Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Nutr Biochem ; 120: 109417, 2023 10.
Article in English | MEDLINE | ID: mdl-37482256

ABSTRACT

The micronutrient selenium (Se) has been shown to exert potential anticancer properties. This study aimed to evaluate the effects of Se (in Se yeast form) on the selenoproteins (SELENO), AR/IGF-1R/EGFR, PI3K/Akt/mTOR and Ras/Raf/ERK cascades, and immune checkpoint blockade in TNBC murine 4T1 cells. We also assessed the effects of combination treatment with chemotherapeutic doxorubicin and Se on trophoblast cell surface antigen 2 (TROP2) levels. Compared with the control groups, cells incubated with Se (0.25, 0.5, 0.75, 1.0, 1.5 µg Se/mL) have lower viability, raised intracellular Se concentrations and SELENO expression, and higher malondialdehyde products in a dose-dependent manner. Se induced the inactivation of AR/IGF-1R/EGFR and downregulation of the PI3K/Akt/mTOR and Ras/Raf/ERK signaling molecules. Se-treated cells also exhibited decreased mitochondrial membrane potential, reduced levels of the cell cycle regulatory protein cyclin D1, cancer stemness, metastatic and EMT-related markers, and increased apoptosis. Subsequently, Se treatment significantly suppressed PD-1/PD-L1 and CTLA-4 mRNA levels and proteins. Doxorubicin decreased 4T1 cell viability and TROP2 expression levels, but the addition of Se to doxorubicin contributed to further reductions. Similar responses to Se treatment were also observed in the human MDA-MB-231 and MCF-7 breast cancer cells. These results show that Se upregulates SELENO and anti-AR/IGF-1R/EGFR signaling in TNBC cells, thus inducing oxidative stress-dependent apoptosis and cell cycle arrest, stemness, EMT, and metastasis, as well as blocking the immune checkpoint molecules. TROP2 down-regulation with Se is also a potential anti-TNBC therapeutic target.


Subject(s)
Breast Neoplasms , Carcinoma , Selenium , Animals , Mice , Humans , Female , Selenium/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Signal Transduction , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , TOR Serine-Threonine Kinases/metabolism , Breast Neoplasms/drug therapy , Apoptosis , ErbB Receptors/metabolism , Doxorubicin/pharmacology , Cell Proliferation
2.
Biochem Cell Biol ; 101(5): 432-442, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37018819

ABSTRACT

Atrial structural remodeling takes on a critical significance to the occurrence and maintenance of atrial fibrillation (AF). As revealed by recent data, insulin-like growth factor-1 receptor (IGF-1R) plays a certain role in tissue fibrosis. In this study, the mechanism of IGF-1R in atrial structural remodeling was examined based on in vivo and in vitro experiments. First, cluster analysis of AF hub genes was conducted, and then the molecular mechanism was proposed by which IGF-1R regulates myocardial fibrosis via the PI3K/Akt/FoxO3a pathway. Subsequently, the mentioned mechanism was verified in human cardiac fibroblasts (HCFs) and rats transduced with IGF-1 overexpression type 9 adeno-associated viruses. The results indicated that IGF-1R activation up-regulated collagen Ⅰ protein expression and Akt phosphorylation in HCFs and rat atrium. The administration of LY294002 reversed the above phenomenon, improved the shortening of atrial effective refractory period, and reduced the increased incidence of AF and atrial fibrosis in rats. The transfection of FoxO3a siRNA reduced the anti-fibrotic effect of LY294002 in HCFs. The above data revealed that activation of IGF-1R takes on a vital significance to atrial structural remodeling by facilitating myocardial fibrosis and expediting the occurrence and maintenance of AF through the regulation of the PI3K/Akt/FoxO3a signaling pathway.


