Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Mol Cell Endocrinol ; 518: 111035, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32941924

ABSTRACT

The insulin-like growth factor (IGF) system comprises two ligands, IGF-I and IGF-II, that regulate multiple physiological processes, including mammalian development, metabolism and growth, through the type 1 IGF receptor (IGF-1R). The growth hormone (GH)-IGF-I axis is the major regulator of longitudinal growth. IGF-II is expressed in many tissues, notably the placenta, to regulate human pre- and post-natal growth and development. This review provides a brief introduction to the IGF system and summarizes findings from reports arising from recent larger genomic sequencing studies of human genetic mutations in IGF1 and IGF2 and genes of proteins regulating IGF action, namely the IGF-1R, IGF-1R signaling pathway components and the IGF binding proteins (IGFBPs). A perspective on the effect of homozygous mutations on structure and function of the IGFs and IGF-1R is also given and this is related to the effects on growth.


Subject(s)
Growth Disorders/genetics , Metabolic Diseases/genetics , Receptors, Somatomedin/genetics , Somatomedins/genetics , Animals , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Growth Disorders/metabolism , Humans , Metabolic Diseases/metabolism , Mutation , Pregnancy , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/physiology , Receptors, Somatomedin/physiology , Signal Transduction/physiology , Somatomedins/physiology
2.
Gynecol Oncol ; 152(1): 185-193, 2019 01.
Article in English | MEDLINE | ID: mdl-30429055

ABSTRACT

OBJECTIVE: Inactivation of tumor suppressor genes promotes initiation and progression of cervical cancer. This study aims to investigate the tumor suppressive effects of TROP-2 in cervical cancer cells and to explain the underlying mechanisms. METHODS: The tumor suppressive functions of TROP-2 in cervical cancer cells were examined by in vitro and in vivo tumorigenic functional assays. Downstream factors of TROP-2 were screened using Human Phospho-Receptor Tyrosine Kinase Array. Small molecule inhibitors were applied to HeLa cells to test the TROP-2 effects on the oncogenicity of IGF-1R and ALK. Protein interactions between TROP-2 and the ligands of IGF-1R and ALK were detected via immunoprecipitation assay and protein-protein affinity prediction. RESULTS: In vitro and in vivo functional assays showed that overexpression of TROP-2 significantly inhibited the oncogenicity of cervical cancer cells; while knockdown of TROP-2 exhibited opposite effects. Human Phospho-Receptor Tyrosine Kinase Array showed that the activity of IGF-1R and ALK was stimulated by TROP-2 knockdown. Small molecule inhibitors AG1024 targeting IGF-1R and Crizotinib targeting ALK were treated to HeLa cells with and without TROP-2 overexpression, and results from cell viability and migration assays indicated that the oncogenicity of vector-transfected cells was repressed to a greater extent by the inhibition of either IGF-1R or ALK than that of the TROP-2-overexpressed cells. Immunoprecipitation assay and protein-protein affinity prediction suggested protein interactions between TROP-2 and the ligands of IGF-1R and ALK. CONCLUSIONS: Collectively, our results support that TROP-2 exhibits tumor suppressor functions in cervical cancer through inhibiting the activity of IGF-1R and ALK.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Receptors, Somatomedin/antagonists & inhibitors , Tumor Suppressor Proteins/physiology , Uterine Cervical Neoplasms/prevention & control , Anaplastic Lymphoma Kinase/physiology , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Cell Proliferation , Female , HeLa Cells , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Midkine/metabolism , Receptor, IGF Type 1 , Receptors, Somatomedin/physiology , Signal Transduction/physiology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology
3.
Chemosphere ; 217: 100-110, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30414542

