Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 178(21): 4389-4407, 2021 11.
Article in English | MEDLINE | ID: mdl-34233013

ABSTRACT

BACKGROUND AND PURPOSE: The multikinase inhibitor sorafenib is a first-line drug for advanced hepatocellular carcinoma. The response to sorafenib varies among hepatocellular carcinoma patients and many of the responders suffer from reduced sensitivity after long-term treatment. This study aims to explore a novel strategy to potentiate or maximize the anti-hepatocellular carcinoma effects of sorafenib. EXPERIMENTAL APPROACH: We used hepatocellular carcinoma cell lines, western blotting, various antagonists, siRNA and tumour xenografts mouse model to determine the anti- hepatocellular carcinoma effects of sorafenib in combination with berbamine or other Na+ /K+ -ATPase ligands. KEY RESULTS: Berbamine and the cardiotonic steroid, ouabain, synergize with sorafenib to inhibit hepatocellular carcinoma cells growth. Mechanistically, berbamine induces Src phosphorylation in Na+ /K+ -ATPase-dependent manner, leading to the activation of p38MAPK and EGFR-ERK pathways. The Na+ /K+ -ATPase ligand ouabain also induces Src, EGFR, type I insulin-like growth factor receptor, ERK1/2 and p38MAPK phosphorylation in hepatocellular carcinoma cells. Treatment of hepatocellular carcinoma cells with Src or EGFR inhibitor inhibits the induction of ERK1/2 phosphorylation by berbamine. Moreover, sorafenib inhibits the induction of Src, p38MAPK, EGFR and ERK1/2 phosphorylation by berbamine and ouabain. Importantly, combination of sorafenib with berbamine or ouabain synergistically inhibits both sorafenib-naïve and sorafenib-resistant hepatocellular carcinoma cells growth. Co-treatment of hepatocellular carcinoma cells with berbamine and sorafenib significantly induces cell death and significantly inhibits hepatocellular carcinoma xenografts growth in vivo. CONCLUSION AND IMPLICATIONS: Berbamine or other Na+ /K+ -ATPase ligands have a potential for improving sorafenib responsiveness in hepatocellular carcinoma. Targeting Na+ /K+ -ATPase represents a novel strategy to potentiate the anti- hepatocellular carcinoma effects of sorafenib.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Benzylisoquinolines , Carcinoma, Hepatocellular/drug therapy , Humans , Liver Neoplasms/drug therapy , Mice , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Sorafenib/pharmacology , src-Family Kinases/metabolism
2.
Exp Hematol Oncol ; 9(1): 32, 2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33292604

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is the first discovered second messenger, which plays pivotal roles in cell signaling, and regulates many physiological and pathological processes. cAMP can regulate the transcription of various target genes, mainly through protein kinase A (PKA) and its downstream effectors such as cAMP-responsive element binding protein (CREB). In addition, PKA can phosphorylate many kinases such as Raf, GSK3 and FAK. Aberrant cAMP-PKA signaling is involved in various types of human tumors. Especially, cAMP signaling may have both tumor-suppressive and tumor-promoting roles depending on the tumor types and context. cAMP-PKA signaling can regulate cancer cell growth, migration, invasion and metabolism. This review highlights the important roles of cAMP-PKA-CREB signaling in tumorigenesis. The potential strategies to target this pathway for cancer therapy are also discussed.

3.
J Hematol Oncol ; 13(1): 64, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32493414

ABSTRACT

Insulin-like growth factors (IGFs) play important roles in mammalian growth, development, aging, and diseases. Aberrant IGFs signaling may lead to malignant transformation and tumor progression, thus providing the rationale for targeting IGF axis in cancer. However, clinical trials of the type I IGF receptor (IGF-IR)-targeted agents have been largely disappointing. Accumulating evidence demonstrates that the IGF axis not only promotes tumorigenesis, but also confers resistance to standard treatments. Furthermore, there are diverse pathways leading to the resistance to IGF-IR-targeted therapy. Recent studies characterizing the complex IGFs signaling in cancer have raised hope to refine the strategies for targeting the IGF axis. This review highlights the biological activities of IGF-IR signaling in cancer and the contribution of IGF-IR to cytotoxic, endocrine, and molecular targeted therapies resistance. Moreover, we update the diverse mechanisms underlying resistance to IGF-IR-targeted agents and discuss the strategies for future development of the IGF axis-targeted agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm/physiology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor, IGF Type 1/physiology , Signal Transduction/physiology , Somatomedins/physiology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Cell Nucleus/metabolism , Cell Physiological Phenomena/drug effects , Cell Physiological Phenomena/physiology , Cell Self Renewal/physiology , Clinical Trials as Topic , Combined Modality Therapy , DNA Damage , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , Disease Progression , Drug Development , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Integrins/physiology , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/physiopathology , Neoplasms/radiotherapy , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Tumor Microenvironment
4.
J Hematol Oncol ; 12(1): 71, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31277692

ABSTRACT

Mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. mTOR is usually assembled into several complexes such as mTOR complex 1/2 (mTORC1/2). In cooperation with raptor, rictor, LST8, and mSin1, key components in mTORC1 or mTORC2, mTOR catalyzes the phosphorylation of multiple targets such as ribosomal protein S6 kinase ß-1 (S6K1), eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), Akt, protein kinase C (PKC), and type-I insulin-like growth factor receptor (IGF-IR), thereby regulating protein synthesis, nutrients metabolism, growth factor signaling, cell growth, and migration. Activation of mTOR promotes tumor growth and metastasis. Many mTOR inhibitors have been developed to treat cancer. While some of the mTOR inhibitors have been approved to treat human cancer, more mTOR inhibitors are being evaluated in clinical trials. Here, we update recent advances in exploring mTOR signaling and the development of mTOR inhibitors for cancer therapy. In addition, we discuss the mechanisms underlying the resistance to mTOR inhibitors in cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy/methods , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
5.
J Biol Chem ; 294(15): 5945-5955, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30782845

ABSTRACT

GADD34 (growth arrest and DNA damage-inducible gene 34) plays a critical role in responses to DNA damage and endoplasmic reticulum stress. GADD34 has opposing effects on different stimuli-induced cell apoptosis events, but the reason for this is unclear. Here, using immunoblotting analyses and various molecular genetic approaches in HepG2 and SMMC-7721 cells, we demonstrate that GADD34 protects hepatocellular carcinoma (HCC) cells from tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by stabilizing a BCL-2 family member, myeloid cell leukemia 1 (MCL-1). We found that GADD34 knockdown decreased MCL-1 levels and that GADD34 overexpression up-regulated MCL-1 expression in HCC cells. GADD34 did not affect MCL-1 transcription but enhanced MCL-1 protein stability. The proteasome inhibitor MG132 abrogated GADD34 depletion-induced MCL-1 down-regulation, suggesting that GADD34 inhibits the proteasomal degradation of MCL-1. Furthermore, GADD34 overexpression promoted extracellular signal-regulated kinase (ERK) phosphorylation through a signaling axis that consists of the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6) and transforming growth factor-ß-activated kinase 1 (MAP3K7)-binding protein 1 (TAB1), which mediated the up-regulation of MCL-1 by GADD34. Of note, TRAIL up-regulated both GADD34 and MCL-1 levels, and knockdown of GADD34 and TRAF6 suppressed the induction of MCL-1 by TRAIL. Correspondingly, GADD34 knockdown potentiated TRAIL-induced apoptosis, and MCL-1 overexpression rescued TRAIL-treated and GADD34-depleted HCC cells from cell death. Taken together, these findings suggest that GADD34 inhibits TRAIL-induced HCC cell apoptosis through TRAF6- and ERK-mediated stabilization of MCL-1.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Proteins/metabolism , Protein Phosphatase 1/metabolism , TNF Receptor-Associated Factor 6/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Proteins/genetics , Protein Phosphatase 1/genetics , Protein Stability , TNF Receptor-Associated Factor 6/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics
6.
Oncol Lett ; 17(1): 944-950, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30655852

