Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
J Neuroendocrinol ; 33(9): e13010, 2021 09.
Article in English | MEDLINE | ID: mdl-34312927

ABSTRACT

Nesfatin-1, an 82 amino acid peptide cleaved from the N-terminal of its precursor nucleobindin-2 (NUCB2), is emerging as a multifunctional peptide in fish. The present study aimed to determine whether nesfatin-1 plays a role in fish somatic growth by modulating the growth hormone (GH)/insulin-like growth factor (IGF) axis, using a representative teleost model, the goldfish (Carassius auratus). The results demonstrated that a single i.p. injection of synthetic goldfish nesfatin-1 significantly decreased the expression of hypothalamic pacap (approximately 90%) and pituitary Gh (approximately 90%) mRNAs at 15 minutes post-injection. Serum GH levels were also reduced as a result of nesfatin-1 administration, by approximately 45% and 55% at 15 and 30 minutes post-injection, respectively. Likewise, in vitro treatment of goldfish dispersed pituitary cells with nesfatin-1 reduced Gh secretion, suggesting that nesfatin-1 acts directly on pituitary somatotrophs to inhibit Gh release. Exposure of cultured liver fragments to nesfatin-1 (0.1, 1 and 10 nmol L-1 ) led to a significant reduction in igf-1 mRNA at 120 minutes and of igf-II mRNA at 30 and 60 minutes post-incubation. Collectively, these results indicate a suppressive role for nesfatin-1 on the goldfish GH/IGF axis. Immunohistochemical studies demonstrated that NUCB2/nesfatin-1-like immunoreactivity, although present in the goldfish pituitary, is not colocalised with GH in goldfish somatotrophs. Thus, nesfatin-1 does not appear to act in an autocrine manner to regulate GH secretion. Taken together, this research found that the pituitary gland is an important source of endogenous NUCB2/nesfatin-1 and also that nesfatin-1 directly suppresses the Gh/IGF axis in goldfish.


Subject(s)
Growth Hormone/antagonists & inhibitors , Nucleobindins/pharmacology , Somatomedins/antagonists & inhibitors , Animals , Cells, Cultured , Female , Gene Expression/drug effects , Goldfish , Growth Hormone/metabolism , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Nucleobindins/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Somatomedins/metabolism
2.
Anticancer Drugs ; 32(7): 734-744, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34001703

ABSTRACT

Curcumin has a vital role in the development of renal carcinoma. Nevertheless, the mechanism of curcumin in renal carcinoma tumorigenesis remains largely unknown. Thirty renal carcinoma patients were recruited. Renal carcinoma cell lines CAKI-1 and ACHN were exposed to curcumin. The levels of circular RNA fibronectin type III domain-containing protein 3B (circ-FNDC3B), microRNA (miR)-138-5p and insulin-like growth factor 2 (IGF2) were detected via quantitative reverse transcription PCR or western blot. Cell proliferation and apoptosis were investigated via 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide, colony formation analysis, flow cytometry and western blot. Target association between miR-138-5p and circ-FNDC3B or IGF2 was analyzed via dual-luciferase reporter analysis. The function of curcumin in vivo was assessed via a xenograft model. circ-FNDC3B level was enhanced and miR-138-5p abundance was declined in renal carcinoma tissues and cells. Curcumin restrained renal carcinoma cell proliferation and promoted apoptosis. circ-FNDC3B overexpression or miR-138-5p knockdown weakened the influence of curcumin. circ-FNDC3B knockdown hindered cell proliferation and promoted apoptosis by increasing miR-138-5p. IGF2 was targeted via miR-138-5p and positively regulated via circ-FNDC3B. Curcumin decreased xenograft tumor growth via reducing circ-FNDC3B in vivo. Curcumin suppressed renal carcinoma tumorigenesis in vitro and in vivo via regulating circ-FNDC3B/miR-138-5p/IGF2 axis, proposing new insight into renal carcinoma tumorigenesis.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Curcumin/pharmacology , Fibronectins/drug effects , Insulin-Like Growth Factor II/drug effects , Kidney Neoplasms/drug therapy , MicroRNAs/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
J Cell Physiol ; 234(12): 21860-21876, 2019 12.
Article in English | MEDLINE | ID: mdl-31066045

ABSTRACT

Mesenchymal stem cells (MSCs) transplantation has emerged as a potential therapeutic approach for Alzheimer's disease (AD). However, the poor proliferation capacity and low survival rate of engrafted MSCs in the hostile microenvironment of AD limit their therapeutic efficiency. Lin28B is a conserved RNA-binding protein associated with cell self-renewal and survival. The purpose of the present study was to explore whether lin28B might influence the functions of implanted MSCs and strengthen their neuroprotective potential in AD. A gain-of-function assay was used to upregulate lin28B expression in MSCs by lentiviral transfection. Our in vitro results indicated that lin28B promoted MSCs proliferation and migration, and protected MSCs against Aß1-42-induced cell death by upregulating insulin-like growth factor-2 (IGF-2). Blockage of IGF-2 partially abrogated the above effects of lin28B. After intracerebroventricular injection into amyloid precursor protein/presenilin 1 mice, implanted MSCs were monitored using bioluminescence imaging. We observed that administration of MSCs transfected with lin28B significantly stimulated their proliferation and prolonged cell retention after delivery. Moreover, administration of the transfected MSCs markedly mitigated cognitive deficits, promoted amyloid plaque clearance, decreased the activation of microglia, and reduced neuronal cell death. The data above confirmed our hypothesis that lin28B is a crucial modulator determining the fate of transplanted MSCs by regulating IGF-2-associated pathways and thereby enhancing their protective effects against AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Insulin-Like Growth Factor II/metabolism , Mesenchymal Stem Cells , RNA-Binding Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Animals , Disease Models, Animal , Insulin-Like Growth Factor II/drug effects , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mice , Transcriptional Activation/physiology
4.
Neurobiol Dis ; 110: 218-230, 2018 02.
Article in English | MEDLINE | ID: mdl-29113829

