role for insulin-like growth factor binding protein-3 (IGFBP-3) in male germ cell apoptosis. Groups of adult Sprague-Dawley male rats received one of the following treatments for 5 days: (i) daily intratesticular (IT) injections with saline (control); (ii) a single subcutaneous injection of the gonadotropin-releasing hormone antagonist (GnRH-A), acyline, on day 1 and a daily IT injection of saline; (iii) daily IT injection of IGFBP-3; and (iv) a GnRH-A injection on day 1 and a daily IT injection of IGFBP-3. Germ cell apoptosis increased significantly after IGFBP-3 or GnRH-A treatment which was further enhanced by the combined treatment. After co-immunoprecipitation with BAX antibody, IGFBP-3 association with BAX was demonstrated in total and mitochondrial fractions but not in the cytosol of testis extracts. BAX-associated IGFBP-3 expression was increased in mitochondria after treatment compared with control, which was confirmed by an IGFBP-3 enzyme-linked immunosorbent assay. Dot blot studies further validated the BAX-IGFBP-3 binding in vitro. IGFBP-3 as well as BAX induced release of cytochrome c and DIABLO from isolated testicular mitochondria in vitro. IGFBP-3, when combined with an ineffective dose of BAX, triggered release of these proteins from isolated mitochondria at a 4-fold lower dose than IGFBP-3 alone. Our data demonstrate that the IGFBP-3 and BAX interaction activates germ cell apoptosis via the mitochondria-dependent pathway. This represents a novel pathway regulating germ call homeostasis that may have significance for male fertility and testicular disease.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mitochondria/metabolism , Spermatozoa/cytology , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Cell Fractionation , Cytochromes c/metabolism , Enzyme-Linked Immunosorbent Assay , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Male , Mice , Mitochondrial Proteins/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Signal Transduction , Spermatozoa/metabolism , Testis/cytology
9.
Metabolism ; 59(3): 343-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19800083

ABSTRACT

Pancreatic beta-cell apoptosis is important in the pathogenesis and potential treatment of type 1 diabetes mellitus. We investigated whether Humanin, a recently described survival factor for neurons, could improve the survival of beta-cells and delay or treat diabetes in the nonobese diabetic (NOD) model. Humanin reduced apoptosis induced by serum starvation in NIT-1 cells and decreased apoptosis induced by cytokine treatment. Humanin induced signal transducer and activator of transcription 3 and extracellular signal-regulated kinase phosphorylation over a 24-hour time course. Specific inhibition of signal transducer and activator of transcription 3 resulted in nullifying the protective effect of Humanin. Humanin normalized glucose tolerance in NOD mice treated for 6 weeks, and their pancreata revealed decreased lymphocyte infiltration and severity. In addition, Humanin delayed/prevented the onset of diabetes in NOD mice treated for 20 weeks. In summary, Humanin treatment decreases cytokine-induced apoptosis in beta-cells in vitro and improved glucose tolerance and onset of diabetes in NOD mice in vivo. This indicates that Humanin may be useful for islet protection and survival in a spectrum of diabetes-related therapeutics.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus/prevention & control , Insulin-Secreting Cells/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , STAT3 Transcription Factor/physiology , Animals , Caspases/metabolism , Diabetes Mellitus/genetics , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glucose Tolerance Test , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred NOD , Neurons/drug effects , Neurosecretory Systems/cytology , Neurosecretory Systems/physiology
10.
J Cell Sci ; 122(Pt 17): 3104-12, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19654213