ABSTRACT

Epidemiological studies have found that women have detectable levels of organic pollutants such as hexachlorobenzene (HCB), 2,2-dichlorodiphenyldichloroethylene (p,p'-DDE), polychlorinated biphenyl 153 (PCB153), perfluorooctanoate (PFOA), and perfluorooctane sulfonate (PFOS) in their follicular fluid. Thus, these compounds may directly affect the function of granulosa cells within the ovary and may promote granulosa cell tumor (GCT) progression. Two human GCT cell lines, COV434 and KGN, have been used as in vitro model systems to represent juvenile (JGCT) and adult (AGCT) GCT subtypes, respectively. In this study, we found that basal expression of estrogen receptor 1 (ESR1), estrogen receptor 2 (ESR2), and insulin-like growth factor 1 receptor (IGF1R) was higher in the AGCT subtype than in the JGCT subtype. All of the compounds acted as mitogenic factors at low nanomolar concentrations in the JGCT and AGCT forms of GCT. Interestingly, PFOA, PFOS, and HCB stimulated cell proliferation through IGF1R, whereas p,p'-DDE acted through GPR30. Moreover, a mixture of the five compounds also significantly stimulated granulosa cell proliferation; however, the observed effect was lower than predicted. Interestingly, the proliferative effect of a mixture of these compounds was dependent on IGF1R and GPR30 but independent of the classic estrogen receptors. Taken together, our results demonstrate for the first time that mixtures of persistent organic pollutants present in follicular fluids may induce granulosa tumor progression through IGF1R and GPR30 by acting as mitogenic factors in granulosa cells.


Subject(s)
Endocrine Disruptors/metabolism , Follicular Fluid/chemistry , Granulosa Cell Tumor/pathology , Receptors, Estrogen/analysis , Receptors, G-Protein-Coupled/physiology , Receptors, Somatomedin/physiology , Adolescent , Adult , Cell Line, Tumor , Cell Proliferation/drug effects , Endocrine Disruptors/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Granulosa Cell Tumor/etiology , Granulosa Cells/drug effects , Granulosa Cells/pathology , Humans , Mitogens , Ovarian Neoplasms , Receptor, IGF Type 1 , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Young Adult
4.
J Physiol ; 596(23): 5947-5964, 2018 12.
Article in English | MEDLINE | ID: mdl-29953638

ABSTRACT

KEY POINTS: The placental insulin-like growth factor (IGF) system is critical for normal fetoplacental growth, which is dysregulated following several pregnancy perturbations including uteroplacental insufficiency and maternal obesity. We report that the IGF system was altered in placentae of mothers born growth restricted compared to normal birth weight mothers, with maternal diet- and fetal sex-specific responses. Additionally, we report increased body weight and plasma IGF1 concentrations in fetuses from chow-fed normal birth weight mothers that exercised prior to and continued during pregnancy compared to sedentary mothers. Exercise initiated during pregnancy, on the other hand, resulted in placental morphological alterations and increased IGF1 and IGF1R protein expression, which may in part be modulated by reduced Let 7f-1 miRNA abundance. Growth restriction of mothers before birth and exercise differentially regulate the placental IGF system with diet- and sex-specific responses, probably as a means to improve fetoplacental growth and development, and hence neonatal survival. This increased neonatal survival may prevent adult disease onset. ABSTRACT: The insulin-like growth factor (IGF) system regulates fetoplacental growth and plays a role in disease programming. Dysregulation of the IGF system is implicated in several pregnancy perturbations associated with altered fetal growth, including intrauterine growth restriction and maternal obesity. Limited human studies have demonstrated that maternal exercise enhances fetoplacental growth and decreases cord IGF ligands, which may restore the placental IGF system in complicated pregnancies. This study investigated the impact maternal exercise has on the placental IGF system in placentae from mothers born growth restricted and if these outcomes are dependent on maternal diet or fetal sex. Uteroplacental insufficiency (Restricted) or sham (Control) surgery was induced on embryonic day (E) 18 in Wistar-Kyoto rats. F1 offspring were fed a chow or high-fat diet from weaning, and at 16 weeks were randomly allocated an exercise protocol: Sedentary, Exercised prior to and during pregnancy (Exercise), or Exercised during pregnancy only (PregEx). Females were mated (20 weeks) with placentae associated with F2 fetuses collected at E20. The placental IGF system mRNA abundance and placental morphology was altered in mothers born growth restricted. Exercise increased fetal weight and Control plasma IGF1 concentrations, and decreased female placental weight. PregEx did not influence fetoplacental growth but increased placental IGF1 and IGF1R (potentially modulated by reduced Let 7f-1 miRNA) and decreased placental IGF2 protein. Importantly, these placental IGF system changes occurred with sex-specific responses. These data highlight that exercise differently influences fetoplacental growth and the placental IGF system depending on maternal exercise initiation, which may prevent the transgenerational transmission of deficits and dysfunction.