ABSTRACT

The selective vulnerability of motor neurons in amyotrophic lateral sclerosis (ALS) is evident by sparing of a few subpopulations during this fast progressing and debilitating degenerative disease. By studying the gene expression profile of resilient vs. vulnerable motor neuron populations we can gain insight in what biomolecules and pathways may contribute to the resilience and vulnerability. Several genes have been found to be differentially expressed in the vulnerable motor neurons of the cervical spinal cord as compared to the spared motor neurons in CNIII/IV. One gene that is differentially expressed and present at higher levels in less vulnerable motor neurons is insulin-like growth factor II (IGF-II). The motor neuron protective effect of IGF-II has been demonstrated both in vitro and in SOD1 transgenic mice. Here, we have screened a library of small molecule compounds and identified inducers of IGF-II mRNA and protein expression. Several identified compounds significantly protected motor neurons from glutamate excitotoxicity in vitro. One of the compounds, vardenafil, resulted in a complete motor neuron protection, an effect that was reversed by blocking receptors of IGF-II. When administered to naïve rats vardenafil was present in the cerebrospinal fluid and increased IGF-II mRNA expression in the spinal cord. When administered to SOD1 transgenic mice, there was a significant delay in motor symptom onset and prolonged survival. Vardenafil also increased IGF-II mRNA and protein levels in motor neurons derived from healthy subject and ALS patient iPSCs, activated a human IGF-II promoter and improved survival of ALS-patient derived motor neurons in culture. Our findings suggest that modulation of genes differentially expressed in vulnerable and resilient motor neurons may be a useful therapeutic approach for motor neuron disease.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Insulin-Like Growth Factor II/biosynthesis , Motor Neurons/drug effects , Motor Neurons/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Vardenafil Dihydrochloride/pharmacology , Animals , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Insulin-Like Growth Factor II/drug effects , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley
5.
Cancer Treat Rev ; 60: 90-99, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28934637

ABSTRACT

The insulin-like growth factor (IGF) system comprises multiple growth factor receptors, including insulin-like growth factor 1 receptor (IGF-1R), insulin receptor (IR) -A and -B. These receptors are activated upon binding to their respective growth factor ligands, IGF-I, IGF-II and insulin, and play an important role in development, maintenance, progression, survival and chemotherapeutic response of ovarian cancer. In many pre-clinical studies anti-IGF-1R/IR targeted strategies proved effective in reducing growth of ovarian cancer models. In addition, anti-IGF-1R targeted strategies potentiated the efficacy of platinum based chemotherapy. Despite the vast amount of encouraging and promising pre-clinical data, anti-IGF-1R/IR targeted strategies lacked efficacy in the clinic. The question is whether targeting the IGF-1R/IR signaling pathway still holds therapeutic potential. In this review we address the complexity of the IGF-1R/IR signaling pathway, including receptor heterodimerization within and outside the IGF system and downstream signaling. Further, we discuss the implications of this complexity on current targeted strategies and indicate therapeutic opportunities for successful targeting of the IGF-1R/IR signaling pathway in ovarian cancer. Multiple-targeted approaches circumventing bidirectional receptor tyrosine kinase (RTK) compensation and prevention of system rewiring are expected to have more therapeutic potential.


Subject(s)
Antineoplastic Agents/therapeutic use , Insulin-Like Growth Factor II/drug effects , Molecular Targeted Therapy/methods , Ovarian Neoplasms/drug therapy , Female , Humans , Insulin-Like Growth Factor II/metabolism , Ovarian Neoplasms/metabolism
6.
Mol Cancer Ther ; 16(7): 1324-1334, 2017 07.
Article in English | MEDLINE | ID: mdl-28468775

ABSTRACT

Insulin receptor (InsR) and the type I insulin-like growth factor (IGF1R) are homologous receptors necessary for signal transduction by their cognate ligands insulin, IGF-I and IGF-II. IGF1R mAbs, intended to inhibit malignant phenotypic signaling, failed to show benefit in patients with endocrine-resistant tumors in phase III clinical trials. Our previous work showed that in tamoxifen-resistant cells, IGF1R expression was lacking, but InsR inhibition effectively blocked growth. In endocrine-sensitive breast cancer cells, insulin was not growth stimulatory, likely due to the presence of hybrid InsR/IGF1R, which has high affinity for IGF-I, but not insulin. Combination inhibition of InsR and IGF1R showed complete suppression of the system in endocrine-sensitive breast cancer cells. To develop InsR-binding agents, we employed a small protein scaffold, T7 phage gene 2 protein (Gp2) with the long-term goal of creating effective InsR inhibitors and diagnostics. Using yeast display and directed evolution, we identified three Gp2 variants (Gp2 #1, #5, and #10) with low nanomolar affinity and specific binding to cell surface InsR. These Gp2 variants inhibited insulin-mediated monolayer proliferation in both endocrine-sensitive and resistant breast cancer, but did not downregulate InsR expression. Gp2 #5 and Gp2 #10 disrupted InsR function by inhibiting ligand-induced receptor activation. In contrast, Gp2 #1 did not block InsR phosphorylation. Notably, Gp2 #1 binding was enhanced by pretreatment of cells with insulin, suggesting a unique receptor-ligand-binding mode. These Gp2 variants are the first nonimmunoglobulin protein scaffolds to target insulin receptor and present compelling opportunity for modulation of InsR signaling. Mol Cancer Ther; 16(7); 1324-34. ©2017 AACR.