ABSTRACT

During the differentiation of muscle satellite cells, committed myoblasts respond to specific signalling cues by exiting the cell cycle, migrating, aligning, expressing muscle-specific genes and finally fusing to form multinucleated myotubes. The predominant foetal growth factor, IGF-2, initiates important signals in myogenesis. The aim of this study was to investigate whether ERK5 and its upstream MKK activator, MEK5, were important in the pro-myogenic actions of IGF-2. ERK5 protein levels, specific phosphorylation and kinase activity increased in differentiating C2 myoblasts. ERK5-GFP translocated from the cytoplasm to the nucleus after activation by upstream MEK5, whereas phospho-acceptor site mutated (dominant-negative) ERK5AEF-GFP remained cytoplasmic. Exogenous IGF-2 increased MHC levels, myogenic E box promoter-reporter activity, ERK5 phosphorylation and kinase activity, and rapidly induced nuclear localisation of ERK5. Transfection with antisense Igf2 decreased markers of myogenesis, and reduced ERK5 phosphorylation, kinase and transactivation activity. These negative effects of antisense Igf2 were rescued by constitutively active MEK5, whereas transfection of myoblasts with dominant-negative MEK5 blocked the pro-myogenic action of IGF-2. Our findings suggest that the MEK5-ERK5 pathway is a novel key mediator of IGF-2 action in myoblast differentiation.


Subject(s)
Insulin-Like Growth Factor II/metabolism , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Muscle Development , Animals , Cell Differentiation , Cell Line , Insulin-Like Growth Factor II/genetics , MAP Kinase Kinase 5/genetics , Mice , Mitogen-Activated Protein Kinase 7/genetics , Myoblasts/cytology , Myoblasts/enzymology , Phosphorylation , Protein Transport , Rats , Signal Transduction , Transcriptional Activation
11.
PLoS One ; 4(7): e6334, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19623253

ABSTRACT

BACKGROUND: Decline in insulin action is a metabolic feature of aging and is involved in the development of age-related diseases including Type 2 Diabetes Mellitus (T2DM) and Alzheimer's disease (AD). A novel mitochondria-associated peptide, Humanin (HN), has a neuroprotective role against AD-related neurotoxicity. Considering the association between insulin resistance and AD, we investigated if HN influences insulin sensitivity. METHODS AND FINDINGS: Using state of the art clamp technology, we examined the role of central and peripheral HN on insulin action. Continuous infusion of HN intra-cerebro-ventricularly significantly improved overall insulin sensitivity. The central effects of HN on insulin action were associated with activation of hypothalamic STAT-3 signaling; effects that were negated by co-inhibition of hypothalamic STAT-3. Peripheral intravenous infusions of novel and potent HN derivatives reproduced the insulin-sensitizing effects of central HN. Inhibition of hypothalamic STAT-3 completely negated the effects of IV HN analog on liver, suggesting that the hepatic actions of HN are centrally mediated. This is consistent with the lack of a direct effect of HN on primary hepatocytes. Furthermore, single treatment with a highly-potent HN analog significantly lowered blood glucose in Zucker diabetic fatty rats. Based upon the link of HN with two age-related diseases, we examined if there were age associated changes in HN levels. Indeed, the amount of detectable HN in hypothalamus, skeletal muscle, and cortex was decreased with age in rodents, and circulating levels of HN were decreased with age in humans and mice. CONCLUSIONS: We conclude that the decline in HN with age could play a role in the pathogenesis of age-related diseases including AD and T2DM. HN represents a novel link between T2DM and neurodegeneration and along with its analogues offers a potential therapeutic tool to improve insulin action and treat T2DM.


Subject(s)
Insulin/physiology , Intracellular Signaling Peptides and Proteins/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance , Liver/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker
12.
Mol Endocrinol ; 23(10): 1624-33, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19556345