Subject(s)
Atrial Fibrillation , Animals , Humans , Rats , Atrial Fibrillation/genetics , Fibrosis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , Signal Transduction
3.
Ann Med ; 55(1): 954-964, 2023 12.
Article in English | MEDLINE | ID: mdl-36896461

ABSTRACT

BACKGROUND/AIMS: Colorectal cancer is the third leading cause of death in patients with cancers in America. Monensin has represented anti-cancer effect on various human cancer cells. We seek to investigate the effect of monensin on proliferation of human colorectal cancer cells and explore whether IGF1R signaling pathway is involved in anti-cancer mechanism of monensin. METHODS: Cell proliferation and migration were assessed by crystal violet staining and cell wounding assay respectively. Cell apoptosis was analyzed by Hoechst 33258 staining and flow cytometry. Cell cycle progression was detected with the use of flow cytometry. Cancer-associated pathways were assessed with the use of pathway-specific reporters. Gene expression was detected by touchdown-quantitative real-time PCR. Inhibition of IGF1R was tested by immunofluorescence staining. Inhibition of IGF1R signaling was accomplished by adenovirus-mediated expression of IGF1. RESULTS: We found that monensin not only effectively inhibited cell proliferation, cell migration as well as cell cycle progression, but also induced apoptosis and G1 arrest in human colorectal cancer cells. Monensin was shown to target multiple cancer-related signaling pathways such as Elk1, AP1, as well as Myc/max, and suppressed IGF1R expression via increasing IGF1 in colorectal cancer cells. CONCLUSION: Monensin could suppressed IGF1R expression via increasing IGF1 in colorectal cancer cells. It has the potential to be repurposed as an anti-colorectal cancer agent, but further studies are still required to investigate the detailed mechanisms of monensin underlying its anti-cancer motion.Key MessagesMonensin inhibits the cell proliferation and the migration, induces apoptosis and inhibits cell cycle progression in human colorectal cancer cells.Monensin may exert anti-cancer activity by targeting multiple signaling pathways, including the IGF1R signaling pathway.Monensin has the potential to be repurposed as an anti-colorectal cancer agent.


Subject(s)
Monensin , Neoplasms , Humans , Anti-Bacterial Agents , Apoptosis , Cell Line, Tumor , Cell Proliferation , Monensin/pharmacology , Receptor, IGF Type 1/pharmacology , Signal Transduction , Colorectal Neoplasms/metabolism
4.
J Pathol ; 259(2): 205-219, 2023 02.
Article in English | MEDLINE | ID: mdl-36373776

ABSTRACT

Colorectal cancer (CRC) is one of the most common cancers worldwide. The tumor microenvironment exerts crucial effects in driving CRC progression. Cancer-associated fibroblasts (CAFs) serve as one of the most important tumor microenvironment components promoting CRC progression. This study aimed to elucidate the novel molecular mechanisms of CAF-secreted insulin-like growth factor (IGF) 2 in colorectal carcinogenesis. Our results indicated that IGF2 was a prominent factor upregulated in CAFs compared with normal fibroblasts. CAF-derived conditioned media (CM) promoted tumor growth, migration, and invasion of HCT 116 and DLD-1 cells. IGF1R expression is significantly increased in CRC, serving as a potent receptor in response to IGF2 stimulation and predicting unfavorable outcomes for CRC patients. Apart from the PI3K-AKT pathway, RNA-seq analysis revealed that the YAP1-target signature serves as a prominent downstream effector to mediate the oncogenic signaling of IGF2-IGF1R. By single-cell RNA sequencing (scRNA-seq) and immunohistochemical validation, IGF2 was found to be predominantly secreted by CAFs, whereas IGF1R was expressed mainly by cancer cells. IGF2 triggers the nuclear accumulation of YAP1 and upregulates YAP1 target signatures; however, these effects were abolished by either IGF1R knockdown or inhibition with picropodophyllin (PPP), an IGF1R inhibitor. Using CRC organoid and in vivo studies, we found that cotargeting IGF1R and YAP1 with PPP and verteporfin (VP), a YAP1 inhibitor, enhanced antitumor effects compared with PPP treatment alone. In conclusion, this study revealed a novel molecular mechanism by which CAFs promote CRC progression. The findings highlight the translational potential of the IGF2-IGF1R-YAP1 axis as a prognostic biomarker and therapeutic target for CRC. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Humans , Cancer-Associated Fibroblasts/pathology , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Signal Transduction , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Cell Proliferation , Tumor Microenvironment , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor II/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology
5.
Tissue Cell ; 79: 101914, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36183441

ABSTRACT

PURPOSE: ß-elemene has a wide range of anticancer effects and can be used in a variety of cancer types. This study mainly explored its mechanism of action on TNBC cells and provided theoretical basis for the treatment of TNBC. METHODS: Firstly, TNBC cells were treated with different concentrations of ß-elemene, and screened out an appropriate concentration for subsequent research. Then, through the bioinformatics website, predicted genes that have a binding relationship with ß-elemene. Then, the overexpression vector of the selected gene was transfected into the cell. The effects of ß-elemene and its target genes on the proliferation and apoptosis of TNBC cells were detected by CCK-8, Edu assay, and flow cytometry, and the senescence of cells was determined by SA-ß-gal experiment. Western blotting was used to detect the expression of apoptosis and aging-related proteins. RESULTS: ß-elemene inhibited TNBC cell viability and proliferation in a concentration-dependent manner, and induces apoptosis and senescence. Through the screening of candidate genes, IGF1 was finally determined to be an effective target gene of ß-elemene. The expression level of IGF1 was decreased in cells treated with ß-elemene. Overexpression of IGF1 significantly alleviated ability of ß-elemene to inhibit cell viability, proliferation, and induced cell apoptosis and senescence. In addition, ß-elemene inhibited the expression of IGF1R and Bcl-2, and promoted the expression of Cleaved Caspase-3 and senescence-related proteins (p27, p16, p53 and p21), and these effects were reversed by overexpression of IGF1. CONCLUSION: ß-elemene induced apoptosis and senescence of triple-negative breast cancer cells through IGF1/IGF1R pathway.


Subject(s)
Sesquiterpenes , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Sesquiterpenes/pharmacology , Apoptosis , Cell Proliferation , Insulin-Like Growth Factor I , Receptor, IGF Type 1/pharmacology
6.
Autophagy ; 17(12): 4029-4042, 2021 12.
Article in English | MEDLINE | ID: mdl-33749521

ABSTRACT

ALS (amyotrophic lateral sclerosis), the most common motor neuron disease, causes muscle denervation and rapidly fatal paralysis. While motor neurons are the most affected cells in ALS, studies on the pathophysiology of the disease have highlighted the importance of non-cell autonomous mechanisms, which implicate astrocytes and other glial cells. In ALS, subsets of reactive astrocytes lose their physiological functions and become toxic for motor neurons, thereby contributing to disease pathogenesis. Evidence of astrocyte contribution to disease pathogenesis are well established in cellular and animal models of familial ALS linked to mutant SOD1, where astrocytes promote motor neuron cell death. The mechanism underlying astrocytes reactivity in conditions of CNS injury have been shown to involve the MTOR pathway. However, the role of this conserved metabolic signaling pathway, and the potential therapeutic effects of its modulation, have not been investigated in ALS astrocytes. Here, we show elevated activation of the MTOR pathway in human-derived astrocytes harboring mutant SOD1, which results in inhibition of macroautophagy/autophagy, increased cell proliferation, and enhanced astrocyte reactivity. We demonstrate that MTOR pathway activation in mutant SOD1 astrocytes is due to post-transcriptional upregulation of the IGF1R (insulin like growth factor 1 receptor), an upstream positive modulator of the MTOR pathway. Importantly, inhibition of the IGF1R-MTOR pathway decreases cell proliferation and reactivity of mutant SOD1 astrocytes, and attenuates their toxicity to motor neurons. These results suggest that modulation of astrocytic IGF1R-MTOR pathway could be a viable therapeutic strategy in SOD1 ALS and potentially other neurological diseases.Abbreviations: ACM: astrocyte conditioned medium; AKT: AKT serine/threonine kinase; ALS: amyotrophic lateral sclerosis; BrdU: thymidine analog 5-bromo-2'-deoxyuridine; CNS: central nervous system; EIF4EBP1/4EBP1: eukaryotic translation initiation factor 4E binding protein 1; GFAP: glial fibrillary acidic protein; IGF1R: insulin like growth factor 1 receptor; INSR: insulin receptor; iPSA: iPSC-derived astrocytes; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta;MTOR: mechanistic target of rapamycin kinase; NES: nestin; PPK1: 3-phosphoinositide dependent protein kinase 1; PI: propidium iodide; PPP: picropodophyllotoxin; PTEN: phosphatase and tensin homolog; S100B/S100ß: S100 calcium binding protein B; SLC1A3/ EAAT1: solute carrier family 1 member 3; SMI-32: antibody to nonphosphorylated NEFH; SOD1: superoxide dismutase 1; TUBB3: tubulin beta 3 class III; ULK1: unc-51 like autophagy activating kinase 1.


Subject(s)
Amyotrophic Lateral Sclerosis , Astrocytes , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/metabolism , Autophagy , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , TOR Serine-Threonine Kinases/metabolism
7.
Brain Res Bull ; 143: 27-35, 2018 10.
Article in English | MEDLINE | ID: mdl-30278200

ABSTRACT

Depression is associated with uncontrolled diabetes, which indicates a lack of insulin effect, yet the role of the insulin receptor in mediating depression is not clearly established because insulin receptors are not required for glucose entry into the brain. However, insulin receptors are important for brain function since insulin receptor knockout mice have depressive-like activity. Depression and cognitive problems are also associated with low insulin-like growth factor-1 (IGF-1) in the elderly. Rodent studies showed chronic IGF-1 administration had antidepressant-like (AD) activity. We asked if insulin in the brain might act through the IGF-1 receptor, as it does in some tissues. We used acute insulin or IGF-1 infusions into the 3rd ventricle (icv) in rats and tested animals in a forced swim test. We found that antidepressive-like behavior is mediated by insulin and IGF-1. Further, administration of the IGF-1 receptor antagonist JB-1 blocked the antidepressive-like activity of the insulin and IGF-1, indicating a strong relationship between insulin, IGF-1 and depression. Insulin acts at least partially through the IGF-1 receptor and is responsive to receptor antagonism. The model offers promise for future studies of the mechanism of depression, and therapy to increase insulin sensitivity and IGF-1 action including exercise and nutrition.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Receptor, IGF Type 1/drug effects , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Depression/etiology , Depressive Disorder/etiology , Fluoxetine/metabolism , Fluoxetine/pharmacology , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Motor Activity/drug effects , Oligopeptides/metabolism , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Swimming/psychology
8.
Biochem Biophys Res Commun ; 498(4): 877-883, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29545181

ABSTRACT

Insulin-like growth factor-1 (IGF-1) levels are known to increase in the bronchoalveolar lavage fluid (BALF) of patients with acute respiratory distress syndrome. Herein, we investigated the role of IGF-1 in lipopolysaccharide (LPS)-induced lung injury. In LPS-treated cells, expressions of receptor-interacting protein 3 (RIP3) and phosphorylated mixed lineage kinase domain-like protein (MLKL) were decreased in IGF-1 receptor small interfering RNA (siRNA)-treated cells compared to control cells. The levels of pro-inflammatory cytokines including interleukin (IL)-1ß, IL-6, IL-10, tumour necrosis factor-α, and macrophage inflammatory protein 2/C-X-C motif chemokine ligand 2 in the supernatant were significantly reduced in IGF-1 receptor siRNA-treated cells compared to control cells. In LPS-induced murine lung injury model, total cell counts, polymorphonuclear leukocytes counts, and pro-inflammatory cytokine levels in the BALF were significantly lower and histologically detected lung injury was less common in the group treated with IGF-1 receptor monoclonal antibody compared to the non-treated group. On western blotting, RIP3 and phosphorylated MLKL expressions were relatively decreased in the IGF-1 receptor monoclonal antibody group compared to the non-treated group. IGF-1 may be associated with RIP3-mediated necroptosis in vitro, while blocking of the IGF-1 pathway may reduce LPS-induced lung injuries in vivo.


Subject(s)
Lung Injury/prevention & control , Necrosis/etiology , Receptor, IGF Type 1/pharmacology , Animals , Antigens, CD/metabolism , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cadherins/metabolism , Lipopolysaccharides , Lung/metabolism , Lung Injury/chemically induced , Macrophages/metabolism , Mice , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
9.
Gen Comp Endocrinol ; 245: 44-54, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28185934

ABSTRACT

Corticosteroids play positive or negative role in the reproductive mechanisms of many fish species but the physiological contexts relating to such biphasic actions are not well defined. In the present study we investigated to what extent corticosteroids (cortisol-Co, 11-deoxycorticosterone-DOC) hormones may interfere with the steroidogenic capacity of Eurasian perch ovarian tissues, and we tested whether the negative effects of corticosteroids may be mitigated by potential stimulating endocrine factors, namely insulin-like growth factor-1 (IGF), human chorionic gonadotropin (HCG) or thyroid hormones (Triidothyronine-T3, thyroxine-T4). Ovarian tissues from six maturing fish at late vitellogenesis developmental stage (LVO) or at the start of the final meiotic oocyte maturation (FMO) were incubated during 6h in Cortland medium containing various endocrine compounds. Both corticosteroids drastically suppressed aromatase activity (AA) and sex-steroid production, namely 17-ß estradiol (E2), 17α-20ß-dihydroxy-4-pregnen-3-one (DHP) and testosterone (T). HCG significantly prevented the suppression of both AA and sex-steroid production by low and high cortisol doses, but a lesser AA protection was observed in the case of DOC. The protection of DHP and T productions by HCG from the negative effects by the two corticosteroids was higher at FMO than at LVO stage. IGF or thyroid hormone treatments were lesser effective or ineffective in mitigating the suppression of AA or sex-steroid production by cortisol. The results suggest that an increase in cortisol or DOC such as after mild or high stress intensity may inhibit drastically the ovarian steroidogenic capacity whatever the final oocyte maturation stage in percid fish by hampering AA and sex-steroid production. That inhibition may be partly mitigated by gonadotropins but not IGF nor thyroid hormones, especially at final meiotic oocyte maturation stage.


Subject(s)
Desoxycorticosterone/pharmacology , Gonadal Steroid Hormones/metabolism , Hydrocortisone/pharmacology , Perches/physiology , Animals , Aromatase/genetics , Aromatase/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Gonadal Steroid Hormones/genetics , Humans , Oocytes/growth & development , Ovary/drug effects , Ovary/metabolism , Receptor, IGF Type 1/pharmacology , Thyroxine/pharmacology , Tissue Culture Techniques , Triiodothyronine/pharmacology
10.
Cancer Res ; 76(17): 4981-93, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27364558

ABSTRACT

Syndecan-1 (Sdc1/CD138) expression is linked to disease severity in multiple myeloma, although the causal basis for this link remains unclear. Here we report that capture of the IGF1 receptor (IGF1R) by Sdc1 suppresses ASK1-dependent apoptosis in multiple myeloma cells. Sdc1 binds two different fractions of IGF1R, one that is constitutively active and a second that is activated by IGF1 ligand. Notably, IGF1R kinase activity in both fractions is blocked by synstatinIGF1R (SSTNIGF1R), a peptide that inhibits IGF1R capture by Sdc1, as well as by a truncated peptide (SSTNIGF1R-T) that appears to be specific for multiple myeloma cells. Mechanistically, we show that ASK1 is bound to active IGF1R and inhibited by Tyr and Ser83/Ser966 phosphorylation. When IGF1R engagement with Sdc1 is blocked by SSTNIGF1R, ASK1 becomes activated, and initiates JNK- and caspase-3-mediated apoptosis. In pharmacologic tests, we find SSTNIGF1R is highly stable in human plasma and displays a half-life of 27 hours in mice, wherein it significantly reduces both the size and neovascularization of CAG myeloma tumor xenografts. Taken together, our results offer a preclinical proof of concept and mechanistic rationale for the exploration of SSTNIGF1R as an experimental therapeutic to dually attack multiple myeloma tumor cell survival and tumor angiogenesis. Cancer Res; 76(17); 4981-93. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/pathology , Receptor, IGF Type 1/pharmacology , Receptors, Somatomedin/metabolism , Syndecan-1/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Flow Cytometry , Gene Knockdown Techniques , Heterografts , Humans , Immunoprecipitation , MAP Kinase Kinase Kinase 5/metabolism , Mice , Mice, Nude , Multiple Myeloma/metabolism , Peptides/pharmacology
11.
Asian Pac J Cancer Prev ; 16(1): 221-5, 2015.
Article in English | MEDLINE | ID: mdl-25640355

ABSTRACT

As metformin can inhibit endometrial carcinoma (EC) cell growth and the insulin growth factor (IGF) system is active in EC, the question of whether t can regulate endometrial carcinoma cell secretion of IGF-1 or expression of IGF-1 receptor (IGF-1R) is of interest. In this study, serum IGF-1 levels in EC patients were found to be comparable with that in the non EC patients (p>0.05). However, the IGF-1 level in the medium of cultured cells after treatment with metformin was decreased (p<0.05). IGF-1R was highly expressed in human endometrial carcinoma paraffin sections, but IGF-1R and phosphor-protein kinase B/protein kinase B (p-Akt/ Akt) expression was down-regulated after metformin treatment (p<0.05). In summary, metformin can reduce the secretion of IGF-1 by Ishikawa and JEC EC cell lines and their expression of IGF-1R to deactivate downstream signaling involving the PI-3K/Akt pathway to inhibit endometrial carcinoma cell growth.


Subject(s)
Carcinoma/drug therapy , Down-Regulation/drug effects , Endometrial Neoplasms/drug therapy , Insulin-Like Growth Factor I/genetics , Metformin/pharmacology , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/pharmacology , Carcinoma/genetics , Cell Line, Tumor , Down-Regulation/genetics , Endometrial Neoplasms/genetics , Female , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Biochem Biophys Res Commun ; 447(2): 271-7, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24704450

ABSTRACT

The molecular mechanisms of multiple myeloma are not well defined. EEN is an endocytosis-regulating molecule. Here we report that EEN regulates the proliferation and survival of multiple myeloma cells, by regulating IGF-1 secretion. In the present study, we observed that EEN expression paralleled with cell proliferation, EEN accelerated cell proliferation, facilitated cell cycle transition from G1 to S phase by regulating cyclin-dependent kinases (CDKs) pathway, and delayed cell apoptosis via Bcl2/Bax-mitochondrial pathway. Mechanistically, we found that EEN was indispensable for insulin-like growth factor-1 (IGF-1) secretion and the activation of protein kinase B-mammalian target of rapamycin (Akt-mTOR) pathway. Exogenous IGF-1 overcame the phenotype of EEN depletion, while IGF-1 neutralization overcame that of EEN over-expression. Collectively, these data suggest that EEN may play a pivotal role in excessive cell proliferation and insufficient cell apoptosis of bone marrow plasma cells in multiple myeloma. Therefore, EEN may represent a potential diagnostic marker or therapeutic target for multiple myeloma.


Subject(s)
Biomarkers, Tumor/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/pathology , Receptor, IGF Type 1/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , G1 Phase Cell Cycle Checkpoints , Humans , Intracellular Signaling Peptides and Proteins/genetics , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/pharmacology , TOR Serine-Threonine Kinases/metabolism
13.
Mol Imaging Biol ; 16(4): 529-37, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24532107

ABSTRACT

PURPOSE: The aim was to assess changes in insulin-like growth factor 1 receptor (IGF-1R) expression with immunoSPECT/CT and to study the dynamics of IGF-1R expression of human breast tumors during endocrine treatment. PROCEDURES: Mice with MCF-7 xenografts were treated with estradiol or tamoxifen, and IGF-1R expression was measured by immunohistochemistry and immunoSPECT/CT using (111)In-R1507 (anti-IGF-1R antibody). Moreover, IGF-1R expression was analyzed immunohistochemically on 22 human breast tumors, treated preoperatively with endocrine therapy. RESULTS: Estradiol resulted in an increased expression of IGF-1R, as measured by immunohistochemistry and immunoSPECT/CT. In contrast, tamoxifen resulted in a downregulation of IGF-1R, whereas this could not be measured with immunoSPECT/CT. A downregulation was also detectable in 9 out of 22 (41 %) human breast tumors after endocrine therapy. CONCLUSIONS: Anti-estrogen treatment can cause a reduction in membranous IGF-1R expression. Based on these results, a combination of anti-IGF-1R antibodies with anti-estrogen therapy might not be a rational treatment strategy.


Subject(s)
Breast Neoplasms/drug therapy , Estradiol/therapeutic use , Receptor, IGF Type 1/metabolism , Tamoxifen/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Breast Neoplasms/pathology , Cell Membrane/drug effects , Cell Membrane/metabolism , Estradiol/pharmacology , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Middle Aged , Receptor, IGF Type 1/pharmacology , Tamoxifen/pharmacology , Xenograft Model Antitumor Assays
14.
Cancer Biol Ther ; 13(7): 559-66, 2012 May.
Article in English | MEDLINE | ID: mdl-22406993

ABSTRACT

We generated novel truncated insulin-like growth factor I receptors (IGF-IRs) designated as 126/STOP, 223/STOP and 325/STOP in order to establish shorter soluble IGF-IRs than previously reported 486/STOP without abrogating the same antitumor effects. Stable transfection of 223/STOP and 325/STOP, but not 126/STOP caused inhibition of anchorage-independent growth of CaOV-3 ovarian cancer cells in vitro. This antitumor effect was reproduced when we used recombinant proteins of these constructs, suggesting a bystander effect of these shorter truncated IGF-IRs. Tumorigenesis in vivo of CaOV-3 cells tranfected with 223/STOP or 325/STOP was strictly inhibited, and inoculation of these cells in nude mice caused massive apoptosis exclusively in vivo. Phosphorylations of IGF-IR and Akt, but not Erk were attenuated in 223/STOP- or 325/STOP-transfected CaOV-3 cells, and downregulations of IGF-IR and Akt phosphorylation seemed to play at least a partial role in the anti-tumor effect of these novel truncated IGF-IRs. Since 223/STOP and 325/STOP are smaller in size than previously reported 486/STOP, and they retain the same antitumor effects, they could be good candidates for clinical application in the future.


Subject(s)
Ovarian Neoplasms/genetics , Receptor, IGF Type 1/genetics , Animals , Apoptosis/genetics , Bystander Effect/drug effects , CHO Cells , Cell Culture Techniques , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cricetinae , Culture Media, Conditioned/chemistry , Female , Gene Expression , Humans , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transfection
15.
Mol Cancer ; 10: 100, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21854628

ABSTRACT

BACKGROUND: The transcription factor, CCAAT enhancer binding protein-ß (C/EBPß), is expressed as several distinct protein isoforms (LAP1, LAP2 and LIP) that have opposing actions in cellular proliferation and differentiation. Increases in the ratio of LIP/LAP are associated with aggressive, metastatic breast cancer; however, little is known regarding the molecular mechanisms that regulate LIP expression or the biological actions of an increase in the LIP/LAP ratio. Metastasis is highly dependent upon the suppression of anoikis and the role of C/EBPß and LIP in this anchorage-independent, survival process is currently not known in mammary epithelial cells. IGF-1R signaling is important for the survival of breast cancer cells and crosstalk between IGF-1R and EGFR signaling pathways have been implicated in the development of more aggressive disease. We therefore evaluated in mammary epithelial cells whether IGF-1R signaling regulates the LIP/LAP ratio, analyzed the potential interplay between EGFR and IGF-1R signaling and addressed the biological significance of increased LIP expression in cellular survival and suppression of anoikis. RESULTS: Our data provide the first evidence that IGF-1R signaling regulates LIP expression in an EGFR independent manner to increase the LIP/LAP ratio in mammary epithelial cells. Although crosstalk between IGF-1R signaling and EGFR signaling is detectable in MCF10A cells, this crosstalk is not required for the IGF-1 mediated regulation of LIP expression. Rather, the critical regulator of IGF-1 induced LIP expression appears to be EGFR-independent, Akt activity. Our data also demonstrate that increases in LIP expression promote cell survival via suppression of anoikis. Likewise, knockdown of total C/EBPß leads to increased cell death and suggest that C/EBPß expression is important for survival and resistance to anoikis. IGF-1 treatment can partially rescue vector control cells from anoikis; however, cells with reduced C/EBPß expression do not survive anoikis. CONCLUSIONS: Taken together, our data demonstrate that IGF-1R signaling regulates LIP expression in an EGFR independent manner to increase the LIP/LAP ratio in mammary epithelial cells. C/EBPß expression and elevations in LIP play an important role in regulating cellular survival via suppression of anoikis, in an IGF-1R mediated context or in a manner independent of IGF-1R signaling.


Subject(s)
Anoikis , CCAAT-Enhancer-Binding Protein-beta/metabolism , Receptor, IGF Type 1/physiology , Signal Transduction , Breast Neoplasms , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line, Tumor , Cell Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gene Expression , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/pharmacology
16.
Pain ; 152(4): 888-895, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21296499

ABSTRACT

Sensitization of primary afferent neurons is one of the most important components of pain hypersensitivity after tissue injury. Insulin-like growth factor 1 (IGF-1), involved in wound repair in injured tissue, also plays an important role in maintaining neuronal function. In the present study, we investigated the effect of tissue IGF-1 on nociceptive sensitivity of primary afferent neurons. Local administration of IGF-1 induced thermal and mechanical pain hypersensitivity in a dose-dependent manner, and was attenuated by IGF-1 receptor (IGF1R) inhibition. Tissue but not plasma IGF-1 levels, as determined by enzyme-linked immunosorbent assay, significantly increased after plantar incision. Immunohistochemistry revealed that IGF1R was predominantly expressed in neurons as well as in satellite glial cells in the dorsal root ganglion (DRG). Double-labeling immunohistochemistry showed that IGF1R expression colocalized with peripherin and TRPV1, but not with NF200 in DRG neurons. The IGF1R inhibitor successfully alleviated mechanical allodynia, heat hyperalgesia, and spontaneous pain behavior observed after plantar incision. Expression of phosphorylated Akt in DRG neurons significantly increased after plantar incision and was suppressed by IGF1R inhibition. These results demonstrate that increased tissue IGF-1 production sensitizes primary afferent neurons via the IGF1R/Akt pathway to facilitate pain hypersensitivity after tissue damage.


Subject(s)
Hyperalgesia/etiology , Hyperalgesia/metabolism , Insulin-Like Growth Factor I/adverse effects , Pain Threshold/physiology , Postoperative Complications/physiopathology , Animals , Cell Count/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Hyperalgesia/drug therapy , Insulin-Like Growth Factor I/metabolism , Male , Oncogene Protein v-akt/metabolism , Pain Measurement , Pain Threshold/drug effects , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/therapeutic use , Postoperative Complications/pathology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...