Subject(s)
Antigens, CD/genetics , Breast Neoplasms/drug therapy , Receptor, Insulin/genetics , Receptors, Somatomedin/genetics , Repressor Proteins/administration & dosage , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/drug effects , Mice , Nuclear Matrix-Associated Proteins/administration & dosage , Nuclear Matrix-Associated Proteins/genetics , Protein Binding , Receptor, IGF Type 1 , Receptor, Insulin/antagonists & inhibitors , Receptors, Somatomedin/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Article in English | MEDLINE | ID: mdl-26212489

ABSTRACT

OBJECTIVES: Estetrol (E4) is a natural estrogen produced by the human fetal liver. In combination with drospirenone (DRSP) or levonorgestrel (LNG), E4 blocks ovulation and has less effect on haemostatic biomarkers in comparison with ethinylestradiol (EE) combined with DRSP. This study evaluates the impact of several doses of E4/DRSP and E4/LNG on safety parameters such as liver function, lipid metabolism, bone markers and growth endocrine parameters. METHODS: This was a dose-finding, single-centre, controlled study performed in healthy women aged 18 to 35 years with a documented pretreatment ovulatory cycle. Participants received 5 mg or 10 mg E4/3 mg DRSP; 5 mg, 10 mg or 20 mg E4/150 µg LNG; or 20 µg EE/3 mg DRSP as a comparator for three consecutive cycles in a 24/4-day regimen. Changes from baseline to end of treatment in liver parameters, lipid metabolism, bone markers and growth endocrinology were evaluated. RESULTS: A total of 109 women were included in the study. Carrier proteins were minimally affected in the E4/DRSP and E4/LNG groups, in comparison with the EE/DRSP group, where a significant increase in sex hormone-binding globulin was observed. Similarly, minor effects on lipoproteins were observed in the E4 groups, and the effects on triglycerides elicited by the E4 groups were significantly lower than those in the EE/DRSP group. No imbalances in bone markers were observed in any groups. No alterations in insulin-like growth factor were observed in the E4 groups. CONCLUSIONS: E4-containing combinations have a limited effect on liver function, lipid metabolism, and bone and growth endocrine parameters.


Subject(s)
Androstenes/pharmacology , Contraceptives, Oral, Combined/pharmacology , Estetrol/pharmacology , Ethinyl Estradiol/pharmacology , Levonorgestrel/pharmacology , Liver/drug effects , Adolescent , Adult , Alkaline Phosphatase/blood , Aspartate Aminotransferases/blood , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Collagen Type I/blood , Collagen Type I/drug effects , Contraceptives, Oral, Combined/pharmacokinetics , Contraceptives, Oral, Synthetic/pharmacology , Estetrol/pharmacokinetics , Estrogens/pharmacology , Female , Growth Hormone/blood , Growth Hormone/drug effects , Humans , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/metabolism , Lipid Metabolism/drug effects , Liver/physiology , Mineralocorticoid Receptor Antagonists/pharmacology , Osteocalcin/blood , Osteocalcin/drug effects , Peptides/blood , Peptides/drug effects , Sex Hormone-Binding Globulin/drug effects , Sex Hormone-Binding Globulin/metabolism , Triglycerides/blood , Young Adult , gamma-Glutamyltransferase/blood
8.
Endocrinology ; 156(7): 2632-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25924104

ABSTRACT

In the hippocampus, estrogens are powerful modulators of neurotransmission, synaptic plasticity and neurogenesis. In women, menopause is associated with increased risk of memory disturbances, which can be attenuated by timely estrogen therapy. In animal models of menopause, 17ß-estradiol (E2) replacement improves hippocampus-dependent spatial memory. Here, we explored the effect of E2 replacement on hippocampal gene expression in a rat menopause model. Middle-aged ovariectomized female rats were treated continuously for 29 days with E2, and then, the hippocampal transcriptome was investigated with Affymetrix expression arrays. Microarray data were analyzed by Bioconductor packages and web-based softwares, and verified with quantitative PCR. At standard fold change selection criterion, 156 genes responded to E2. All alterations but 4 were transcriptional activation. Robust activation (fold change > 10) occurred in the case of transthyretin, klotho, claudin 2, prolactin receptor, ectodin, coagulation factor V, Igf2, Igfbp2, and sodium/sulfate symporter. Classification of the 156 genes revealed major groups, including signaling (35 genes), metabolism (31 genes), extracellular matrix (17 genes), and transcription (16 genes). We selected 33 genes for further studies, and all changes were confirmed by real-time PCR. The results suggest that E2 promotes retinoid, growth factor, homeoprotein, neurohormone, and neurotransmitter signaling, changes metabolism, extracellular matrix composition, and transcription, and induces protective mechanisms via genomic effects. We propose that these mechanisms contribute to effects of E2 on neurogenesis, neural plasticity, and memory functions. Our findings provide further support for the rationale to develop safe estrogen receptor ligands for the maintenance of cognitive performance in postmenopausal women.