ABSTRACT

IGF-binding protein 3 (IGFBP-3) promotes apoptosis by both IGF-dependent and -independent mechanisms. We have previously reported that phosphorylation of IGFBP-3 (S156) by DNA-dependent protein kinase enhances its nuclear accumulation and is essential for its ability to interact with retinoid X receptor-alpha and induce apoptosis in cultured prostate cancer cells. Using specific chemical inhibitors and small interfering RNA, we demonstrate that preventing casein kinase 2 (CK2) activation enhanced the apoptotic potential of IGFBP-3. We mapped potential CK2 phosphosphorylation sites in IGFBP-3 to S167 and S175 and identified that wild-type IGFBP-3- and IGFBP-3-S175A-induced apoptosis to a comparable extent. In contrast, IGFBP-3-S167A was far more potently apoptosis inducing due to inability to undergo CK2 phosphorylation. Pretreatment of 22RV1 cells with IGFBP-3 small interfering RNA also limits the ability of high doses of CK2 inhibitor to induce apoptosis. These effects can be reversed by the addition of exogenous IGFBP-3 protein, suggesting reciprocal regulation of cell survival and apoptosis by IGFBP-3 and CK2. These studies reveal multisite phosphorylation of IGFBP-3 that both positively and negatively regulate its apoptotic potential. Understanding such intrinsic regulation of IGFBP-3 action may enhance the development of potential cancer therapies.


Subject(s)
Apoptosis , Casein Kinase II/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Amino Acid Sequence , Animals , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , DNA-Activated Protein Kinase/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Organ Specificity , Phosphorylation , Phosphoserine/metabolism , Protein Transport
13.
FASEB J ; 23(8): 2616-26, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19332648

ABSTRACT

IGF activity is regulated tightly by a family of IGF binding proteins (IGFBPs). IGFBP-5 is the most conserved of these and is up-regulated significantly during differentiation of several key lineages and in some cancers. The function of IGFBP-5 in these physiological and pathological situations is unclear, however, several IGFBP-5 sequence motifs and studies in vitro suggest IGF-independent actions. Therefore, we aimed to compare the phenotypes of mice overexpressing wild-type Igfbp5 or an N-terminal mutant Igfbp5 with negligible IGF binding affinity. Both significantly inhibited growth, even at low expression levels. Even though wild-type IGFBP-5 severely disrupted the IGF axis, we found no evidence for interaction of mutant IGFBP-5 with the IGF system. Further, overexpression of wild-type IGFBP-5 rescued the lethal phenotype induced by "excess" IGF-II in type 2 receptor-null mice; mutant IGFBP-5 overexpression could not. Therefore, wild-type IGFBP-5 provides a very effective mechanism for the inhibition of IGF activity and a powerful in vivo mechanism to inhibit IGF activity in pathologies such as cancer. This study is also the first to suggest significant IGF-independent actions for IGFBP-5 during development.


Subject(s)
Insulin-Like Growth Factor Binding Protein 5/metabolism , Somatomedins/metabolism , Animals , Base Sequence , DNA Primers/genetics , Female , Gene Expression , Insulin-Like Growth Factor Binding Protein 5/deficiency , Insulin-Like Growth Factor Binding Protein 5/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Muscle Development , Muscle, Skeletal/metabolism , Mutation , Phenotype , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tissue Distribution
14.
Mol Endocrinol ; 23(2): 169-75, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19095771

ABSTRACT

IGF-binding proteins (IGFBPs) have multiple cellular effects, which occur by both IGF-dependent and -independent mechanisms. IGFBP-2 is involved in the regulation of both normal and carcinogenic cell growth. To further understand the actions of IGFBP-2, we carried out a yeast two-hybrid screen to search for intracellular partner proteins using a human prostate cDNA library. We isolated Pim-1-associated protein-1 (PAP-1)-associated protein-1 (PAPA-1) as an IGFBP-2-binding protein, whose expression and subcellular localization is regulated by both IGFBP-2 and androgens. Coimmunoprecipitation and glutathione S-transferase pull-down assay confirmed the interaction in vitro, and confocal microscopy showed the colocalization of IGFBP-2 and PAPA-1 in the nucleus. Suppression of PAPA-1 by small interfering RNA treatment enhanced the growth-promoting effect of IGFBP-2. Conversely, IGFBP-2-promoted bromodeoxyuridine incorporation into LNCaP cells was abrogated by the simultaneous overexpression of myc-hPAPA-1. Mouse embryonic fibroblasts from IGFBP-2 knockout mouse showed diminished growth activity compared with wild type, and expression of FLAG-mPAPA-1 decreased cell proliferation in IGFBP-2 knockout, but not control mouse embryonic fibroblasts. These studies suggest that the growth-promoting role of IGFBP-2 in prostate cancer is inhibited by its intracellular interaction with PAPA-1.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Nuclear Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Pancreatitis-Associated Proteins , Prostate/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Two-Hybrid System Techniques
15.
Cancer Res ; 68(9): 3342-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451161