Subject(s)
Diet, High-Fat , Fetal Growth Retardation/metabolism , Fetus/physiology , Physical Conditioning, Animal/physiology , Placenta/metabolism , Somatomedins/physiology , Animals , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/physiopathology , Male , Mothers , Pregnancy , Rats, Inbred WKY , Receptors, Somatomedin/physiology , Sex Factors , Up-Regulation
5.
Oncogene ; 37(14): 1869-1884, 2018 04.
Article in English | MEDLINE | ID: mdl-29353882

ABSTRACT

Antiestrogen resistance in estrogen receptor positive (ER+) breast cancer is associated with increased expression and activity of insulin-like growth factor 1 receptor (IGF1R). Here, a kinome siRNA screen has identified 10 regulators of IGF1R-mediated antiestrogen with clinical significance. These include the tamoxifen resistance suppressors BMPR1B, CDK10, CDK5, EIF2AK1, and MAP2K5, and the tamoxifen resistance inducers CHEK1, PAK2, RPS6KC1, TTK, and TXK. The p21-activated kinase 2, PAK2, is the strongest resistance inducer. Silencing of the tamoxifen resistance inducing genes, particularly PAK2, attenuates IGF1R-mediated resistance to tamoxifen and fulvestrant. High expression of PAK2 in ER+ metastatic breast cancer patients is correlated with unfavorable outcome after first-line tamoxifen monotherapy. Phospho-proteomics has defined PAK2 and the PAK-interacting exchange factors PIXα/ß as downstream targets of IGF1R signaling, which are independent from PI3K/ATK and MAPK/ERK pathways. PAK2 and PIXα/ß modulate IGF1R signaling-driven cell scattering. Targeting PIXα/ß entirely mimics the effect of PAK2 silencing on antiestrogen re-sensitization. These data indicate PAK2/PIX as an effector pathway in IGF1R-mediated antiestrogen resistance.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/therapeutic use , Receptors, Somatomedin/physiology , Rho Guanine Nucleotide Exchange Factors/metabolism , p21-Activated Kinases/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , High-Throughput Screening Assays , Humans , MCF-7 Cells , RNA, Small Interfering/pharmacology , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Tamoxifen/therapeutic use , p21-Activated Kinases/genetics
6.
Biochem J ; 474(24): 4105-4118, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29054976

ABSTRACT

Insulin-like growth factor-1 receptor (IGF1R) is a receptor tyrosine kinase that mediates growth, proliferation and survival. Dysregulation of IGF pathway contributes to the initiation, progression and metastasis of cancer and is also involved in diseases of glucose metabolism, such as diabetes. We have identified Ubiquilin1 (UBQLN1) as a novel interaction partner of IGF1R, IGF2R and insulin receptor (INSR). UBQLN family of proteins have been studied primarily in the context of protein quality control and in the field of neurodegenerative disorders. Our laboratory discovered a link between UBQLN1 function and tumorigenesis, such that UBQLN1 is lost and underexpressed in 50% of human lung adenocarcinoma cases. We demonstrate here that UBQLN1 regulates the expression and activity of IGF1R. Following loss of UBQLN1 in lung adenocarcinoma cells, there is accelerated loss of IGF1R. Despite decreased levels of total receptors, the ratio of active : total receptors is higher in cells that lack UBQLN1. UBQLN1 also regulates INSR and IGF2R post-stimulation with ligand. We conclude that UBQLN1 is essential for normal regulation of IGF receptors. UBQLN-1-deficient cells demonstrate increased cell viability compared with control when serum-starved and stimulation of IGF pathway in these cells increased their migratory potential by 3-fold. As the IGF pathway is involved in processes of normal growth, development, metabolism and cancer progression, understanding its regulation by Ubiquilin1 can be of tremendous value to many disciplines.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Receptors, Somatomedin/physiology , A549 Cells , Adaptor Proteins, Signal Transducing , Autophagy-Related Proteins , Cell Survival/physiology , HEK293 Cells , HeLa Cells , Humans , Protein Transport/physiology , Receptor, IGF Type 1
7.
PLoS One ; 12(9): e0185164, 2017.
Article in English | MEDLINE | ID: mdl-28945762

ABSTRACT

The insulin-like growth factor-1 receptor (IGF1R) mediates the biological actions of IGF1 and IGF2. The IGF1R is involved in both physiological and pathological activities and is usually overexpressed in most types of cancer. In addition to its classical mechanism of action, recent evidence has shown a nuclear presence of IGF1R, associated with novel genomic/transcriptional types of activities. The present study was aimed at evaluating the hypothesis that nuclear IGF1R localization is not restricted to cancer cells and might constitute a novel physiologically relevant regulatory mechanism. Our data shows that nuclear translocation takes place in a wide array of cells, including normal diploid fibroblasts. In addition, we provide evidence for a synergistic effect of a nuclear translocation blocker along with selective IGF1R inhibitors in terms of decreasing cell proliferation. Given the important role of the IGF1R in mitogenesis, the present results may be of translational relevance in cancer research. In conclusion, results are consistent with the concept that nuclear IGF1R fulfills important physiological and pathological roles.


Subject(s)
Cell Proliferation/physiology , Receptors, Somatomedin/physiology , Active Transport, Cell Nucleus/drug effects , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Nucleus/physiology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , MCF-7 Cells , Microscopy, Confocal , Receptor, IGF Type 1 , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/genetics , Signal Transduction
8.
J Exp Biol ; 220(Pt 15): 2777-2786, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28515235

ABSTRACT

Annual killifishes exhibit a number of unique life history characters including the occurrence of embryonic diapause, unique cell movements associated with dispersion and subsequent reaggregation of the embryonic blastomeres, and a short post-embryonic life span. Insulin-like growth factor (IGF) signaling is known to play a role in the regulation of metabolic dormancy in a number of animals but has not been explored in annual killifishes. The abundance of IGF proteins during development and the developmental effects of blocking IGF signaling by pharmacological inhibition of the insulin-like growth factor I receptor (IGF1R) were explored in embryos of the annual killifish Austrofundulus limnaeus Blocking of IGF signaling in embryos that would normally escape entrance into diapause resulted in a phenotype that was remarkably similar to that of embryos entering diapause. IGF-I protein abundance spikes during early development in embryos that will not enter diapause. In contrast, IGF-I levels remain low during early development in embryos that will enter diapause II. IGF-II protein is packaged at higher levels in escape-bound embryos compared with diapause-bound embryos. However, IGF-II levels quickly decrease and remain low during early development and only increase substantially during late development in both developmental trajectories. Developmental patterns of IGF-I and IGF-II protein abundance under conditions that would either induce or bypass entrance into diapause are consistent with a role for IGF signaling in the regulation of developmental trajectory and entrance into diapause in this species. We propose that IGF signaling may be a unifying regulatory pathway that explains the larger suite of characters that are associated with the complex life history of annual killifishes.


Subject(s)
Cyprinodontiformes/physiology , Diapause/physiology , Embryonic Development/physiology , Fish Proteins/physiology , Signal Transduction , Animals , Cyprinodontiformes/growth & development , Embryo, Nonmammalian/metabolism , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/physiology , Receptors, Somatomedin/physiology
9.
Oncotarget ; 7(34): 54445-54462, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27437872

ABSTRACT

Amplification of seven oncogenes: HER2, EGFR, FGFR1, FGFR2, MET, KRAS and IGF1R has been identified in gastric cancer. The first five are targeted therapeutically in patients with HER2-positivity, FGFR2- or MET-amplification but the majority of patients are triple-negative and require alternative strategies. Our aim was to evaluate the importance of the IGF1R tyrosine kinase in triple-negative gastric cancer with and without oncogenic KRAS, BRAF or PI3K3CA mutations. Cell lines and metastatic tumor cells isolated from patients expressed IGF1R, and insulin-like growth factor-1 (IGF-1) activated the PI3-kinase/Akt and Ras/Raf/MAP-kinase pathways. IGF-1 protected triple-negative cells from caspase-dependent apoptosis and anoikis. Protection was mediated via the PI3-kinase/Akt pathway. Remarkably, IGF-1-dependent cell survival was greater in patient samples. IGF-1 stimulated triple-negative gastric cancer cell growth was prevented by IGF1R knockdown and Ras/Raf/MAP-kinase pathway inhibition. The importance of the receptor in cell line and metastatic tumor cell growth in serum-containing medium was demonstrated by knockdown and pharmacological inhibition with figitumumab. The proportions of cells in S-phase and mitotic-phase, and Ras/Raf/MAP-kinase pathway activity, were reduced concomitantly. KRAS-addicted and BRAF-impaired gastric cancer cells were particularly susceptible. In conclusion, IGF1R and the IGF signal transduction pathway merit consideration as potential therapeutic targets in patients with triple-negative gastric cancer.


Subject(s)
Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/analysis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptors, Somatomedin/physiology , Stomach Neoplasms/pathology , ras Proteins/physiology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Amplification , Humans , Mutation , Phosphorylation , Proto-Oncogene Proteins B-raf/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, IGF Type 1 , Signal Transduction/physiology , Stomach Neoplasms/chemistry
10.
Tumour Biol ; 37(6): 7767-76, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26695144

ABSTRACT

A growing body of evidence suggests that microRNA-494 (miR-494) could act as tumor-suppressive or oncogenic microRNAs (miRNAs) in different types of tumors. However, the biological roles and underlying mechanisms of miR-494 remain unknown in human epithelial ovarian carcinoma (EOC). Therefore, the aims of this study were to investigate the miR-494 expression and the significance of its clinical diagnosis in patients suffering EOC and to analyze its role and underlying molecular mechanism on the carcinogenesis of EOC. Here, we found that miR-494 was significantly decreased in EOC cell lines and tissues and its expression was negatively correlated with advanced International Federation of Gynecology and Obstetrics (FIGO) stage, high pathological grade, and lymph node metastasis (all P < 0.01). Functional studies showed that overexpression of miR-494 in EOC cells could remarkably inhibit proliferation, colony formation, migration, and invasion and induce cell apoptosis, G0/G1 phase arrest. An in vivo analysis revealed that the overexpression of miR-494 suppressed tumor growth in a nude mouse xenograft model system. Bioinformatic assay and dual-luciferase assay confirmed that insulin-like growth factor 1 receptor (IGF1R) was as a direct target of miR-494 in EOC cells. Western blot assay showed that overexpression of miR-494 inhibited IGF1R expression and its downstream signal protein expression. In addition, downregulation of IGF1R has similar effects with miR-494 overexpression on EOC cells and overexpression of IGF1R effectively rescued the inhibition of overexpressed miR-494 in EOC cells. These data suggested that miR-494 functions as a tumor suppressor in EOC by targeting IGF1R.


Subject(s)
Carcinoma/pathology , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Neoplasm Proteins/physiology , Ovarian Neoplasms/pathology , RNA, Neoplasm/genetics , Receptors, Somatomedin/physiology , 3' Untranslated Regions/genetics , Animals , Apoptosis , Carcinoma/genetics , Cell Division , Cell Line , Cell Movement , Epithelial Cells/metabolism , Female , G1 Phase Cell Cycle Checkpoints , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovary/cytology , Receptor, IGF Type 1 , Receptors, Somatomedin/biosynthesis , Receptors, Somatomedin/genetics , Signal Transduction/genetics , Tumor Stem Cell Assay
11.
Am J Physiol Endocrinol Metab ; 309(6): E503-10, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26219867

ABSTRACT

The somatotropic signaling pathway has been implicated in aging and longevity studies in mice and other species. The physiology and lifespans of a variety of mutant mice, both spontaneous and genetically engineered, have contributed to our current understanding of the role of growth hormone and insulin-like growth factor I on aging-related processes. Several other mice discovered to live longer than their wild-type control counterparts also exhibit differences in growth factor levels; however, the complex nature of the phenotypic changes in these animals may also impact lifespan. The somatotropic axis impacts several pathways that dictate insulin sensitivity, nutrient sensing, mitochondrial function, and stress resistance as well as others that are thought to be involved in lifespan regulation.


Subject(s)
Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Longevity/physiology , Aging/physiology , Animals , Fibroblast Growth Factors/physiology , Mice , Mice, Knockout , Models, Animal , Pituitary Gland, Anterior/metabolism , Pituitary Hormones, Anterior/physiology , Receptors, Somatomedin/physiology , Signal Transduction
12.
Drug Metab Pers Ther ; 30(2): 75-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25941918

ABSTRACT

Among various auto/paracrine growth-regulating signaling pathways an important role belongs to that of insulin-like growth factors (IGFs) and insulin. IGF-signaling system is actively involved in the regulation of both normal ovarian function and ovarian tumor growth. On the one hand, all members of this system are expressed in malignant ovarian epithelial cells, and the prognostic significance of this expression has been revealed for some of them in ovarian cancer patients in several studies. On the other hand, circulating IGFs/IGFBPs levels have not been undoubtedly associated with ovarian cancer risk or disease progression, but some of them can be regarded as supplementary serological ovarian cancer markers. An important route to the clinical application of IGF-signaling system studies in ovarian cancer is the growing possibility of using specific molecular targeted agents to suppress its growth-stimulating and other activities. However, the introduction of such agents to practical oncology has met serious problems, with the main difficulties resulting from the absence of reliable predictive molecular markers and metabolic side effects due to the tight connection between IGF-signaling and insulin-regulated processes. The prognostic and diagnostic values of various IGF system components and the current state of corresponding molecular targeted therapies development for ovarian cancer are reviewed.


Subject(s)
Ovarian Neoplasms/diagnosis , Receptors, Somatomedin/physiology , Somatomedins/physiology , Female , Humans , Molecular Targeted Therapy/methods , Ovarian Neoplasms/drug therapy , Prognosis , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Signal Transduction/physiology , Somatomedins/antagonists & inhibitors , Somatomedins/metabolism
13.
Oncol Rep ; 33(4): 1899-907, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25647264

ABSTRACT

Inhibition of histone deacetylases (HDACs) has emerged as a new target for cancer therapies. The present study examined the antitumor effect and molecular mechanism of the HDAC inhibitor apicidin in YD-15 human salivary mucoepidermoid carcinoma (MEC) cells. The cells were treated with apicidin and cell death was quantified using an MTT assay. Apoptosis and autophagy were measured using flow cytometry, immunoblot analysis and cell staining. Regulation of the signaling pathways was monitored using immunoblot analysis and co-treatment with specific inhibitors. Insulin-like growth factor 1 receptor (IGF-1R) was knocked down using specific siRNA. Apicidin significantly inhibited the proliferation of MEC cells. Apicidin also induced apoptosis through the inactivation of extracellular signal-regulated kinase (ERK) and AKT/mTOR signaling and activation of c-Jun NH2-terminal kinase (JNK), whereas apicidin promoted autophagy through inactivation of the AKT/mTOR signaling. These effects may be mediated by the inhibition of IGF-1R, an upstream regulator of MAPK and AKT/mTOR pathways. These results suggested that apicidin is an attractive chemotherapeutic agent against salivary MEC and may be a good candidate for targeting IGF-1R for cancer therapies.


Subject(s)
Carcinoma, Mucoepidermoid/pathology , Histone Deacetylase Inhibitors/pharmacology , Neoplasm Proteins/biosynthesis , Peptides, Cyclic/pharmacology , Receptors, Somatomedin/biosynthesis , Salivary Gland Neoplasms/pathology , Signal Transduction/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Mucoepidermoid/genetics , Carcinoma, Mucoepidermoid/metabolism , Cell Division/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , RNA Interference , RNA, Small Interfering/genetics , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Receptors, Somatomedin/physiology , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology
14.
Eur Neuropsychopharmacol ; 24(12): 1947-53, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24529663

ABSTRACT

The involvement of insulin, the insulin-like growth factors (IGF1, IGF2) and their receptors in central nervous system development and function has been the focus of scientific interest for more than 30 years. The insulin-like peptides, both locally-produced proteins as well as those transported from the circulation into the brain via the blood-brain barrier, are involved in a myriad of biological activities. These actions include, among others, neuronal survival, neurogenes, angiogenesis, excitatory and inhibitory neurotransmission, regulation of food intake, and cognition. In recent years, a linkage between brain insulin/IGF1 and certain neuropathologies has been identified. Epidemiological studies have demonstrated a correlation between diabetes (mainly type 2) and Alzheimer׳s disease. In addition, an aberrant decline in IGF1 values was suggested to play a role in the development of Alzheimer׳s disease. The present review focuses on the expression and function of insulin, IGFs and their receptors in the brain in physiological and pathological conditions.