Subject(s)
Estradiol/pharmacology , Estrogen Replacement Therapy , Estrogens/pharmacology , Gene Expression/drug effects , Hippocampus/drug effects , Menopause/drug effects , RNA, Messenger/drug effects , Animals , Cation Transport Proteins/drug effects , Cation Transport Proteins/genetics , Claudins/drug effects , Claudins/genetics , Factor V/drug effects , Factor V/genetics , Female , Glucuronidase/drug effects , Glucuronidase/genetics , Insulin-Like Growth Factor Binding Protein 2/drug effects , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Intracellular Signaling Peptides and Proteins , Klotho Proteins , Models, Animal , Prealbumin/drug effects , Prealbumin/genetics , Proteins/drug effects , Proteins/genetics , RNA, Messenger/metabolism , Rats , Receptors, Prolactin/drug effects , Receptors, Prolactin/genetics , Sodium Sulfate Cotransporter , Symporters/drug effects , Symporters/genetics
9.
Endocrinology ; 156(7): 2395-408, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25885794

ABSTRACT

Medulloblastoma (Med) is the most common malignant brain tumor in children. The role of ESR2 [estrogen receptor (ER)-ß] in promoting Med growth was comprehensively examined in three in vivo models and human cell lines. In a novel Med ERß-null knockout model developed by crossing Esr2(-/-) mice with cerebellar granule cell precursor specific Ptch1 conditional knockout mice, the tumor growth rate was significantly decreased in males and females. The absence of Esr2 resulted in increased apoptosis, decreased B-cell lymphoma 2 (BCL2), and IGF-1 receptor (IGF1R) expression, and decreased levels of active MAPKs (ERK1/2) and protein kinase B (AKT). Treatment of Med in Ptch1(+/-) Trp53(-/-) mice with the antiestrogen chemotherapeutic drug Faslodex significantly increased symptom-free survival, which was associated with increased apoptosis and decreased BCL2 and IGF1R expression and signaling. Similar effects were also observed in nude mice bearing D283Med xenografts. In vitro studies in human D283Med cells metabolically stressed by glutamine withdrawal found that 17ß-estradiol and the ERß selective agonist 2,3-bis(4-hydroxyphenyl)-propionitrile dose dependently protected Med cells from caspase-3-dependent cell death. Those effects were associated with increased phosphorylation of IGF1R, long-term increases in ERK1/2 and AKT signaling, and increased expression of IGF-1, IGF1R, and BCL2. Results of pharmacological experiments revealed that the cytoprotective actions of estradiol were dependent on ERß and IGF1R receptor tyrosine kinase activity and independent of ERα and G protein-coupled estrogen receptor 1 (G protein coupled receptor 30). The presented results demonstrate that estrogen promotes Med growth through ERß-mediated increases in IGF1R expression and activity, which induce cytoprotective mechanisms that decrease apoptosis.


Subject(s)
Apoptosis/genetics , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic/genetics , Medulloblastoma/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Medulloblastoma/metabolism , Mice , Mice, Knockout , Patched Receptors , Patched-1 Receptor , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/drug effects , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
10.
Asian J Androl ; 16(5): 675-83, 2014.
Article in English | MEDLINE | ID: mdl-24713826

ABSTRACT

We aimed to determine the mechanisms of the anabolic actions of androgens in skeletal muscle by investigating potential androgen receptor (AR)-regulated genes in in vitro and in vivo models. The expression of the myogenic regulatory factor myogenin was significantly decreased in skeletal muscle from testosterone-treated orchidectomized male mice compared to control orchidectomized males, and was increased in muscle from male AR knockout mice that lacked DNA binding activity (AR(ΔZF2)) versus wildtype mice, demonstrating that myogenin is repressed by the androgen/AR pathway. The ubiquitin ligase Fbxo32 was repressed by 12 h dihydrotestosterone treatment in human skeletal muscle cell myoblasts, and c-Myc expression was decreased in testosterone-treated orchidectomized male muscle compared to control orchidectomized male muscle, and increased in AR(∆ZF2) muscle. The expression of a group of genes that regulate the transition from myoblast proliferation to differentiation, Tceal7 , p57(Kip2), Igf2 and calcineurin Aa, was increased in AR(∆ZF2) muscle, and the expression of all but p57(Kip2) was also decreased in testosterone-treated orchidectomized male muscle compared to control orchidectomized male muscle. We conclude that in males, androgens act via the AR in part to promote peak muscle mass by maintaining myoblasts in the proliferative state and delaying the transition to differentiation during muscle growth and development, and by suppressing ubiquitin ligase-mediated atrophy pathways to preserve muscle mass in adult muscle.


Subject(s)
Muscle Proteins/genetics , Myoblasts, Skeletal/metabolism , Myogenin/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , SKP Cullin F-Box Protein Ligases/genetics , Animals , Calcineurin/drug effects , Calcineurin/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p57/drug effects , Cyclin-Dependent Kinase Inhibitor p57/genetics , Dihydrotestosterone/pharmacology , Gene Expression/drug effects , Humans , In Vitro Techniques , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Male , Mice , Mice, Knockout , Muscle Proteins/drug effects , Muscle, Skeletal , Myoblasts, Skeletal/drug effects , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Orchiectomy , Proto-Oncogene Proteins c-myc/drug effects , RNA, Messenger/drug effects , SKP Cullin F-Box Protein Ligases/drug effects , Testosterone/pharmacology
11.
Pediatr Surg Int ; 30(2): 137-42, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24352370

ABSTRACT

BACKGROUND: The high mortality rate in congenital diaphragmatic hernia (CDH) is attributed to pulmonary hypoplasia (PH). Insulin-like growth factor 2 (IGF2) is an important regulator of fetal growth. The highest levels of IGF2 expression are found in the placenta, which are negatively regulated by decidual retinoid acid receptor alpha (RARα). It has been demonstrated that prenatal administration of retinoic acid (RA) suppresses decidual RARα expression. Previous studies have further shown that prenatal administration of RA can reverse PH in nitrofen-induced CDH model. In IGF2 knockout animals, low levels of IGF2 are associated with decreased placental growth and PH. We therefore hypothesized that nitrofen decreases trophoblastic IGF2 expression and prenatal administration of RA increases it through decidual RARα in the nitrofen-induced CDH model. METHODS: Pregnant rats were exposed to either olive oil or nitrofen on day 9 of gestation (D9). RA was given intraperitoneally on D18, D19 and D20. Fetuses were harvested on D21 and divided into three groups: control, CDH and nitrofen+RA. Immunohistochemistry was performed to evaluate decidual RARα and trophoblastic IGF2 expression. Protein levels of IGF2 in serum, intra-amniotic fluid and left lungs were measured by enzyme-linked immunosorbent assay. RESULTS: Significant growth retardation of placenta and left lungs was observed in the CDH group compared to control and nitrofen+RA group. Markedly increased decidual RARα and decreased IGF2 immunoreactivity were found in the CDH group compared to control and nitrofen+RA group. Significantly decreased IGF2 protein levels were detected in serum, intra-amniotic fluid and left lungs in the CDH group compared to control and nitrofen+RA group. CONCLUSION: Our findings suggest that nitrofen may disturb trophoblastic IGF2 expression through decidual RARα resulting in retarded placental growth and PH in the nitrofen-induced CDH. Prenatal administration of RA may promote lung and placental growth by increasing trophoblastic IGF2 expression.


Subject(s)
Antineoplastic Agents/pharmacology , Hernias, Diaphragmatic, Congenital , Insulin-Like Growth Factor II/metabolism , Tretinoin/pharmacology , Trophoblasts/metabolism , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Hernia, Diaphragmatic/embryology , Hernia, Diaphragmatic/genetics , Hernia, Diaphragmatic/metabolism , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Lung/drug effects , Lung/embryology , Lung/metabolism , Phenyl Ethers , Pregnancy , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Trophoblasts/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
12.
Res Vet Sci ; 94(3): 462-70, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23566927

ABSTRACT

The objective of this study was to evaluate the effects of a single intramammary infusion of Panax ginseng extract (GS) on insulin-like growth factors (IGF) in bovine mammary gland during early involution. Eight mammary quarters from six nonpregnant cows in late lactation were infused with 10 mL of ginseng extract solution (3 mg/mL), six quarters were treated with 10 mL of placebo (vehicle alone) and six quarters were maintained as uninoculated controls. Milking was interrupted after infusion. Concentrations of IGF1 in mammary secretions were higher in GS-treated quarters than in placebo and uninoculated control quarters at 24, 48 and 72 h post-treatment (p<0.05). Treatment with GS did not affect mammary secretion of IGF2 (p=0.942). At 7 d of post-lactational involution, a decrease of immunostained area and mRNA expression for IGF1 was observed in mammary tissue of GS-treated quarters compared with placebo-treated quarters and uninoculated controls (p<0.05). The IGF2 immunostained area and mRNA expression for this growth factor were not affected by GS treatment (p=0.216 and p=0.785, respectively). An increase in protein levels and mRNA expression in mammary tissue of IGFBP3, IGFBP4 and IGFBP5 was observed in GS-treated quarters compared with placebo-treated quarters and uninoculated controls (p<0.05). These results provide evidence that intramammary inoculation of GS extract at cessation of milking may promote early mammary involution through the inhibition of IGF1 local production and bioavailability.


Subject(s)
Mammary Glands, Animal/drug effects , Panax , Plant Extracts/pharmacology , Somatomedins/drug effects , Animals , Cattle , Female , Insulin-Like Growth Factor Binding Protein 3/analysis , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 3/drug effects , Insulin-Like Growth Factor Binding Protein 4/analysis , Insulin-Like Growth Factor Binding Protein 4/biosynthesis , Insulin-Like Growth Factor Binding Protein 4/drug effects , Insulin-Like Growth Factor Binding Protein 5/analysis , Insulin-Like Growth Factor Binding Protein 5/biosynthesis , Insulin-Like Growth Factor Binding Protein 5/drug effects , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor II/analysis , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/drug effects , Lactation/drug effects , Mammary Glands, Animal/chemistry , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Somatomedins/analysis , Somatomedins/biosynthesis
13.
J Physiol Pharmacol ; 60 Suppl 3: 77-81, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19996486