ABSTRACT

Cisplatin (DDP)-based anticancer therapy is an important chemotherapeutic strategy for the treatment of colorectal cancer. However, its beneficial effect is largely compromised by adverse reactions, and more importantly, by the development of drug resistance. Therefore, it is crucial to determine the potential mechanism underlying the development of DDP resistance in colorectal cancer. Interleukin-17 (IL-17) is a proinflammatory cytokine that has been found to serve an important role in the host defense during cancer development. It has been suggested that IL-17 is key to promoting the development of resistance to DDP in several major types of cancer. However, the role of IL-17 in DDP resistance in colorectal cancer has not been extensively investigated. In the present study, it was observed that IL-17 was significantly upregulated in colorectal tumor samples, compared with the adjacent tissues. Furthermore, IL-17 was found to promote the viability of HCT116 colorectal cells treated with DDP, whilst blocking IL-17 signaling leading to HCT116 cell apoptosis. IL-17 was also shown to regulate the expression of several apoptosis-related proteins, including phosphorylated-protein kinase B (p-Akt), apoptosis regulator BAX (Bax), apoptosis regulator Bcl-2 (Bcl-2) and serine/threonine-protein kinase mTOR (mTOR). These findings indicated that IL-17 facilitates the development of DDP resistance in colorectal cancer by inhibiting cancer cell apoptosis through targeting p-Akt, Bax, Bcl-2 and mTOR. Overall, the findings of the present study suggest that a combination of DDP and an IL-17 inhibitor may prove to be a highly efficient strategy for colorectal cancer treatment.

7.
Med Res Rev ; 39(1): 114-145, 2019 01.
Article in English | MEDLINE | ID: mdl-29855050

ABSTRACT

The nonsteroidal anti-inflammatory agent aspirin is widely used for preventing and treating cardiovascular and cerebrovascular diseases. In addition, epidemiologic evidences reveal that aspirin may prevent a variety of human cancers, while data on the association between aspirin and some kinds of cancer are conflicting. Preclinical studies and clinical trials also reveal the therapeutic effect of aspirin on cancer. Although cyclooxygenase is a well-known target of aspirin, recent studies uncover other targets of aspirin and its metabolites, such as AMP-activated protein kinase, cyclin-dependent kinase, heparanase, and histone. Accumulating evidence demonstrate that aspirin may act in different cell types, such as epithelial cell, tumor cell, endothelial cell, platelet, and immune cell. Therefore, aspirin acts on diverse hallmarks of cancer, such as sustained tumor growth, metastasis, angiogenesis, inflammation, and immune evasion. In this review, we focus on recent progress in the use of aspirin for cancer chemoprevention and therapy, and integratively analyze the mechanisms underlying the anticancer effects of aspirin and its metabolites. We also discuss mechanisms of aspirin resistance and describe some derivatives of aspirin, which aim to overcome the adverse effects of aspirin.


Subject(s)
Aspirin/therapeutic use , Chemoprevention , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Aspirin/adverse effects , Aspirin/chemistry , Biomarkers, Tumor/metabolism , Clinical Trials as Topic , Humans , Neoplasms/epidemiology
8.
Exp Hematol Oncol ; 7: 24, 2018.
Article in English | MEDLINE | ID: mdl-30250760

ABSTRACT

Estrogen is a steroid hormone that has critical roles in reproductive development, bone homeostasis, cardiovascular remodeling and brain functions. However, estrogen also promotes mammary, ovarian and endometrial tumorigenesis. Estrogen antagonists and drugs that reduce estrogen biosynthesis have become highly successful therapeutic agents for breast cancer patients. The effects of estrogen are largely mediated by estrogen receptor (ER) α and ERß, which are members of the nuclear receptor superfamily of transcription factors. The mechanisms underlying the aberrant expression of ER in breast cancer and other types of human tumors are complex, involving considerable alternative splicing of ERα and ERß, transcription factors, epigenetic and post-transcriptional regulation of ER expression. Elucidation of mechanisms for ER expression may not only help understand cancer progression and evolution, but also shed light on overcoming endocrine therapy resistance. Herein, we review the complex mechanisms for regulating ER expression in human cancer.

9.
Cell Death Dis ; 9(6): 625, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29795373

ABSTRACT

Transforming growth factor ß (TGF-ß) is critical for embryonic development, adult tissue homeostasis, and tumor progression. TGF-ß suppresses tumors at early stage, but promotes metastasis at later stage through oncogenes such as Twist1. Gamma-synuclein (SNCG) is overexpressed in a variety of invasive and metastatic cancer. Here, we show that TGF-ß induces SNCG expression by Smad-Twist1 axis, thus promoting TGF-ß- and Twist1-induced cancer cell migration and invasion. We identify multiple Twist1-binding sites (E-boxes) in SNCG promoter. Chromatin immunoprecipitation and luciferase assays confirm the binding of Twist1 to the E-boxes of SNCG promoter sequence (-129/-1026 bp). Importantly, the Twist1-binding site close to the transcription initiation site is critical for the upregulation of SNCG expression by TGF-ß and Twist1. Mutations of Twist1 motif on the SNCG promoter constructs markedly reduces the promoter activity. We further show that TGF-ß induces Twist1 expression through Smad thereby enhancing the binding of Twist1 to SNCG promoter, upregulating SNCG promoter activity and increasing SNCG expression. SNCG knockdown abrogates TGF-ß- or Twist1-induced cancer cell migration and invasion. Finally, SNCG knockdown inhibits the promotion of cancer metastasis by Twist1. Together, our data demonstrate that SNCG is a novel target of TGF-ß-Smad-Twist1 axis and a mediator of Twist1-induced cancer metastasis.


Subject(s)
Cell Movement , Nuclear Proteins/metabolism , Transforming Growth Factor beta/pharmacology , Twist-Related Protein 1/metabolism , gamma-Synuclein/metabolism , E-Box Elements , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Hep G2 Cells , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Promoter Regions, Genetic , Smad Proteins/metabolism , Up-Regulation/drug effects , gamma-Synuclein/genetics
10.
J Cell Mol Med ; 22(1): 589-599, 2018 01.
Article in English | MEDLINE | ID: mdl-29024409

ABSTRACT

The natural agent rhein is an ananthraquinone derivative of rhubarb, which has anticancer effects. To determine the mechanisms underlying the anticancer effects of rhein, we detected the effect of rhein on several oncoproteins. Here, we show that rhein induces ß-catenin degradation in both hepatoma cell HepG2 and cervical cancer cell Hela. Treatment of HepG2 and Hela cells with rhein shortens the half-life of ß-catenin. The proteasome inhibitor MG132 blunts the downregulation of ß-catenin by rhein. The induction of ß-catenin degradation by rhein is dependent on GSK3 but independent of Akt. Treatment of HepG2 and Hela cells with GSK3 inhibitor or GSK3ß knockdown abrogates the effect of rhein on ß-catenin. GSK3ß knockdown compromises the inhibition of HepG2 and Hela cell growth by rhein. Furthermore, rhein dose not downregulate ß-catenin mutant that is deficient of phosphorylation at multiple residues including Ser33, Ser37, Thr41 and Ser45. Moreover, rhein induces cell cycle arrest at S phase in both HepG2 and Hela cells. Intraperitoneal administration of rhein suppresses tumour cells proliferation and tumour growth in HepG2 xenografts model. Finally, the levels of ß-catenin are reduced in rhein-treated tumours. These data demonstrate that rhein can induce ß-catenin degradation and inhibit tumour growth.


Subject(s)
Anthraquinones/pharmacology , Biological Products/pharmacology , Cell Cycle Checkpoints/drug effects , Neoplasms/pathology , Proteolysis/drug effects , beta Catenin/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Female , Glycogen Synthase Kinase 3 beta/metabolism , HeLa Cells , Hep G2 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , S Phase/drug effects , Xenograft Model Antitumor Assays
11.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 34(3): 415-420, 2017 Jun 01.
Article in Chinese | MEDLINE | ID: mdl-29745508

ABSTRACT

AMP-activated protein kinase (AMPK) is involved in the development and progression of tumors including hepatocellular carcinoma (HCC). However, studies on AMPK and tumorigenesis were largely based on experiments in vitro or tumor xenografts model. Here, we introduce a liver-specific AMPKα1 knockout mice model, which is achieved by Alb-Cre recombinase system. The expression of AMPKα1 in the liver of AMPKα1 -/--Alb-Cre mice is absent. AMPKα1 knockout in the liver does not affect the growth and histological structure of mouse liver. This model provides a favorable tool to the study of the roles of AMPKα1 in liver metabolism or tumorigenesis.

12.
Oncotarget ; 7(13): 16349-61, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26918349

ABSTRACT

AMP-activated protein kinase (AMPK) is an important energy sensor that may inhibit cell proliferation or promote cell survival during stresses. Besides cyclooxygenase, AMPK is another target of the nonsteroid anti-inflammatory agent aspirin. Preclinical and clinical investigations demonstrate that aspirin can inhibit several types of cancer such as colorectal adenomas and hepatocellular carcinoma (HCC). However, little is known about the cellular response to aspirin that may lead to aspirin resistance. Here, we show that aspirin induces the expression of MCL-1 in HepG2 and SW480 cells through AMPK-mTOR-Akt/ERK axis. Treatment of HepG2 and SW480 cells with aspirin leads to increased MCL-1 expression, Akt and ERK1/2 phosphorylation. Inhibition of Akt/MEK abrogates the induction of MCL-1 by aspirin. Aspirin activates AMPK, which in turn up-regulates mTORC2 activity, Akt, ERK1/2 phosphorylation and MCL-1 expression. MCL-1 knockdown sensitizes cancer cells to aspirin-induced apoptosis. Combination of aspirin and AMPK, Akt or MEK inhibitor results in more significant inhibition of cell proliferation and induction of apoptosis than single agent. Moreover, sorafenib blocks aspirin-induced MCL-1 up-regulation. Combination of aspirin and sorafenib leads to much more cell death and less cell proliferation than each drug alone. Treatment of HCC and colon cancer xenografts with both aspirin and sorafenib results in more significant tumor suppression than single agent. These data demonstrate that AMPK-mediated up-regulation of mTORC2 and MCL-1 may compromise the anticancer effects of aspirin. Combination of aspirin and sorafenib may be an effective regimen to treat HCC and colon cancer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anticarcinogenic Agents/pharmacology , Aspirin/pharmacology , Neoplasms, Experimental/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Sorafenib , TOR Serine-Threonine Kinases/biosynthesis , Up-Regulation , Xenograft Model Antitumor Assays
13.
Cell Res ; 26(1): 46-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26584640

ABSTRACT

Mammalian target of rapamycin (mTOR) is a core component of raptor-mTOR (mTORC1) and rictor-mTOR (mTORC2) complexes that control diverse cellular processes. Both mTORC1 and mTORC2 regulate several elements downstream of type I insulin-like growth factor receptor (IGF-IR) and insulin receptor (InsR). However, it is unknown whether and how mTOR regulates IGF-IR and InsR themselves. Here we show that mTOR possesses unexpected tyrosine kinase activity and activates IGF-IR/InsR. Rapamycin induces the tyrosine phosphorylation and activation of IGF-IR/InsR, which is largely dependent on rictor and mTOR. Moreover, mTORC2 promotes ligand-induced activation of IGF-IR/InsR. IGF- and insulin-induced IGF-IR/InsR phosphorylation is significantly compromised in rictor-null cells. Insulin receptor substrate (IRS) directly interacts with SIN1 thereby recruiting mTORC2 to IGF-IR/InsR and promoting rapamycin- or ligand-induced phosphorylation of IGF-IR/InsR. mTOR exhibits tyrosine kinase activity towards the general tyrosine kinase substrate poly(Glu-Tyr) and IGF-IR/InsR. Both recombinant mTOR and immunoprecipitated mTORC2 phosphorylate IGF-IR and InsR on Tyr1131/1136 and Tyr1146/1151, respectively. These effects are independent of the intrinsic kinase activity of IGF-IR/InsR, as determined by assays on kinase-dead IGF-IR/InsR mutants. While both rictor and mTOR immunoprecitates from rictor(+/+) MCF-10A cells exhibit tyrosine kinase activity towards IGF-IR and InsR, mTOR immunoprecipitates from rictor(-/-) MCF-10A cells do not induce IGF-IR and InsR phosphorylation. Phosphorylation-deficient mutation of residue Tyr1131 in IGF-IR or Tyr1146 in InsR abrogates the activation of IGF-IR/InsR by mTOR. Finally, overexpression of rictor promotes IGF-induced cell proliferation. Our work identifies mTOR as a dual-specificity kinase and clarifies how mTORC2 promotes IGF-IR/InsR activation.