ABSTRACT

The role of systemic and local insulin-like growth factor I (IGF-I) in the development of prostate cancer is still controversial. Transgenic adenocarcinoma mouse prostate (TRAMP) mice express the SV40 T-antigen under the control of the probasin promoter, and spontaneously develop prostate cancer. We crossed TRAMP mice with liver IGF-deficient (LID) mice to produce LID-TRAMP mice, a mouse model of prostate cancer with low serum IGF-I, to allow us to study the effect of circulatory IGF-I levels on the development of prostate cancer. LID mice have a targeted deletion of the hepatic Igf1 gene but retain normal expression of Igf1 in extrahepatic tissues. Serum IGF-I and IGFBP-3 levels in LID and LID-TRAMP mice were measured using novel assays, which showed that they are approximately 10% and 60% of control L/L- mice, respectively. Serum growth hormone (GH) levels of LID-TRAMP mice were 3.5-fold elevated relative to L/L-TRAMP mice (P < 0.001), but IGFBP-2 levels were not different. Surprisingly, rates of survival, metastasis, and the ratio of genitourinary tissue weight to body weight were not significantly different between LID-TRAMP and L/L-TRAMP mice. There was also no difference in the pathologic stage of the prostate cancer between the two groups at 9 to 19 weeks of age. LID-TRAMP tumors displayed increased levels of GH receptors and increased Akt phosphorylation. These results are in striking contrast with the published model of the GH-deficient lit/lit-TRAMP, which has smaller tumors and improved survival, and indicate that the reduction in systemic IGF-I is not sufficient to inhibit prostate cancer tumor progression in the TRAMP model, which may require a reduction of GH levels as well.


Subject(s)
Adenocarcinoma/blood , Adenocarcinoma/genetics , Gene Deletion , Insulin-Like Growth Factor I/genetics , Mutagenesis, Site-Directed , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Adenocarcinoma/pathology , Animals , Disease Progression , Female , Genotype , Growth Hormone/blood , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Serum/chemistry , Signal Transduction/genetics , Tumor Burden
16.
Carcinogenesis ; 28(8): 1653-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17434920

ABSTRACT

Tumor suppression by insulin-like growth factor-binding protein-3 (IGFBP-3) has been demonstrated to occur via insulin-like growth factor-dependent and -independent mechanisms in vitro and in vivo. We have recently described IGFBP-3-induced mitochondrial translocation of the nuclear receptors RXRalpha/Nur77 in the induction of prostate cancer (CaP) cell apoptosis. Herein, we demonstrate that IGFBP-3 and Nur77 associate in the cytoplasmic compartment in 22RV1 CaP cells. Nur77 is a major component of IGFBP-3-induced apoptosis as shown by utilizing mouse embryonic fibroblasts (MEFs) derived from Nur77 wild-type and knockout (KO) mice. However, dose-response experiments revealed that a small component of IGFBP-3-induced apoptosis is Nur77 independent. Reintroduction of Nur77 into Nur77 KO MEFs restores full responsiveness to IGFBP-3. IGFBP-3 induces phosphorylation of Jun N-terminal kinase and inhibition of Akt phosphorylation and activity, which have been associated with Nur77 translocation. Finally, IGFBP-3 administration to CaP xenografts on SCID mice induced apoptosis and translocated Nur77 out of the nucleus. Taken together, our results verify an important role for the orphan nuclear receptor Nur77 in the apoptotic actions of IGFBP-3.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/physiology , Insulin-Like Growth Factor Binding Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Transcription Factors/physiology , Animals , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Nuclear Receptor Subfamily 4, Group A, Member 1 , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding/physiology , Protein Transport/physiology , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Subcellular Fractions/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
17.
J Cell Sci ; 119(Pt 23): 4828-40, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17105766

ABSTRACT

Cell-cell contact is essential for appropriate co-ordination of development and it initiates significant signalling events. During myogenesis, committed myoblasts migrate to sites of muscle formation, align and form adhesive contacts that instigate cell-cycle exit and terminal differentiation into multinucleated myotubes; thus myogenesis is an excellent paradigm for the investigation of signals derived from cell-cell contact. PI3-K and p38 MAPK are both essential for successful myogenesis. Pro-myogenic growth factors such as IGF-II activate PI3-K via receptor tyrosine kinases but the extracellular cues and upstream intermediates required for activation of the p38 MAPK pathway in myoblast differentiation are not known. Initial observations suggested a correlation between p38 MAPK phosphorylation and cell density, which was also related to N-cadherin levels and Igf2 expression. Subsequent studies using N-cadherin ligand, dominant-negative N-cadherin, constitutively active and dominant-negative forms of RhoA, and MKK6 and p38 constructs, reveal a novel pathway in differentiating myoblasts that links cell-cell adhesion via N-cadherin to Igf2 expression (assessed using northern and promoter-reporter analyses) via RhoA and p38alpha and/or beta but not gamma. We thus define a regulatory mechanism for p38 activation that relates cell-cell-derived adhesion signalling to the synthesis of the major fetal growth factor, IGF-II.


Subject(s)
Cell Adhesion/physiology , Cell Differentiation , Insulin-Like Growth Factor II/metabolism , Myoblasts/cytology , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cadherins/metabolism , Cell Count , Cells, Cultured , Isoenzymes/metabolism , Mice , Muscle Development , Mutant Proteins/metabolism , Myoblasts/metabolism , Signal Transduction
18.
Cancer Res ; 66(22): 10878-84, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108124

ABSTRACT

Insulin-like growth factor (IGF) binding protein-3 (IGFBP-3) promotes apoptosis of cancer cells by both IGF-dependent and IGF-independent mechanisms. In vitro phosphorylation of IGFBP-3 by DNA-dependent protein kinase (DNA-PK) has been reported but with unknown functional relevance. Using a chemical inhibitor for DNA-PK in prostate cancer cells and a paired system of glioblastoma cell lines that either lack or express DNA-PK, we show that the apoptosis-promoting and growth-inhibitory actions of IGFBP-3 are completely abrogated in the absence of catalytically active DNA-PK. In the absence of DNA-PK activity, IGFBP-3 has reduced nuclear accumulation and is unable to bind its nuclear binding partner retinoid X receptor (RXR) alpha. We assessed the importance of the three potential DNA-PK phosphorylation sites in IGFBP-3 using PCR-based site-directed mutagenesis. When transfected into 22RV1 cells, IGFBP-3-S165A and IGFBP-3-T170A functioned in an identical manner to wild-type IGFBP-3 to induce apoptosis. In contrast, IGFBP-3-S156A was unable to promote apoptosis and exhibited reduced nuclear accumulation, suggesting a key role for DNA-PK-dependent phosphorylation in the regulation of IGFBP-3 action. These studies reveal a novel regulatory mechanism for the actions of IGFBP-3 in prostate cancer and show phosphorylation of Ser(156) to be functionally critical in its apoptosis-inducing actions.


Subject(s)
Apoptosis/physiology , DNA-Activated Protein Kinase/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Prostatic Neoplasms/metabolism , Binding Sites , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Male , Phosphorylation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Retinoid X Receptor alpha/metabolism
19.
J Cell Sci ; 117(Pt 9): 1737-46, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15075235

ABSTRACT

Igfbp5 is upregulated during the differentiation of several key cell lineages and in some tumours; the function of IGFBP-5 in these physiological and pathological situations is unknown. Since IGFBP-5 contains sequence motifs consistent with IGF-independent actions, the aim of these studies was to distinguish between IGF-dependent and -independent actions of IGFBP-5. Myc-tagged wild-type (termed wtIGFBP-5) and non-IGF binding mouse Igfbp5 (termed mutIGFBP-5) cDNAs were generated and used to transfect C2 myoblasts, a cell line that undergoes differentiation to myotubes in an IGF- and IGFBP-5-regulated manner. WtIGFBP-5, but not mutIGFBP-5, inhibited myogenesis, as assessed by cell morphology, MHC immunocytochemistry and caveolin 3 expression. However, both wt- and mutIGFBP-5 increased cell survival and decreased apoptosis, as indicated by decreased caspase-3 activity and cell surface annexin V binding. Further examination of apoptotic pathways revealed that wt- and mutIGFBP-5 ameliorated the increase in caspase-9 but not the modest increase in caspase-8 during myogenesis, suggesting that IGFBP-5 increased cell survival via inhibition of intrinsic cell death pathways in an IGF-independent manner. The relationship between IGF-II and IGFBP-5 was examined further by cotransfecting C2 myoblasts with antisense Igf2 (previously established to induce increased cell death) and Igfbp5; both wt- and mutIGFBP-5 conferred equivalent protection against the decreased cell survival and increased apoptosis. In conclusion, we have partitioned IGFBP-5 action in myogenesis into IGF-dependent inhibition of differentiation and IGF-independent cell survival. Our findings suggest that, by regulation of cell survival, IGFBP-5 has an autonomous role in the regulation of cell fate in development and in tumourigenesis.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 5/metabolism , Insulin-Like Growth Factor II/metabolism , Muscle Development , Animals , Caspase 3 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/metabolism , Cell Count , Cell Line , Enzyme Activation , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor II/genetics , Mice , Myosin Heavy Chains/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
20.
Mol Cell Biol ; 24(9): 3607-22, 2004 May.
Article in English | MEDLINE | ID: mdl-15082758

ABSTRACT

Activation of either the phosphatidylinositol 3-kinase (PI 3-kinase)/Akt or the p38 mitogen-activated protein kinase (MAPK) signaling pathways accelerates myogenesis but only when the reciprocal pathway is functional. We therefore examined the hypothesis that cross-activation between these signaling cascades occurs to orchestrate myogenesis. We reveal a novel and reciprocal cross-talk and activation between the PI 3-kinase/Akt and p38 MAPK pathways that is essential for efficient myoblast differentiation. During myoblast differentiation, Akt kinase activity correlated with S473 but not T308 phosphorylation and occurred 24 h after p38 activation. Inhibition or activation of p38 with SB203580, dominant-negative p38, or MKK6EE regulated Akt kinase activity. Analysis of Akt isoforms revealed a specific increase in Akt2 protein levels that coincided with AktS473 phosphorylation during myogenesis and an enrichment of S473-phosphorylated Akt2. Akt2 promoter activity and protein levels were regulated by p38 activation, thus providing a mechanism for communication. Subsequent Akt activation by S473 phosphorylation was PI 3-kinase dependent and specific for Akt2 rather than Akt1. Complementary to p38-mediated transactivation of Akt, activation or inhibition of PI 3-kinase regulated p38 activity upstream of MKK6, demonstrating reciprocal communication and positive feedback characteristic of myogenic regulation. Our findings have identified novel communication between p38 MAPK and PI 3-kinase/Akt via Akt2.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Muscle Development/physiology , Muscle, Skeletal/growth & development , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Enzyme Activation , Enzyme Inhibitors/metabolism , Genes, Reporter , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mitogen-Activated Protein Kinases/genetics , Muscle, Skeletal/cytology , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...