ABSTRACT

The present study aimed at researching the synergistic effect between an ectopic bone substitute and surrounding muscle tissue. To describe this effect, changes of insulin like growth factors (IGF1, IGF2), myostatin (GDF8) and vascular endothelial growth factor (VEGF) mRNA content of 12 Wistar-King rats musculus latissimus dorsi with implanted poly-3-hydroxybutyrate (PHB) scaffold were examined after 6 and 12 weeks. At each time interval six rats were killed and implants and surrounding tissues prepared for genetic evaluation. Eight rats without any implants served as controls. RNA was extracted from homogenized muscle tissue and reverse transcribed. Changes in mRNA content were measured by Real-Time PCR using specific primers for IGF1, IGF2, GDF8 and VEGF. Comparing the level of VEGF mRNA in muscle after 6 and 12 weeks to the controls, we could assess a significant increase of VEGF gene expression (p<0.05) whereas the level of mRNA expression was higher after 6 than after 12 weeks of treatment. Expression of IGF1 gene was also significantly increased as compared to the controls over the observed period of time (p<0.05). In the case of the IGF2 gene, the expression was significantly elevated after 6 weeks (p<0.05), but not significantly increased after 12 weeks (p>0.05). We observed a significantly decreased GDF8 gene expression (p<0.05) both after retrieval of implants after 6 as well as after 12 weeks. Moreover, mRNA level of GDF8 after 6 and 12 weeks were comparable the same. Our results show that PHB implants in rat musculus latissimus dorsi interact with the surrounding muscle tissue. This interaction works itself on growth potential of the muscle.


Subject(s)
Adaptation, Physiological/drug effects , Bone Substitutes/pharmacology , Hydroxybutyrates/pharmacology , Muscle, Skeletal/metabolism , Polyesters/pharmacology , Wound Healing/drug effects , Adaptation, Physiological/physiology , Animals , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Myostatin/drug effects , Myostatin/genetics , Myostatin/metabolism , Ossification, Heterotopic/metabolism , Osteogenesis/physiology , Prohibitins , RNA, Messenger/analysis , Random Allocation , Rats , Tissue Engineering , Tissue Scaffolds , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/physiology
14.
Cancer Detect Prev ; 32(4): 286-91, 2009.
Article in English | MEDLINE | ID: mdl-19185431

ABSTRACT

BACKGROUND: The mechanism of emodin-mediated cell apoptosis has been investigated extensively in many types of human cancer cells. Our previous study demonstrated that emodin induced apoptosis through the decrease of Bcl-2/Bax ratio and the increase of cytoplasm cytochrome c concentration in human breast cancer BCap-37 cells. However, emodin's reaction to breast cancer cells remains elusive. MATERIALS AND METHODS: An apoptosis-associated cDNA microarray comprised of 458 known genes, namely, death receptors, calpains, death kinases, granzymes, DNA fragmentation proteins, caspases and Bcl-2 family, was used to determine the impact of emodin in breast cancer BCap-37 cells. Furthermore, the candidate emodin target genes were further evaluated via real-time quantitative PCR and Western blot analysis. RESULTS: We found that gene expression profiling in human breast cancer BCap-37 cells was altered when exposed to emodin. Thirty of the unique genes that were either induced or repressed in response to emodin-induced apoptosis were also identified. A follow-up study characterized p53, emodin-induced gene, IGF-2, and emodin-repressed gene, and the downstream proteins were also seen as possible molecular targets of emodin. CONCLUSION: Data from this study provide novel evidence that emodin induces gene expression profiling changes, but has no effects on caspases. In addition, the p53 pathway may cooperate with the IGF-2 pathway, resulting in an emodin-induced apoptosis through disruption of the mitochondrial signaling pathway in BCap-37 cells.


Subject(s)
Breast Neoplasms/genetics , Emodin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Caspase 3/drug effects , Caspase 3/genetics , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Profiling , Genes, p53/drug effects , Humans , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Oligonucleotide Array Sequence Analysis , Receptors, Death Domain/drug effects , Signal Transduction/drug effects
15.
Clin Adv Hematol Oncol ; 6(1): 44-54, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18322441

ABSTRACT

BACKGROUND: Animal models suggest that growth hormone participates in hepatocarcinogenesis. OBJECTIVE: To correlate the effect of octreotide long-acting release (LAR) on insulin-like growth factor-I (IGF-I) and -II (IGF-II) with response and survival in patients with unresectable and metastatic hepatocellular carcinoma. METHODS: We conducted a phase II, single-institution trial of octreotide-LAR (30 mg intramuscularly every 4 weeks) in 15 patients while monitoring serum IGF-I and -II levels. RESULTS: Patients (median CLIP score 2, Okuda stage II, and ECOG performance status 1) were treated for a median of 2.0 cycles. No responses occurred. Median overall survival was 116 days (range, 27-937 days) and median progression-free survival was 60 days (range, 27-444 days). One patient had prolonged stable disease (16 months). There were no grade 4 and four grade 3 toxicities: abdominal cramping, elevated creatinine, diarrhea, and dyspnea. Median serum IGF-I decreased from baseline (42.2 ng/mL; range, 14.2-109 ng/mL) to day 29 (27.9 ng/mL; range, 5.7-71.1 ng/mL), and median serum IGF-II decreased from baseline (25,000 ng/mL; range, 12,400-93,600 ng/mL) to day 29 (18,400 ng/mL; range, 4,061-79,400 ng/mL; 2-sided P<.006 and P<.04, respectively; Wilcoxon signed rank test). This suppression did not correlate with clinical activity. Baseline serum IGF-I >30 ng/mL was associated with greater progression-free survival and overall survival (P=.0005 and P=.0173, respectively; 2-sided log-rank test). CONCLUSIONS: Octreotide-LAR lowered serum IGF-I and -II levels; however, this lowering did not correlate with clinical activity. There were no responses, and progression-free survival and overall survival were similar to historical patients not on treatment. Baseline serum IGF-I predicted prognosis.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Carcinoma, Hepatocellular/drug therapy , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor I/drug effects , Liver Neoplasms/drug therapy , Octreotide/pharmacology , Aged , Delayed-Action Preparations , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis/drug therapy , Prognosis
16.
Mol Reprod Dev ; 75(4): 588-97, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17886265