Subject(s)
Multiprotein Complexes/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , TOR Serine-Threonine Kinases/metabolism , Tyrosine/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , HEK293 Cells , Hep G2 Cells , Humans , Mechanistic Target of Rapamycin Complex 2 , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein , Sirolimus/metabolism
14.
Tumour Biol ; 36(8): 6507-13, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25820822

ABSTRACT

AflatoxinB1 (AFB1) is well known as a potent carcinogen. Epidemiological studies have shown an association between AFB1 exposure and lung cancer in humans. AFB1 can induce the mutations of genes such as tumor suppressor p53 through its metabolite AFB1-8,9-exo-epoxide, which acts as a mutagen to react with DNA. In addition, recent study demonstrates AFB1 positively regulates type I insulin-like growth factor receptor (IGF-IR) signaling in hepatoma cells. The current study aims to determine the effects of AFB1 on Src kinase and insulin receptor substrate (IRS) in lung cancer cells and the effects of AFB1 on lung cancer cell migration. To this end, the effects of AFB1 on IRS expression, Src, Akt, and ERK phosphorylation were measured by Western blot analysis. The migration of lung cancer cells was detected by wound-healing assay. AFB1 downregulates IRS1 but paradoxically upregulates IRS2 through positive regulation of the stability of IRS2 and the proteasomal degradation of IRS1 in lung cancer cell lines A549 and SPCA-1. In addition, AFB1 induces Src, Akt, and ERK1/2 phosphorylation. Treatment of lung cancer cells with Src inhibitor saracatinib abrogates AFB1-induced IRS2 accumulation. Moreover, AFB1 stimulates lung cancer cell migration, which can be inhibited by saracatinib. We conclude that AFB1 may upregulate IRS2 and stimulate lung cancer cell migration through Src.


Subject(s)
Cell Movement/drug effects , Insulin Receptor Substrate Proteins/genetics , Lung Neoplasms/genetics , src-Family Kinases/genetics , Aflatoxin B1/toxicity , Benzodioxoles/administration & dosage , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Phosphorylation/drug effects , Quinazolines/administration & dosage , Signal Transduction/drug effects , src-Family Kinases/biosynthesis
15.
Int J Mol Sci ; 15(9): 16246-56, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25226534

ABSTRACT

c-Jun N-terminal kinases (JNK) are members of the mitogen-activated protein kinase (MAPK) family that have important roles in signal transduction. The small molecule SP600125 is widely used in biochemical studies as a JNK inhibitor. However, recent studies indicate that SP600125 may also act independent of JNK. Here, we report that SP600125 can induce Src, type I insulin-like growth factor receptor (IGF-IR), Akt and Erk1/2 phosphorylation. Notably, these effects are independent of its inhibition of JNK. Inhibition of Src abrogates the stimulation of IGF-IR, Akt and Erk1/2 phosphorylation. IGF-IR knockdown blunts the induction of both Akt and Erk1/2 phosphorylation by SP600125. Moreover, combination of SP600125 and the Src inhibitor saracatinib synergistically inhibits cell proliferation. We conclude that SP600125 can activate Src-IGF-IR-Akt/Erk1/2 signaling pathways independent of JNK.


Subject(s)
Anthracenes/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Receptor, IGF Type 1/metabolism , Up-Regulation/drug effects , src-Family Kinases/metabolism , Cell Line , Cell Proliferation/drug effects , HeLa Cells , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
16.
J Biol Chem ; 289(36): 24759-70, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25053419

ABSTRACT

Glycogen synthase kinase-3 (GSK3) has either tumor-suppressive roles or pro-tumor roles in different types of human tumors. A number of GSK3 targets in diverse signaling pathways have been uncovered, such as tuberous sclerosis complex subunit 2 and ß-catenin. The O subfamily of forkhead/winged helix transcription factors (FOXO) is known as tumor suppressors that induce apoptosis. In this study, we find that FOXO binds to type I insulin-like growth factor receptor (IGF-IR) promoter and stimulates its transcription. GSK3 positively regulates the transactivation activity of FOXO and stimulates IGF-IR expression. Although kinase-dead GSK3ß cannot up-regulate IGF-IR, the constitutively active GSK3ß induces IGF-IR expression in a FOXO-dependent manner. Serum starvation or Akt inhibition leads to an increase in IGF-IR expression, which could be blunted by GSK3 inhibition. GSK3ß knockdown or GSK3 inhibitor suppresses IGF-I-induced IGF-IR, Akt, and ERK1/2 phosphorylation. Moreover, knockdown of GSK3ß or FOXO1/3/4 leads to a decrease in cellular proliferation and abrogates IGF-I-induced hepatoma cell proliferation. These results suggest that GSK3 and FOXO may positively regulate IGF-I signaling and hepatoma cell proliferation.


Subject(s)
Forkhead Transcription Factors/genetics , Glycogen Synthase Kinase 3/genetics , Receptor, IGF Type 1/genetics , Transcription Factors/genetics , Blotting, Western , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Insulin-Like Growth Factor I/pharmacology , Models, Genetic , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Transcriptional Activation
17.
PLoS One ; 7(10): e47961, 2012.
Article in English | MEDLINE | ID: mdl-23112878

ABSTRACT

Aflatoxin B1 (AFB1) is a potent carcinogen that can induce hepatocellular carcinoma. AFB1-8,9-exo-epoxide, one of AFB1 metabolites, acts as a mutagen to react with DNA and induce gene mutations, including the tumor suppressor p53. In addition, AFB1 reportedly stimulates IGF receptor activation. Aberrant activation of IGF-I receptor (IGF-IR) signaling is tightly associated with various types of human tumors. In the current study, we investigated the effects of AFB1 on key elements in IGF-IR signaling pathway, and the effects of AFB1 on hepatoma cell migration. The results demonstrated that AFB1 induced IGF-IR, Akt, and Erk1/2 phosphorylation in hepatoma cell lines HepG2 and SMMC-7721, and an immortalized human liver cell line Chang liver. AFB1 also down-regulated insulin receptor substrate (IRS) 1 but paradoxically up-regulated IRS2 through preventing proteasomal degradation. Treatment of hepatoma cells and Chang liver cells with IGF-IR inhibitor abrogated AFB1-induced Akt and Erk1/2 phosphorylation. In addition, IRS2 knockdown suppressed AFB1-induced Akt and Erk1/2 phosphorylation. Finally, AFB1 stimulated hepatoma cell migration. IGF-IR inhibitor or IRS2 knockdown suppressed AFB1-induced hepatoma cell migration. These data demonstrate that AFB1 stimulates hepatoma cell migration through IGF-IR/IRS2 axis.


Subject(s)
Aflatoxin B1/metabolism , Carcinogens/metabolism , Carcinoma, Hepatocellular/metabolism , Insulin Receptor Substrate Proteins/metabolism , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Movement , Hep G2 Cells , Humans , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Receptor, IGF Type 1/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...