Subject(s)
Brain/pathology , Brain/physiology , Insulin/physiology , Receptor, Insulin/physiology , Receptors, Somatomedin/physiology , Animals , Appetite/physiology , Brain/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Disease Models, Animal , Glucose/metabolism , Humans , Insulin/metabolism , Life Expectancy , Lipid Metabolism/physiology , Receptor, Insulin/metabolism , Receptors, Somatomedin/metabolism , Somatomedins/metabolism , Somatomedins/physiology
15.
Circulation ; 127(21): 2097-106, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23625957

ABSTRACT

BACKGROUND: Several microRNAs (miRs) have been shown to regulate gene expression in the heart, and dysregulation of their expression has been linked to cardiac disease. miR-378 is strongly expressed in the mammalian heart but so far has been studied predominantly in cancer, in which it regulates cell survival and tumor growth. METHODS AND RESULTS: Here, we report tight control of cardiomyocyte hypertrophy through miR-378. In isolated primary cardiomyocytes, miR-378 was found to be both necessary and sufficient to repress cardiomyocyte hypertrophy. Bioinformatic prediction suggested that factors of the mitogen-activated protein kinase (MAPK) pathway are enriched among miR-378 targets. Using mRNA and protein expression analysis along with luciferase assays, we validated 4 key components of the MAPK pathway as targets of miR-378: MAPK1 itself, insulin-like growth factor receptor 1, growth factor receptor-bound protein 2, and kinase suppressor of ras 1. RNA interference with these targets prevented the prohypertrophic effect of antimiR-378, suggesting their functional relation with miR-378. Because miR-378 significantly decreases in cardiac disease, we sought to compensate for its loss through adeno-associated virus-mediated, cardiomyocyte-targeted expression of miR-378 in an in vivo model of cardiac hypertrophy (pressure overload by thoracic aortic constriction). Restoration of miR-378 levels significantly attenuated thoracic aortic constriction-induced cardiac hypertrophy and improved cardiac function. CONCLUSIONS: Our data identify miR-378 as a regulator of cardiomyocyte hypertrophy, which exerts its activity by suppressing the MAPK signaling pathway on several distinct levels. Restoration of disease-associated loss of miR-378 through cardiomyocyte-targeted adeno-associated virus-miR-378 may prove to be an effective therapeutic strategy in myocardial disease.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , MicroRNAs/physiology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/physiology , Signal Transduction/physiology , Adenoviridae/genetics , Animals , Cells, Cultured , Disease Models, Animal , Down-Regulation/physiology , GRB2 Adaptor Protein/antagonists & inhibitors , GRB2 Adaptor Protein/physiology , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Kinases/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/physiology
16.
Minerva Endocrinol ; 37(2): 173-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22691890

ABSTRACT

The major issue currently being faced in the management of prostate cancer is the inability to distinguish between indolent prostate tumors that will not present clinically from more aggressive and metastatic prostate cancers that will impact on men's lives. Only a small proportion of prostate cancers can be accounted for by unmistakable hereditary cancer syndromes and the predominant contribution to the progression of most sporadic cancers is thought to be environmental, with nutrition having the greatest influence. Population studies have clearly implicated metabolic factors as contributors to disease progression and poor response to therapy. It is well established that the IGF system is key in regulating growth and metabolism and mediates the effects of nutrition on these processes. It consists of two ligands (IGF-I and IGF-II), two receptors [type 1 IGF-IR and IGF-II/mannose 6-phosphate receptor], and six high affinity IGF-binding proteins (IGFBP-1 to -6). This review provides evidence from in vitro, in vivo, clinical and epidemiology studies that indicates an important role for the IGF axis in the development of prostate cancer and the likely role that it plays in mediating the effects of nutrition on disease progression. We suggest that the IGF axis is central to understanding how lifestyle impacts on prostate cancer and we highlight this by describing numerous strategies being developed to target this axis.


Subject(s)
Adenocarcinoma/metabolism , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Receptors, Somatomedin/physiology , Somatomedins/physiology , Adenocarcinoma/epidemiology , Adenocarcinoma/physiopathology , Adenocarcinoma/therapy , Androgens/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Disease Progression , Energy Metabolism , Humans , Incidence , Insulin-Like Growth Factor Binding Proteins , Male , Molecular Targeted Therapy , Neoplasms, Hormone-Dependent/epidemiology , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/physiopathology , Neoplasms, Hormone-Dependent/therapy , Overnutrition/complications , Overnutrition/metabolism , Overnutrition/physiopathology , Prevalence , Prostatectomy , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/therapy , Tumor Cells, Cultured , Zinc/metabolism
17.
Endocrinol Metab Clin North Am ; 41(2): 249-63, v, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22682629

ABSTRACT

Although very similar to insulin and its receptor; the modus operandi of the insulin-like growth factors (IGFs) within the body is very different from that of the traditional peptide hormone. The IGF-binding proteins bind the IGFs with greater affinity than the cell surface receptors, enabling them to tightly control tissue activity. In addition to their role in fetal and childhood growth, IGFs play an important role in metabolic regulation. This article describes the basic underlying human physiology of IGFs, how this differs from that of experimental models, and why some information can only be learned from human clinical studies.


Subject(s)
Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor I/physiology , Aging/physiology , Animals , Cell Survival/physiology , Humans , Insulin-Like Growth Factor Binding Proteins/physiology , Mice , Receptors, Somatomedin/physiology , Sedentary Behavior
18.
Endocrinol Metab Clin North Am ; 41(2): 375-93, vii, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22682636

ABSTRACT

Insulin-like growth factors (IGFs) play an integral role in development, growth, and survival. This article details the current understanding of the effects of IGFs in the peripheral nervous system (PNS) during health and disease, and introduces how the IGF system regulates PNS development and impacts growth and survival of PNS cells. Also discussed are implications of IGF signaling in neurodegeneration and the status and prospects of IGF therapies for PNS conditions. There is substantial support for the application of IGF therapies in the treatment of PNS injury and disease.


Subject(s)
Peripheral Nervous System/physiology , Somatomedins/physiology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Survival , Humans , Insulin-Like Growth Factor Binding Proteins/physiology , Male , Mice , Peripheral Nervous System/growth & development , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/physiopathology , Rats , Receptors, Somatomedin/physiology , Somatomedins/therapeutic use , Translational Research, Biomedical
19.
Gen Comp Endocrinol ; 167(3): 344-51, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20403355

ABSTRACT

The insulin-like growth factor (IGF) signaling pathway consists of multiple IGF ligands, IGF receptors, and IGF-binding proteins (IGFBPs). Studies in a variety of animal and cellular systems suggest that the IGF signaling pathway plays a key role in regulating skeletal muscle growth, differentiation, and in maintaining homeostasis of the adult muscle tissues. Intriguingly, IGFs stimulate both myoblast proliferation and differentiation, which are two mutually exclusive biological events during myogenesis. Both of these actions are mediated through the same IGF-1 receptor. Recent studies have shed new insights into the molecular mechanisms underlying these paradoxical actions of IGFs in muscle cells. In this article, we provide a brief review of our current understanding of the IGF signaling system and discuss recent findings on how local oxygen availability and IGFBPs act to specify IGF actions in muscle cells.


Subject(s)
Cell Differentiation , Insulin-Like Growth Factor Binding Proteins/physiology , Muscle, Skeletal/growth & development , Receptors, Somatomedin/physiology , Somatomedins/physiology , Animals , Cell Differentiation/genetics , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Models, Biological , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Somatomedins/genetics , Somatomedins/metabolism
20.
Reprod Fertil Dev ; 22(1): 75-87, 2010.
Article in English | MEDLINE | ID: mdl-20003848

ABSTRACT

Development of the post-hatching conceptus in ruminants involves a period of morphological expansion that is driven by complex interactions between the conceptus and its intrauterine environment. As a result of these interactions, endometrial physiology is altered, leading to establishment of the pregnancy and continued development of the placenta. Disruption of normal fetal and placental development can occur when embryos are exposed to manipulations in vitro or when inappropriate endocrine sequencing occurs in vivo during the pre- and peri-implantation periods. The present review addresses the development of the post-hatching bovine conceptus, its interactions with the maternal system and changes in development that can occur as a result of in vivo and in vitro manipulations of the bovine embryo.


Subject(s)
Cattle/physiology , Congenital Abnormalities/veterinary , Pregnancy, Animal/physiology , Animals , Congenital Abnormalities/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Female , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/physiology , Pregnancy , Pregnancy, Animal/genetics , RNA, Antisense/genetics , RNA, Antisense/physiology , Receptors, Somatomedin/genetics , Receptors, Somatomedin/physiology , Syndrome , Uterus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...