ABSTRACT

Amino acids play a multitude of roles during early embryonic development and have been demonstrated to facilitate improved development of in vivo or in vitro fertilized and parthenogenetic embryos in several species. However, review of emerging literatures, shows that culture milieu of cloned embryos might be different from those of in vitro fertilized embryos. This study therefore, evaluated the effect of nonessential amino acids (NEAA) on yield and quality of porcine embryos produced by somatic cell nuclear transfer (SCNT) and compared them with parthenogenetic embryos as control. Analysis showed that, supplementation of NEAA to culture medium significantly improved the blastocyst rate of parthenogenetic (38.9 +/- 8.8 vs. 27.5 +/- 9.0%) as well as SCNT (22.5 +/- 2.2 vs. 13.8 +/- 3.4%) embryos although cleavage rates were not different (P < 0.05). These blastocysts also had higher hatching ability and contained higher cell number than control blastocysts (P < 0.05). TUNEL labeling revealed that blastocysts cultured in the presence of NEAA were less predisposed to biochemical apoptosis and showed lower indices of TUNEL, fragmentation, and total apoptosis than those cultured in the absence of NEAA (P < 0.05). Real-time qRT-PCR analysis further revealed that NEAA decreased the expression ratio of BAX:BCL-xL and enhanced the relative abundance of IGF2 transcripts. Therefore, our study suggests that NEAA improves the yield and quality of cloned porcine embryos by enhancing blastocyst expansion, hatching, and total cell number and decreasing the apoptosis by positively modulating the expression of embryo survival related genes, similar to those reported for in vivo or in vitro fertilized embryos. Nonessential amino acids improve the yield and quality of cloned and parthenogenetic porcine embryos and modulate the expression of embryo survival related genes.


Subject(s)
Amino Acids/pharmacology , Embryonic Development/drug effects , Embryonic Stem Cells/drug effects , Nuclear Transfer Techniques , Oocytes/metabolism , Parthenogenesis/drug effects , Animals , Embryo Culture Techniques/methods , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Oocytes/cytology , Oocytes/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Swine , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/genetics , bcl-X Protein/drug effects , bcl-X Protein/genetics
17.
Placenta ; 28(11-12): 1147-57, 2007.
Article in English | MEDLINE | ID: mdl-17658597

ABSTRACT

The human first trimester placenta experiences a low oxygen environment. The hypoxia inducible factors (HIFs) mediate the response to low oxygen, inducing genes such as insulin-like growth factor (IGF)-II. Interestingly, IGF-II has been shown to promote placental growth and function. Currently, the interaction between oxygen, IGF-II and HIFs in the regulation of trophoblast behaviour are unclear. Murine implantation sites from days 5.5-10.5 were collected for immunohistochemical analyses. Use of the hypoxia marker pimonidazole indicated that the early mouse implantation site is exposed to low oxygen levels similar to those seen in the early human placenta. HIF-1alpha protein immunostaining was also observed in the implantation site. Culturing murine ectoplacental cones in decreasing oxygen concentrations (20%, 5% and 1% O(2)), either with or without the addition of IGF-II, induced complex responses by trophoblasts in terms of their migration and differentiation. Following 3 days exposure to low oxygen there was reduced EPC outgrowth, reduced Igf2 and increased Tpbp mRNA levels, suggesting commitment to the spongiotrophoblast lineage. In addition, Hif-1alpha mRNA levels were decreased, whilst Hif-2alpha mRNA was unchanged. This decrease in Hif-1alpha may be due to the observed increase in antisense (as) Hif-1alpha mRNA levels in 1% cultures. Furthermore, expression of Hif-2alpha and the HIF target genes: asHif-1alpha, Vegf and Slc2a1 were reduced under low oxygen with the addition of IGF-II. In conclusion, Hif-1alpha and Hif-2alpha are differentially regulated by oxygen and IGF-II in cultured trophoblast cells and asHif-1alpha may mediate the response to prolonged hypoxia in murine trophoblasts.


Subject(s)
Cell Differentiation/physiology , Decidua/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin-Like Growth Factor II/metabolism , Oxygen/physiology , Trophoblasts/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Decidua/cytology , Decidua/growth & development , Female , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Male , Mice , Mice, Inbred C57BL , Oxygen/pharmacology , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Receptor, IGF Type 2/genetics , Trophoblasts/cytology
18.
J Photochem Photobiol B ; 86(3): 240-5, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17182253

ABSTRACT

In this work, mycosporine-like amino acids (MAAs) of Synechocystis sp. PCC 6803 were characterized and were investigated on UV induction and protective ability. High performance liquid chromatographic (HPLC) studies revealed three major compounds in the MAAs. By UV absorption and mass spectra analysis, one of the compounds was tentatively identified as mycosporine-tau (M-tau). One novel compound similar to usujirene was tentatively named as dehydroxylusujirene, and the other novel compound was named as M-343 according to its absorption maximum. In vivo experiments indicated that M-tau was induced by both UV-A and UV-B, while dehydroxylusujirene and M-343 were only induced by UV-A, suggesting that different chromophores were involved in MAAs synthesis in Synechocystis sp. PCC 6803. It was also indicated that M-343 could be photochemically synthesized from some precursors. Under both UV and oxidation stresses, M-343 was more stable than dehydroxylusujirene and M-tau. Considering the reaction with H2O2, M-tau and dehydroxylusujirene might be potential antioxidants in reaction with physiological reactive oxygen species in vivo. In protection experiments, the MAAs exhibited efficient protective ability towards UV-B and H2O2 stresses, with maximal protection rates of 30% and 21.5%, respectively. These results indicate that the MAAs in Synechocystis sp. PCC 6803 act as both UV-screen and antioxidant.


Subject(s)
Amino Acids/biosynthesis , Radiation-Protective Agents/metabolism , Synechocystis/metabolism , Synechocystis/radiation effects , Ultraviolet Rays , Amino Acids/isolation & purification , Amino Acids/pharmacology , Chromatography, High Pressure Liquid , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/radiation effects , Oxidative Stress/drug effects , Peptide Fragments/drug effects , Peptide Fragments/radiation effects , Protein Precursors/drug effects , Protein Precursors/radiation effects , Radiation-Protective Agents/pharmacology , Sunscreening Agents/metabolism , Sunscreening Agents/pharmacology
19.
J Infect ; 52(6): 389-98, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16249033

ABSTRACT

OBJECTIVES: Low-dose growth hormone (GH) administration has been suggested as a treatment for HIV-lipodystrophy. METHODS: Postglucose GH-secretion, kinetics of insulin-like growth factors (IGFs), insulin, and glucose metabolism were examined in six male HIV-infected lipodystrophic patients (two normal-weight patients with normal glucose-tolerance (NGT), two normal-weight with impaired glucose-tolerance (IGT), and two obese patients with diabetes (DM)) during a 16 weeks open-labelled pilot-study of low-dose GH, 0.7 mg/day. RESULTS: DM, compared to NGT and IGT, displayed an impaired rebound of GH during a 5h oral glucose-tolerance test. Near lower normal limits in all patients before GH-therapy, total and free IGF-I increased between 87 and 152% during the GH-therapy (P<0.001), approaching upper normal limits in all patients with the highest incremental percentages shown in DM. A slight and temporary reduction in insulin sensitivity was caused by a reduction in non-oxidative glucose metabolism (n=5). GH-administration reduced hepatic extraction of insulin alleviating the demand for insulin secretion (n=5). No adverse effects of GH were detected. CONCLUSIONS: As judged from effects on circulating IGF-I, glucose metabolism, and insulin kinetics, 0.7 mg/day of GH may be expedient for treatment of HIV-infected males with lipodystrophy. Whether the patients' glucose metabolic status matters for the IGF-response to low-dose GH-therapy awaits further investigation.


Subject(s)
Glucose/metabolism , Growth Hormone/administration & dosage , HIV-Associated Lipodystrophy Syndrome/drug therapy , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor I/drug effects , Insulin/pharmacokinetics , Antiretroviral Therapy, Highly Active , C-Peptide/blood , Fatty Acids, Nonesterified/blood , Glucose Tolerance Test , Growth Hormone/metabolism , HIV Infections/complications , HIV-Associated Lipodystrophy Syndrome/etiology , Humans , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor II/analysis , Male , Middle Aged , Oxidation-Reduction , Time Factors
20.
J Neurooncol ; 67(1-2): 19-28, 2004.
Article in English | MEDLINE | ID: mdl-15072444

ABSTRACT

Tamoxifen, a non-steroidal anti-estrogen widely used against breast cancer, is also useful for treatment of other malignancies, due to its sensitizing effect on other chemotherapeutic agents and radiation. We have investigated the advantages of combining tamoxifen with one of the commonly used cancer chemotherapeutic drug, etoposide (VP-16) in brain tumor cell lines. While tamoxifen (10 microM) increased etoposide cytotoxicity 8.3-fold in the human glioma cell line (HTB-14), it increased etoposide cytotoxicity 47.5- and 40-fold in two primary cell lines established from pediatric medulloblastoma patients (MCH-BT-31 and MCH-BT-39), respectively. Similarly, in the pediatric ependymoma cell lines (MCH-BT-30 and MCH-BT-52), tamoxifen enhanced etoposide cytotoxicity 6- and 2.68-fold, respectively. CalcuSyn analysis of cytotoxicity data showed that tamoxifen and etoposide combinations were synergistic with combination index values ranging from 0.243 to 0.369 at IC50 level among different pediatric brain tumor cell lines. Tamoxifen is also cytotoxic at higher concentrations (> 20 microM) in brain tumor cells. To understand the mechanism underlying the tamoxifen modulation of etoposide cytotoxicity, we analyzed expression of P-glycoprotein (P-gp), insulin-like growth factor-I receptor (IGF-IR), IGF-I, IGF-II and estrogen receptor as well as protein kinase C (PKC) activity. While P-gp, IGF-IR and IGF-I were not affected, enhanced inhibition of PKC, and IGF-II were observed in brain tumor cells treated with tamoxifen and etoposide combination as compared to cells treated with either drug alone. Tamoxifen at 10 microM when combined with etoposide at 0-100 microM concentrations reduced PKC activity 77% compared to only 58% without tamoxifen. IGF-II expression decreased to 48.6% of the untreated control in the combination treatment as compared to 31.2% for etoposide alone and 26.2% for tamoxifen alone treatments. These results suggest that inhibitory effect of tamoxifen on brain tumor cells manifest through different mechanisms involving inhibition of targets such as PKC and IGF-II.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Etoposide/administration & dosage , Protein Kinase C/drug effects , Tamoxifen/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Child , Drug Synergism , Humans , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/drug effects , Protein Kinase C/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL