Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Neurosci Res ; 98(5): 902-920, 2020 05.
Article in English | MEDLINE | ID: mdl-31840315

ABSTRACT

To understand the cellular basis for the neurodevelopmental effects of intrauterine growth restriction (IUGR), we examined the global and regional expression of various cell types within murine (Mus musculus) fetal brain. Our model employed maternal calorie restriction to 50% daily food intake from gestation day 10-19, producing IUGR offspring. Offspring had smaller head sizes with larger head:body ratios indicating a head sparing IUGR effect. IUGR fetuses at embryonic day 19 (E19) had reduced nestin (progenitors), ß-III tubulin (immature neurons), Glial fibrillary acidic protein (astrocytes), and O4 (oligodendrocytes) cell lineages via immunofluorescence quantification and a 30% reduction in cortical thickness. No difference was found in Bcl-2 or Bax (apoptosis) between controls and IUGR, though qualitatively, immunoreactivity of doublecortin (migration) and Ki67 (proliferation) was decreased. In the interest of examining a potential therapeutic peptide, we next investigated a novel pro-survival peptide, mouse Humanin (mHN). Ontogeny examination revealed highest mHN expression at E19, diminishing by postnatal day 15 (P15), and nearly absent in adult (3 months). Subanalysis by sex at E19 yielded higher mHN expression among males during fetal life, without significant difference between sexes postnatally. Furthermore, female IUGR mice at E19 had a greater increase in cortical mHN versus the male fetus over their respective controls. We conclude that maternal dietary restriction-associated IUGR interferes with neural progenitors differentiating into the various cellular components populating the cerebral cortex, and reduces cerebral cortical size. mHN expression is developmental stage and sex specific, with IUGR, particularly in the females, adaptively increasing its expression toward mediating a pro-survival approach against nutritional adversity.


Subject(s)
Cerebral Cortex/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Maternal Nutritional Physiological Phenomena/physiology , Neurons/metabolism , Animals , Caloric Restriction , Female , Fetal Growth Retardation/metabolism , Glial Fibrillary Acidic Protein/metabolism , Male , Mice
2.
PLoS One ; 13(3): e0193583, 2018.
Article in English | MEDLINE | ID: mdl-29590129

ABSTRACT

BACKGROUND: Intrauterine growth restriction (IUGR) results from a lack of nutrients transferred to the developing fetus, particularly oxygen and glucose. Increased expression of the cytoprotective mitochondrial peptide, humanin (HN), and the glucose transporter 8, GLUT8, has been reported under conditions of hypoxic stress. However, the presence and cellular localization of HN and GLUT8 in IUGR-related placental pathology remain unexplored. Thus, we undertook this study to investigate placental expression of HN and GLUT8 in IUGR-affected versus normal pregnancies. RESULTS: We found 1) increased HN expression in human IUGR-affected pregnancies on the maternal aspect of the placenta (extravillous trophoblastic (EVT) cytoplasm) compared to control (i.e. appropriate for gestational age) pregnancies, and a concomitant increase in GLUT8 expression in the same compartment, 2) HN and GLUT8 showed a protein-protein interaction by co-immunoprecipitation, 3) elevated HN and GLUT8 levels in vitro under simulated hypoxia in human EVT cells, HTR8/SVneo, and 4) increased HN expression but attenuated GLUT8 expression in vitro under serum deprivation in HTR8/SVneo cells. CONCLUSIONS: There was elevated HN expression with cytoplasmic localization to EVTs on the maternal aspect of the human placenta affected by IUGR, also associated with increased GLUT8 expression. We found that while hypoxia increased both HN and GLUT8, serum deprivation increased HN expression alone. Also, a protein-protein interaction between HN and GLUT8 suggests that their interaction may fulfill a biologic role that requires interdependency. Future investigations delineating molecular interactions between these proteins are required to fully uncover their role in IUGR-affected pregnancies.


Subject(s)
Fetal Growth Retardation/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Placenta/metabolism , Adult , Cytoplasm/metabolism , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Glucose Transport Proteins, Facilitative/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Placenta/pathology , Pregnancy , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trophoblasts/pathology , Up-Regulation
3.
Pediatr Blood Cancer ; 64(7)2017 Jul.
Article in English | MEDLINE | ID: mdl-28371314

ABSTRACT

This is a case presentation describing a high insulin requirement that suddenly resolved in a patient with acute lymphoblastic leukemia treated with stem cell transplantation complicated by chronic graft-versus-host disease. The patient was diagnosed with acquired partial lipodystrophy that did not require alternative therapies such as leptin or insulin-like growth factor 1.


Subject(s)
Graft vs Host Disease/complications , Hematopoietic Stem Cell Transplantation/adverse effects , Lipodystrophy/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Child , Chronic Disease , Female , Humans , Hypertriglyceridemia/etiology , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Lipodystrophy/drug therapy
4.
Reprod Health ; 13(1): 121, 2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27662834

ABSTRACT

BACKGROUND: An increased incidence of central precocious puberty has been recently reported in South Korea, which suggests an ongoing downward trend in pubertal development in the Korean population. We aimed to verify the trend in age at menarche in young Korean women during the last decade and associated factors. METHODS: We analyzed a population-based sample of 3409 Korean girls, aged 10-18 years, using data from the Korean National Health and Nutrition Examination Surveys (KNHANES) II (2001), III (2005), IV (2007-2009), and V (2010 and 2011). Average age at menarche was studied using the Kaplan-Meier survival method and predictors were analyzed using Cox proportional hazards model. The percentage of subjects who had experienced menarche at each age level was compared by using the Cochran-Armitage test. RESULTS: Overall mean age at menarche was 12.7 years. The percentage of subjects who experienced menarche before the age of 12 years was 21.4 % in 2001 but increased to 34.6 % in 2010/2011 (p < 0.01). In addition, the percentage of girls who experienced menarche before the age of 14 years increased from 76 % in 2001 to 92 % in 2010/2011 (p < 0.005). Adolescents whose mothers who had experienced early menarche (HR 1.48, 95 % CI [1.22-1.80]), and adolescents who were overweight (HR 1.24, 95 % CI [1.04-1.49]) were more likely to have experienced menarche. Additionally, underweight adolescents (HR 0.27, 95 % CI [0.12-0.60]) and adolescents who had a mother having late menarche (HR 0.68, 95 % CI [0.59-0.79]) were expected to have late menarche. None of the socioeconomic factors assessed in our study showed an association with age at menarche. CONCLUSIONS: A downward trend in age at menarche was defined in Korean adolescents during the last decade. Furthermore, influences of genetic and nutritional parameters on individual variance in age at menarche were defined.

5.
Eur J Endocrinol ; 174(6): C1-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27009113

ABSTRACT

OBJECTIVE: The Growth Hormone (GH) Research Society (GRS) convened a workshop to address important issues regarding trial design, efficacy, and safety of long-acting growth hormone preparations (LAGH). PARTICIPANTS: A closed meeting of 55 international scientists with expertise in GH, including pediatric and adult endocrinologists, basic scientists, regulatory scientists, and participants from the pharmaceutical industry. EVIDENCE: Current literature was reviewed for gaps in knowledge. Expert opinion was used to suggest studies required to address potential safety and efficacy issues. CONSENSUS PROCESS: Following plenary presentations summarizing the literature, breakout groups discussed questions framed by the planning committee. Attendees reconvened after each breakout session to share group reports. A writing team compiled the breakout session reports into a draft document that was discussed and revised in an open forum on the concluding day. This was edited further and then circulated to attendees from academic institutions for review after the meeting. Participants from pharmaceutical companies did not participate in the planning, writing, or in the discussions and text revision on the final day of the workshop. Scientists from industry and regulatory agencies reviewed the manuscript to identify any factual errors. CONCLUSIONS: LAGH compounds may represent an advance over daily GH injections because of increased convenience and differing phamacodynamic properties, providing the potential for improved adherence and outcomes. Better methods to assess adherence must be developed and validated. Long-term surveillance registries that include assessment of efficacy, cost-benefit, disease burden, quality of life, and safety are essential for understanding the impact of sustained exposure to LAGH preparations.


Subject(s)
Dwarfism, Pituitary/drug therapy , Hormone Replacement Therapy/methods , Human Growth Hormone/therapeutic use , Clinical Trials as Topic , Consensus , Hormone Replacement Therapy/adverse effects , Human Growth Hormone/adverse effects , Humans , Research Design
6.
PLoS One ; 10(11): e0141844, 2015.
Article in English | MEDLINE | ID: mdl-26539988

ABSTRACT

BACKGROUND: Few studies have explored the trends in central precocious puberty (CPP) in Asian populations. This study assessed the prevalence and annual incidence of CPP among Korean children. METHODS: Using data from the Korean Health Insurance Review Agency from 2004 to 2010, we reviewed the records of 21,351 children, including those registered with a diagnosis of CPP for the first time and those diagnosed with CPP who were treated with gonadotropin-releasing hormone analogs. RESULTS: The prevalence of CPP was 55.9 per 100,000 girls and 1.7 per 100,000 boys, respectively. The overall incidence of CPP was 15.3 per 100,000 girls, and 0.6 per 100,000 boys. The annual incidence of CPP in girls significantly increased from 3.3 to 50.4 per 100,000 girls; whereas in boys, it gradually increased from 0.3 to 1.2 per 100,000 boys. The annual incidence of CPP in girls consistently increased at all ages year by year, with greater increases at older ages (≥6 years of age), and smaller increases in girls aged < 6 years. In contrast, the annual incidence remained relatively constant in boys aged < 8 years, while a small increase was observed only in boys aged 8 years. The increase of annual incidence showed significant differences depending on age and gender (P <0.0001). CONCLUSIONS: The annual incidence of CPP has substantially increased among Korean girls over the past 7 years. Continued monitoring of CPP trends among Korean children will be informative.


Subject(s)
Puberty, Precocious/epidemiology , Asian People , Child , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Incidence , Korea/epidemiology , Male , Prevalence
7.
Mol Cell Endocrinol ; 413: 96-100, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26116236

ABSTRACT

Evidence for the putative mitochondrial origin of the Humanin (HN) peptide has been lacking, although its cytoprotective activity has been demonstrated in a variety of organismal and cellular systems. We sought to establish proof-of-principle for a mitochondria-derived peptide (MDP) in a rat-derived cellular system as the rat HN sequence is predicted to lack nuclear insertions of mitochondrial origin (NUMT). We found that the rat HN (Rattin; rHN) homologue is derived from the mitochondrial genome as evidenced by decreased production in Rho-0 cells, and that peptide translation occurs in the mitochondria as it is unaffected by cycloheximide. Rat HN localizes to the mitochondria in cellular subfractionation and immunohistochemical studies. Addition of a HN analogue to isolated mitochondria from rat INS-1 beta cells reduced hydrogen peroxide production by 55%. In summary, a locally bioactive peptide is derived and translated from an open reading frame (ORF) within rat mitochondrial DNA encoding 16S rRNA.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Line, Tumor , Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Rats
8.
Horm Cancer ; 4(1): 12-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23011725

ABSTRACT

Insulin-like growth factor binding protein-3 (IGFBP-3) is a pro-apoptotic, anti-metastasic, and anti-angiogenic protein. Low serum IGFBP-3 has been associated with risk of more aggressive prostate cancer (PCa). We investigated the impact of nuclear and cytoplasmic IGFBP-3 protein expression levels in PCa by examining their in situ expression across a wide spectrum of primary tumors by immunohistochemical analysis of tissue microarrays. Immunohistochemistry was performed on PCa microarrays constructed from 226 hormone naïve patients who underwent radical prostatectomy. Both cytoplasmic and nuclear IGFBP-3 expressions were scored in a semi-quantitative fashion using an integrated measure of intensity and positivity. The distribution of IGFBP-3 protein expression was examined across the spectrum of epithelial tissues, and its association with standard clinicopathological covariates and tumor recurrence was examined. There was a broad range of IGFBP-3 staining across all histologies examined. Tumor had higher IGFBP-3 cytoplasmic and nuclear staining than benign histologies. For IGFBP-3 nuclear staining, PCa was significantly different than benign prostatic hyperplasia, normal prostate, and prostate intraepithelial neoplasia. As both a continuous and dichotomized variable, higher nuclear IGFBP-3 expression had statistically significant associations with PCa recurrence. The cytoplasmic staining had no significance in any patient subgroup. In patients with low-grade cancer, IGFBP-3 nuclear positivity was a better predictor of recurrence than baseline PSA, tumor margin status, TNM tumor stage, or presence of capsular invasion. High nuclear IGFBP-3 is amongst the strongest predictors of cancer recurrence in patients with low-grade prostate cancers and may therefore play an important role in risk stratification.


Subject(s)
Cell Nucleus/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Neoplasm Recurrence, Local/metabolism , Prostatic Neoplasms/metabolism , Aged , Cytoplasm/metabolism , Epithelium/metabolism , Epithelium/pathology , Humans , Immunohistochemistry/methods , Insulin-Like Growth Factor Binding Protein 3/genetics , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prostate/metabolism , Prostate/pathology , Prostate/surgery , Prostatectomy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Retrospective Studies
9.
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
10.
Endocr Relat Cancer ; 17(2): 293-302, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20228135

ABSTRACT

Tumor suppression by IGF-binding protein 3 (IGFBP3) may occur in an IGF-independent manner, in addition to its role as a regulator of IGF bioavailability. After secretion, IGFBP3 is internalized, rapidly localized to the nucleus, and is later detected in the cytoplasm. We identified a putative nuclear export sequence (NES) in IGFBP3 between amino acids 217 and 228, analogous to the leucine-rich NES sequence of p53 and HIV Rev. Mutation of the NES prevents nucleocytoplasmic shuttling of IGFBP3 and blocks its ability to induce apoptosis. Targeting of IGFBP3 to the mitochondria and endoplasmic reticulum (ER) was confirmed by co-localization with organelle markers using fluorescence confocal microscopy and subcellular fractionation. Mitochondrial targeting was also demonstrated in vivo in IGFBP3-treated prostate cancer xenografts. These results show that IGFBP3 shuttles from the nucleus to the mitochondria and ER, and that nuclear export is essential for its effects on prostate cancer apoptosis.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Humans , Male , Mice , Mice, SCID , Nuclear Export Signals , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
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
12.
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
13.
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
14.
Am J Physiol Cell Physiol ; 296(5): C954-76, 2009 May.
Article in English | MEDLINE | ID: mdl-19279229

ABSTRACT

Insulin-like growth factor (IGF) binding protein (IGFBP)-3 has traditionally been defined by its role as a binding protein and its association with IGF delivery and availability. Development of non-IGF binding IGFBP-3 analogs and the use of cell lines devoid of type 1 IGF receptors (IGF-R) have led to critical advances in the field of IGFBP-3 biology. These studies show that IGFBP-3 has IGF-independent roles in inhibiting cell proliferation in cancer cell lines. Nuclear transcription factor, retinoid X receptor (RXR)-alpha, and IGFBP-3 functionally interact to reduce prostate tumor growth and prostate-specific antigen in vivo. Moreover, IGFBP-3 inhibits insulin-stimulated glucose uptake into adipocytes independent of IGF. The purpose of this review is to highlight IGFBP-3 as a novel effector molecule and not just another "binding protein" by discussing its IGF-independent actions on metabolism and cell growth. Although this review presents studies that assume the role of IGFBP-3 as either an endocrine or autocrine/paracrine molecule, these systems may not exist as distinct entities, justifying the examination of IGFBP-3 in an integrated model. Also, we provide an overview of factors that regulate IGFBP-3 availability, including its production, methylation, and ubiquitination. We conclude with the role of IGFBP-3 in whole body systems and possible future applications of IGFBP-3 in physiology.


Subject(s)
Autocrine Communication/physiology , Endocrine System/physiology , Insulin-Like Growth Factor Binding Protein 3/physiology , Paracrine Communication/physiology , Animals , Cell Division/physiology , Humans , Mammals , Neoplasms/physiopathology
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.
Pediatr Res ; 61(2): 159-64, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17237715

ABSTRACT

Insulin-like growth factor binding protein (IGFBP)-3 binds to IGF and modulates their actions and also possesses intrinsic activities. We investigated its effects on insulin action and found that when IGFBP-3 was added to fully differentiated 3T3-L1 adipocytes in culture, insulin-stimulated glucose transport was significantly inhibited to 60% of control in a time- and dose-dependent manner. Tumor necrosis factor (TNF)-alpha treatment also inhibited glucose transport to the same degree as IGFBP-3 and, in addition, increased IGFBP-3 levels 3-fold. Co-treatment with TNF-alpha and IGFBP-3 antisense partially prevented the inhibitory effect of TNF-alpha on glucose transport, indicating a role for IGFBP-3 in cytokine-induced insulin resistance. Insulin-stimulated phosphorylation of the insulin receptor was markedly decreased by IGFBP-3 treatment. IGFBP-3 treatment suppressed adiponectin expression in 3T3-L1 adipocytes. Infusion of IGFBP-3 to Sprague-Dawley rats for 3 h decreased peripheral glucose uptake by 15% compared with controls as well as inhibiting glycogen synthesis. Systemic administration of IGFBP-3 to rats for 7 d resulted in a dramatic 40% decrease in peripheral glucose utilization and glycogen synthesis. These in vitro and in vivo findings demonstrate that IGFBP-3 has potent insulin-antagonizing capability and suggest a role for IGFBP-3 in cytokine-induced insulin resistance and other mechanisms involved in the development of type-2 diabetes.


Subject(s)
Adipocytes/metabolism , Insulin Resistance , Insulin-Like Growth Factor Binding Protein 3/metabolism , 3T3-L1 Cells , Adiponectin/antagonists & inhibitors , Animals , Glucose/metabolism , Humans , Mice , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
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.
Clin Cancer Res ; 11(13): 4851-6, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-16000583

ABSTRACT

We have previously identified the retinoid X receptor-alpha (RXRalpha) as an insulin-like growth factor binding protein-3 (IGFBP-3) nuclear binding partner, which is required for IGFBP-3-induced apoptosis. In the current study, we investigated the biological interactions of the RXR ligand, VTP194204 and rhIGFBP-3, in vitro and in vivo. In vitro, IGFBP-3 and VTP194204 individually induced apoptosis, and suppressed cell growth in prostate cancer cell lines in an additive manner. In vivo, LAPC-4 xenograft-bearing severe combined immunodeficiency mice treated daily with saline, IGFBP-3, and/or VTP194204 for 3 weeks showed no effect of individual treatments with IGFBP-3 or VTP194204 on tumor growth. However, the combination of IGFBP-3 and VTP194204 treatments inhibited tumor growth by 50% and induced a significant reduction in serum prostate-specific antigen levels. In terminal nucleotidyl transferase-mediated nick end labeling immunohistochemistry of LAPC-4 xenografts, there was modest induction of apoptosis with either IGFBP-3 or VTP194204 individual treatment, but combination therapy resulted in massive cell death, indicating that IGFBP-3 and VTP194204 have a synergistic effect in preventing tumor growth by apoptosis induction. In summary, this is an initial description of the successful therapeutic use of IGFBP-3 as a cancer therapy in vivo, and shows that combination treatment of IGFBP-3 and RXR ligand has a synergistic effect on apoptosis induction leading to substantial inhibition of prostate cancer xenograft growth. Taken together, these observations suggest that combination therapy with IGFBP-3 and RXR ligands may have therapeutic potential for prostate cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Prostatic Neoplasms/drug therapy , Retinoids/pharmacology , Animals , Cell Line, Tumor , Drug Synergism , Humans , Insulin-Like Growth Factor Binding Protein 3/administration & dosage , Ligands , Male , Mice , Mice, SCID , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Retinoid X Receptors/metabolism , Retinoids/administration & dosage , Retinoids/metabolism , Time Factors , Xenograft Model Antitumor Assays
20.
J Biol Chem ; 280(17): 16942-8, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15731112

ABSTRACT

Insulin-like growth factor-binding protein-3 (IGFBP-3) induces apoptosis by its ability to bind insulin-like growth factors (IGFs) as well as its IGF-independent effects involving binding to other molecules including the retinoid X receptor-alpha (RXRalpha). Here we describe that in response to IGFBP-3, the RXRalpha binding partner nuclear receptor Nur77 rapidly undergoes translocation from the nucleus to the mitochondria, initiating an apoptotic cascade resulting in caspase activation within 6 h. This translocation is a type 1 IGF receptor-signaling independent event as IGFBP-3 induces Nur77 translocation in R-cells. IGFBP-3 and Nur77 are additive in inducing apoptosis. GFP-Nur77 transfection into RXRalpha wild-type and knock-out mouse embryonic fibroblasts and subsequent treatment with IGFBP-3 show that RXRalpha is required for IGFBP-3-induced Nur77 translocation and apoptosis. Addition of IGFBP-3 to 22RV1 cell lysates enhanced the ability of GST-RXRalpha to "pull down" Nur77, and overexpression of IGFBP-3 enhanced the accumulation of mitochondrial RXRalpha. This unique nongenotropic nuclear pathway supports an emerging role for IGFBP-3 as a novel, multicompartmental signaling molecule involved in induction of apoptosis in malignant cells.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Insulin-Like Growth Factor Binding Protein 3/physiology , Mitochondria/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Retinoid X Receptor alpha/metabolism , Somatomedins/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , Blotting, Western , Caspases/metabolism , Cells, Cultured , Cytoplasm/metabolism , Densitometry , Dimerization , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Glutathione Transferase/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mice , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nuclear Receptor Subfamily 4, Group A, Member 1 , Protein Transport , Signal Transduction , Time Factors , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL