Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 528
Filter
1.
Epilepsia ; 58 Suppl 2: 50-59, 2017 06.
Article in English | MEDLINE | ID: mdl-28591479

ABSTRACT

The most common, and usually the only, endocrine disturbance in patients with hypothalamic hamartoma (HH) and epilepsy is central precocious puberty (CPP). The mechanism for CPP associated with HH may relate to ectopic generation and pulsatile release of gonadotropin-releasing hormone (GnRH) from the HH, but this remains an unproven hypothesis. Possible regulators of GnRH release that are intrinsic to HH tissue include the following: (1) glial factors (such as transforming growth factor α[TGFα) and (2) γ-aminobutyric acid (GABA)-mediated excitation. Both are known to be present in surgically-resected HH tissue, but are present in patients with and without a history of CPP, suggesting the possibility that symptoms related to HH are directly associated with the region of anatomic attachment of the HH to the hypothalamus, which determines functional network connections, rather than to differences in HH tissue expression or pathophysiology. CPP associated with HH presents with isosexual development prior to the age of 8 years in girls and 9 years in boys. It is not uncommon for CPP with HH to present in children at an earlier age in comparison to other causes of CPP, including in infancy. Surgical resection of the HH can be effective for treating CPP, but is reserved for patients with intractable epilepsy, since GnRH agonists are widely available and effective treatment. Other endocrine disturbances with HH are rare, but can include growth hormone deficiency, hypothyroidism, and adrenal insufficiency. Diabetes insipidus is commonly encountered postoperatively, but is not observed with HH prior to surgical intervention.


Subject(s)
Drug Resistant Epilepsy/physiopathology , Epilepsies, Partial/physiopathology , Hamartoma/physiopathology , Hypothalamic Diseases/physiopathology , Puberty, Precocious/physiopathology , Child , Child, Preschool , Comorbidity , Drug Resistant Epilepsy/diagnosis , Drug Resistant Epilepsy/therapy , Endocrine System Diseases/diagnosis , Endocrine System Diseases/physiopathology , Endocrine System Diseases/therapy , Epilepsies, Partial/diagnosis , Epilepsies, Partial/therapy , Female , Gonadotropin-Releasing Hormone/blood , Hamartoma/diagnosis , Hamartoma/therapy , Hormones, Ectopic/blood , Humans , Hypothalamic Diseases/diagnosis , Hypothalamic Diseases/therapy , Hypothalamus/physiopathology , Infant , Male , Nerve Net/physiopathology , Puberty, Precocious/diagnosis , Puberty, Precocious/therapy , Transforming Growth Factor alpha/physiology , gamma-Aminobutyric Acid/physiology
2.
Mol Neurobiol ; 54(2): 964-976, 2017 03.
Article in English | MEDLINE | ID: mdl-26797516

ABSTRACT

Transforming growth factor-alpha (TGF-α) is known to play multifunctional roles in the central nervous system (CNS), including the provision of neurotropic properties that protect neurons against various neurotoxic insults. Previously, we reported that TGF-α mediates estrogen-induced enhancement of glutamate transporter GLT-1 function in astrocytes. However, the regulatory mechanism of TGF-α at the transcriptional level remains to be established. Our findings revealed that the human TGF-α promoter contains consensus sites for several transcription factors, such as NF-κB and yin yang 1 (YY1). NF-κB served as a positive regulator of TGF-α promoter activity, corroborated by observations that overexpression of NF-κB p65 increased, while mutation in the NF-κB binding sites in the TGF-α promoter reduced the promoter activity in rat primary astrocytes. Pharmacological inhibition of NF-κB with pyrrolidine dithiocarbamate (PDTC; 50 µM) or quinazoline (QNZ; 10 µM) also abolished TGF-α promoter activity, and NF-κB directly bound to its consensus site in the TGF-α promoter as evidenced by electrophoretic mobility shift assay (EMSA). Dexamethasone (DX) increased TGF-α promoter activity by activation of NF-κB. Treatment of astrocytes with 100 nM of DX for 24 h activated its glucocorticoid receptor and signaling proteins, including MAPK, PI3K/Akt, and PKA, via non-genomic pathways, to enhance TGF-α promoter activity and expression. YY1 served as a critical negative regulator of the TGF-α promoter as overexpression of YY1 decreased, while mutation of YY1 binding site in the promoter increased TGF-α promoter activity. Treatment for 3 h with 250 µM of manganese (Mn), an environmental neurotoxin, decreased astrocytic TGF-α expression by activation of YY1. Taken together, our results suggest that NF-κB is a critical positive regulator, whereas YY1 is a negative regulator of the TGF-α promoter. These findings identify potential molecular targets for neurotherapeutics that may modulate TGF-α regulation and afford neuroprotection.


Subject(s)
Astrocytes/physiology , Transcription, Genetic/physiology , Transforming Growth Factor alpha/physiology , Animals , Astrocytes/drug effects , Base Sequence , Cells, Cultured , Humans , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
3.
Oncol Rep ; 35(2): 1153-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26718770

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) endows cancer cells with enhanced invasive and metastatic potential during cancer progression. Fractalkine, also known as chemokine (C-X3-C motif) ligand 1 (CX3CL1), the only member recognized so far that belongs to the CX3C chemokine subfamily, was reported to participate in the molecular events that regulate cell adhesion, migration and survival of human prostate cancer cells. However, the relationship between CX3CL1 and EMT remains unknown. We treated DU145 and PC-3 cells with CX3CL1 under hypoxic conditions. The migration and invasion abilities of DU145 and PC-3 cells were detected by Transwell assays. Induction of EMT was verified by morphological changes in the DU145 and PC-3 cells and analysis of protein expression of EMT markers such as E-cadherin and vimentin. To identify the involved signaling pathway in CX3CL1-induced EMT, activation of epidermal growth factor receptor (EGFR) was measured using western blot analysis, and Slug expression was detected with or without an EGFR inhibitor prior to CX3CL1 treatment. Concentrations of soluble and total TGF-α in the CX3CL­treated DU145 cells were detected by ELISA. Additionally, we determined the involvement of the TACE/TGF-α/EGFR pathway in CX3CL1­induced EMT using RNA interference and specific inhibitors. CX3CL1 increased the migration and invasiveness of the DU145 and PC-3 cells, and resulted in characteristic alterations of EMT. Our results demonstrated that TACE/TGF-α/EGFR pathway activation and subsequent upregulation of Slug expression were responsible for CX3CL1­induced EMT, and contributed to the migration and inva-sion of prostate cancer cells. Inhibition of TACE/TGF-α/EGFR signaling reversed EMT and led to reduced migration and invasion abilities of the prostate cancer cells. We provide initial evidence that CX3CL1 exposure resulted in EMT occurrence and enhancement of cell migration and invasion through a mechanism involving activation of TACE/TGF-α/EGFR signaling. These findings revealed that CX3CL1 may serve as a new target for the treatment of prostate cancer.


Subject(s)
ADAM Proteins/physiology , Adenocarcinoma/pathology , Chemokine CX3CL1/physiology , Epithelial-Mesenchymal Transition/physiology , ErbB Receptors/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Transforming Growth Factor alpha/physiology , ADAM Proteins/genetics , ADAM17 Protein , Adenocarcinoma/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation
4.
Arthritis Rheumatol ; 67(10): 2691-701, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26138996

ABSTRACT

OBJECTIVE: Transforming growth factor α (TGFα) is increased in osteoarthritic (OA) cartilage in rats and humans and modifies chondrocyte phenotype. CCL2 is increased in OA cartilage and stimulates proteoglycan loss. This study was undertaken to test whether TGFα and CCL2 cooperate to promote cartilage degradation and whether inhibiting either reduces disease progression in a rat model of posttraumatic OA. METHODS: Microarray analysis was used to profile expression of messenger RNA (mRNA) for Tgfa, Ccl2, and related genes in a rat model of posttraumatic OA. Rat primary chondrocytes and articular cartilage explants were treated with TGFα in the presence or absence of MEK-1/2, p38, phosphatidylinositol 3-kinase, Rho-associated protein kinase, or CCR2 inhibitors and immunostained for markers of cartilage degradation. The rat model was used to administer pharmacologic inhibitors of TGFα (AG1478) and CCL2 (RS504393) signaling for up to 10 weeks and assess histopathology and serum biomarkers of cartilage synthesis (C-propeptide of type II collagen [CPII]) and breakdown (C2C). RESULTS: Tgfa and Ccl2 mRNA were simultaneously up-regulated in articular cartilage in the rat model of posttraumatic OA. TGFα induced expression of CCL2, Mmp3, and Tnf in primary chondrocytes. Cleavage of type II collagen and aggrecan (by matrix metalloproteinases and ADAMTS-4/5, respectively) induced by TGFα was blocked by pharmacologic inhibition of CCL2 in cartilage explants. In vivo pharmacologic inhibition of TGFα or CCL2 signaling reduced Osteoarthritis Research Society International cartilage histopathology scores and increased serum CPII levels, but only TGFα inhibition reduced C2C levels intreated versus untreated rat OA cartilage. CONCLUSION: TGFα signaling stimulates cartilage degradation via a CCL2-dependent mechanism, but pharmacologic inhibition of the TGFα-CCL2 axis reduces experimental posttraumatic OA progression in vivo.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Disease Progression , Osteoarthritis/prevention & control , Osteoarthritis/physiopathology , Signal Transduction/physiology , Wounds and Injuries/complications , Animals , Benzoxazines/pharmacology , Cartilage, Articular/pathology , Cartilage, Articular/physiopathology , Chemokine CCL2/drug effects , Chemokine CCL2/physiology , Disease Models, Animal , Male , Osteoarthritis/etiology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spiro Compounds/pharmacology , Transforming Growth Factor alpha/antagonists & inhibitors , Transforming Growth Factor alpha/drug effects , Transforming Growth Factor alpha/physiology , Tyrphostins/pharmacology , Up-Regulation/physiology
5.
Exp Cell Res ; 333(1): 116-26, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25743092

ABSTRACT

Fibronectin (FN), an extracellular matrix ligand, plays a pivotal role in cell adhesion, migration, and oncogenic transformation. Aberrant FN expression is associated with poor prognoses in various types of cancer, including breast cancer. In the current study, we investigated the relationship between FN induction and HER2 expression in breast cancer cells. Our results showed that the level of FN expression increased in response to HER family ligands, EGF and TGF-α in a time- and dose-dependent manner. On the other hand, EGF-induced FN expression decreased in response to trastuzumab, which is a HER2-targeted monoclonal antibody. However, EGF-induced FN expression was not affected by trastuzumab in JIMT-1 breast cancer cells, which are trastuzumab insensitive cells. Next, we introduced the HER2 gene into MDA-MB231 cells to verify the relationship between FN and HER2. The level of FN expression significantly increased in HER2-overexpressed MDA-MB231 cells. In contrast, the induction of FN by HER2 was significantly decreased in response to trastuzumab treatment. In addition, the induction of FN by HER2 was down-regulated by the MEK 1/2 specific inhibitor, U0126. Using conditioned culture media of vec- and HER2-overexpressed MDA-MB231 cells, we observed the cell morphology, adhesion, and invasion of MDA-MB231 cells. Interestingly, in conditioned culture media of HER2-overexpressed MDA-MB231 cells, the cell morphology was altered, and adhesion and invasion of MDA-MB231 cells significantly increased. In addition, our results showed that recombinant human FN augmented cell adhesion and invasion of MDA-MB231 cells while these inductions decreased in response to an FN inhibitor. Therefore, we demonstrated that the induction of FN by HER2 triggers cell adhesion and invasion capacities.


Subject(s)
Cell Adhesion , Fibronectins/metabolism , Receptor, ErbB-2/physiology , Transcriptional Activation , Breast Neoplasms , Cell Line, Tumor , Cell Shape , Epidermal Growth Factor/physiology , Female , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Neoplasm Invasiveness , Transforming Growth Factor alpha/physiology
6.
Am J Respir Cell Mol Biol ; 52(4): 471-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25180535

ABSTRACT

Staphylococcus aureus (SA) colonization and infection is common, and may promote allergic or inflammatory airway diseases, such as asthma, cystic fibrosis, and chronic rhinosinusitis by interacting with airway epithelial cells. Airway epithelial cells not only comprise a physical barrier, but also play key roles in immune, inflammatory, repair, and remodeling responses upon encounters with pathogens. To elucidate the impact of SA on epithelial-mediated remodeling of allergic airways, we tested the hypothesis that SA can enhance the remodeling process. Normal human bronchial epithelial (NHBE) cells were stimulated with heat-killed SA (HKSA) or transforming growth factor (TGF) α. Cell extracts were collected to measure mRNA (real-time RT-PCR) and signaling molecules (Western blot); supernatants were collected to measure protein (ELISA) after 24 hours of stimulation. Epidermal growth factor receptor (EGFR) signaling inhibition experiments were performed using a specific EGFR kinase inhibitor (AG1478) and TGF-α was blocked with an anti-TGF-α antibody. HKSA induced both mRNA and protein for TGF-α and matrix metalloproteinase (MMP) 1 from NHBE cells by a Toll-like receptor 2-dependent mechanism. Recombinant human TGF-α also induced mRNA and protein for MMP-1 from NHBE cells; anti-TGF-α antibody inhibited HKSA-induced MMP-1, suggesting that endogenous TGF-α mediates the MMP-1 induction by HKSA. HKSA-induced MMP-1 expression was suppressed when a specific EGFR kinase inhibitor was added, suggesting that EGFR signaling was mediating the HKSA-induced MMP-1 release. Exposure or colonization by SA in the airway may enhance the remodeling of tissue through a TGF-α-dependent induction of MMP-1 expression, and may thereby promote remodeling in airway diseases in which SA is implicated, such as asthma and chronic rhinosinusitis.


Subject(s)
Airway Remodeling , ErbB Receptors/metabolism , Respiratory Mucosa/metabolism , Toll-Like Receptor 2/metabolism , Cells, Cultured , Enzyme Induction , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , NF-kappa B/metabolism , Signal Transduction , Transforming Growth Factor alpha/physiology
7.
Development ; 141(20): 3910-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25231760

ABSTRACT

In Drosophila melanogaster, the anteroposterior (AP) and dorsoventral (DV) axes of the oocyte and future embryo are established through the localization and translational regulation of gurken (grk) mRNA. This process involves binding of specific factors to the RNA during transport and a dynamic remodeling of the grk-containing ribonucleoprotein (RNP) complexes once they have reached their destination within the oocyte. In ovaries of spindle-class females, an activated DNA damage checkpoint causes inefficient Grk translation and ventralization of the oocyte. In a screen for modifiers of the oocyte DV patterning defects, we identified a mutation in the eIF1A gene as a dominant suppressor. We show that reducing the function of eIF1A in spnB ovaries suppresses the ventralized eggshell phenotype by restoring Grk expression. This suppression is not the result of more efficient DNA damage repair or of disrupted checkpoint activation, but is coupled to an increase in the amount of grk mRNA associated with polysomes. In spnB ovaries, the activated meiotic checkpoint blocks Grk translation by disrupting the accumulation of grk mRNA in a translationally competent RNP complex that contains the translational activator Oo18 RNA-binding protein (Orb); this regulation involves the translational repressor Squid (Sqd). We further propose that reduction of eIF1A allows more efficient Grk translation possibly because of the presence of specific structural features in the grk 5'UTR.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Eukaryotic Initiation Factor-1/metabolism , Gene Expression Regulation, Developmental , Oogenesis , Transforming Growth Factor alpha/physiology , Animals , DNA Damage , Drosophila Proteins/metabolism , Egg Proteins/metabolism , Eukaryotic Initiation Factor-1/genetics , Female , Genotype , Male , Meiosis , Mutation , Oocytes/cytology , Ovary/metabolism , Phenotype , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribonucleoproteins/chemistry
8.
Radiat Res ; 182(3): 350-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25117621

ABSTRACT

Radiation fibrosis of the lung is a late toxicity of thoracic irradiation. Epidermal growth factor (EGF) signaling has previously been implicated in radiation lung injury. We hypothesized that TGF-α, an EGF receptor ligand, plays a key role in radiation-induced fibrosis in lung. Mice deficient in transforming growth factor (TGF-α(-/-)) and control C57Bl/6J (C57-WT) mice were exposed to thoracic irradiation in 5 daily fractions of 6 Gy. Cohorts of mice were followed for survival (n ≥ 5 per group) and tissue collection (n = 3 per strain and time point). Collagen accumulation in irradiated lungs was assessed by Masson's trichrome staining and analysis of hydroxyproline content. Cytokine levels in lung tissue were assessed with ELISA. The effects of TGF-α on pneumocyte and fibroblast proliferation and collagen production were analyzed in vitro. Lysyl oxidase (LOX) expression and activity were measured in vitro and in vivo. Irradiated C57-WT mice had a median survival of 24.4 weeks compared to 48.2 weeks for irradiated TGF-α(-/-) mice (P = 0.001). At 20 weeks after irradiation, hydroxyproline content was markedly increased in C57-WT mice exposed to radiation compared to TGF-α(-/-) mice exposed to radiation or unirradiated C57-WT mice (63.0, 30.5 and 37.6 µg/lung, respectively, P = 0.01). C57-WT mice exposed to radiation had dense foci of subpleural fibrosis at 20 weeks after exposure, whereas the lungs of irradiated TGF-α (-/-) mice were largely devoid of fibrotic foci. Lung tissue concentrations of IL-1ß, IL-4, TNF-α, TGF-ß and EGF at multiple time points after irradiation were similar in C57-WT and TGF-α(-/-) mice. TGF-α in lung tissue of C57-WT mice rose rapidly after irradiation and remained elevated through 20 weeks. TGF-α(-/-) mice had lower basal LOX expression than C57-WT mice. Both LOX expression and LOX activity were increased after irradiation in all mice but to a lesser degree in TGF-α(-/-) mice. Treatment of NIH-3T3 fibroblasts with TGF-α resulted in increases in proliferation, collagen production and LOX activity. These studies identify TGF-α as a critical mediator of radiation-induced lung injury and a novel therapeutic target in this setting. Further, these data implicate TGF-α as a mediator of collagen maturation through a TGF-ß independent activation of lysyl oxidase.


Subject(s)
Lung/radiation effects , Radiation Injuries/etiology , Transforming Growth Factor alpha/physiology , Animals , Collagen/metabolism , Cytokines/biosynthesis , Female , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Fibrosis/etiology , Radiation Injuries/metabolism , Radiation Injuries/pathology
9.
Am J Physiol Renal Physiol ; 307(5): F551-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24899059

ABSTRACT

Polycystic kidney disease (PKD) is a common genetic disorder leading to cyst formation in the kidneys and other organs that ultimately results in kidney failure and death. Currently, there is no therapy for slowing down or stopping the progression of PKD. In this study, we identified the disintegrin metalloenzyme 17 (ADAM17) as a key regulator of cell proliferation in kidney tissues of conditional knockout Ift88(-/-) mice and collecting duct epithelial cells from Ift88°(rpk) mice, animal models of autosomal recessive polycystic kidney disease (ARPKD). Using Western blotting, an enzyme activity assay, and a growth factor-shedding assay in the presence or absence of the specific ADAM17 inhibitor TMI-005, we show that increased expression and activation of ADAM17 in the cystic kidney and in collecting duct epithelial cells originating from the Ift88°(rpk) mice (designated as PKD cells) lead to constitutive shedding of several growth factors, including heparin-binding EGF-like growth factor (HB-EGF), amphiregulin, and transforming growth factor-α (TGF-α). Increased growth factor shedding induces activation of the EGFR/MAPK/ERK pathway and maintains higher cell proliferation rate in PKD cells compared with control cells. PKD cells also displayed increased lactate formation and extracellular acidification indicative of aerobic glycolysis (Warburg effect), which was blocked by ADAM17 inhibition. We propose that ADAM17 is a key promoter of cellular proliferation in PKD cells by activating the EGFR/ERK axis and a proproliferative glycolytic phenotype.


Subject(s)
ADAM Proteins/physiology , Cell Proliferation/physiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/physiology , Glycolysis/physiology , Kidney Tubules, Collecting/pathology , Polycystic Kidney Diseases/physiopathology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/drug effects , ADAM17 Protein , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/physiology , ErbB Receptors/physiology , Female , Heparin-binding EGF-like Growth Factor/physiology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/physiopathology , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Transforming Growth Factor alpha/physiology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
10.
Toxicol Appl Pharmacol ; 279(1): 76-86, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24844442

ABSTRACT

Amorphous silica nanoparticles (SiNPs) have previously been shown to induce marked cytokine (interleukin-6; IL-6 and interleukin-8; CXCL8/IL-8) responses independently of particle uptake in human bronchial epithelial BEAS-2B cells. In this study the involvement of the mitogen-activated protein kinases (MAP-kinases), nuclear factor-kappa Β (NF-κΒ) and in particular tumour necrosis factor-α converting enzyme (TACE) and-epidermal growth factor receptor (EGFR) signalling pathways were examined in triggering of IL-6 and CXCL8 release after exposure to a 50nm silica nanoparticle (Si50). Exposure to Si50 increased phosphorylation of NF-κΒ p65 and MAP-kinases p38 and JUN-N-terminal protein kinase pathways (JNK), but not extracellular signal regulated kinases (ERK). Inhibition of NF-κΒ and p38 reduced the cytokine responses to Si50, whereas neither JNK- nor ERK-inhibition exerted any significant effect on the responses to Si50. Increases in membrane-bound transforming growth factor-α (TGF-α) release and EGFR phosphorylation were also observed after Si50 exposure, and pre-treatment with inhibitors of these pathways reduced the release of IL-6 and CXCL8, but did not affect the Si50-induced phosphorylation of p38 and p65. In contrast, p38-inhibition partially reduced Si50-induced TGF-α release, while the p65-inhibition was without effect. Overall, our results indicate that Si50-induced IL-6 and CXCL8 responses in BEAS-2B cells were regulated through combined activation of several pathways, including NF-κΒ and p38/TACE/TGF-α/EGFR signalling. The study identifies critical, initial events in the triggering of pro-inflammatory responses by nanoparticles.


Subject(s)
ADAM Proteins/physiology , Cytokines/biosynthesis , Epithelial Cells/metabolism , ErbB Receptors/physiology , Lung/metabolism , NF-kappa B/physiology , Nanoparticles/toxicity , Silicon Dioxide/toxicity , Transforming Growth Factor alpha/physiology , p38 Mitogen-Activated Protein Kinases/physiology , ADAM17 Protein , Blotting, Western , Cell Line , Cell Survival/drug effects , Humans , Interleukin-5/biosynthesis , Interleukin-8/biosynthesis , Lung/cytology , Lung/drug effects , Phosphorylation , Signal Transduction/drug effects , Transcription Factor RelA/biosynthesis , Transcription Factor RelA/genetics
11.
Life Sci ; 100(1): 25-34, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24496038

ABSTRACT

AIMS: The mechanism by which SR48692 inhibits non-small cell lung cancer (NSCLC) proliferation was investigated. MAIN METHODS: The ability of SR48692 to inhibit the proliferation of NSCLC cell lines NCI-H1299 and A549 was investigated in vitro in the presence or absence of neurotensin (NTS). The ability of NTS to cause epidermal growth factor receptor (EGFR) transactivation was investigated by Western blot using NSCLC cells and various inhibitors. The growth effects and Western blot results were determined in cell lines treated with siRNA for NTSR1. KEY FINDINGS: Treatment of A549 or NCI-H1299 cells with siRNA for NTSR1 reduced significantly NTSR1 protein and the ability of SR48692 to inhibit the proliferation of A549 or NCI-H1299 NSCLC cells. Treatment of A549 and NCI-H1299 cells with siRNA for NTSR1 reduced the ability of NTS to cause epidermal growth factor receptor (EGFR) transactivation. SR48692 or gefitinib (EGFR tyrosine kinase inhibitor) inhibited the ability of NTS to cause EGFR and ERK tyrosine phosphorylation. NTS transactivation of the EGFR was inhibited by GM6001 (matrix metalloprotease inhibitor), Tiron (superoxide scavenger) or U73122 (phospholipase C inhibitor) but not H89 (PKA inhibitor). NTS stimulates whereas SR48692 or gefitinib inhibits the clonal growth of NSCLC cells. SIGNIFICANCE: These results suggest that SR48692 may inhibit NSCLC proliferation in an EGFR-dependent mechanism.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Pyrazoles/pharmacology , Quinolines/pharmacology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases , Gefitinib , Gene Knockdown Techniques , Humans , Phosphorylation , Protein Processing, Post-Translational , Quinazolines/pharmacology , RNA, Small Interfering/genetics , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Transcriptional Activation , Transforming Growth Factor alpha/physiology
12.
Cancer Res ; 74(7): 2062-72, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24520077

ABSTRACT

Secondary bile acids (BA) such as deoxycholic acid (DCA) promote the development of several gastrointestinal malignancies, but how they mediate this effect is unclear. In this study, we offer evidence of a mechanism involving ectodomain shedding of the EGFR ligands amphiregulin (AREG) and TGF-α, which rely upon the cell surface protease TACE/ADAM-17. Specifically, we show that AREG participates in DCA-induced EGFR and STAT3 signaling, cell-cycle progression, and tumorigenicity in human colorectal cancer and pancreatic ductal adenocarcinoma (PDAC). TACE and AREG, but not TGF-α, were overexpressed in both colorectal cancer and PDAC tissues compared with normal tissues. Exposure of colorectal cancer and PDAC cells to DCA resulted in colocalization of Src and TACE to the cell membrane, resulting in AREG-dependent activation of EGFR, mitogen-activated protein kinase (MAPK), and STAT3 signaling. Src or TACE inhibition was sufficient to attenuate DCA-induced AREG, but not TGF-α shedding. We also examined a role for the BA transporter TGR5 in DCA-mediated EGFR and STAT3 signaling. RNA interference-mediated silencing of TGR5 or AREG inhibited DCA-induced EGFR, MAPK, and STAT3 signaling, blunted cyclin D1 expression and cell-cycle progression, and attenuated DCA-induced colorectal cancer or PDAC tumorigenicity. Together, our findings define an AREG-dependent signaling pathway that mediates the oncogenic effects of secondary BAs in gastrointestinal cancers, the targeting of which may enhance therapeutic responses in their treatment.


Subject(s)
Bile Acids and Salts/toxicity , Gastrointestinal Neoplasms/chemically induced , Glycoproteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Transforming Growth Factor alpha/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Amphiregulin , Cyclin D1/genetics , Deoxycholic Acid/toxicity , EGF Family of Proteins , ErbB Receptors/physiology , HCT116 Cells , Humans , Pancreas/metabolism , Phosphorylation , Receptors, G-Protein-Coupled/physiology , STAT3 Transcription Factor/physiology , src-Family Kinases/metabolism
13.
Nephrol Dial Transplant ; 27(10): 3686-91, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23114895

ABSTRACT

Renal fibrosis is a major hallmark of chronic kidney disease, regardless of the initial causes, and prominent renal fibrosis predicts poor prognosis for renal insufficiency. Transforming growth factor (TGF)-ß plays a pivotal role in the progression of renal fibrosis, and therapeutic interventions targeting TGF-ß have been successful and well tolerated in animal models. However, these interventions might have adverse effects by inducing systemic inflammation due to the strong bifunctional role of TGF-ß (pro-fibrotic and anti-inflammatory). This review of the current literature focuses on the inhibitors/antagonists of TGF-ß, and discusses possible therapeutic approaches targeting them, describing the effectiveness of orally active bone morphogenetic protein 7 mimetics in reversing established fibrosis. It will conclude with a brief discussion of possible future directions for research.


Subject(s)
Kidney/pathology , Renal Insufficiency, Chronic/drug therapy , Transforming Growth Factor alpha/antagonists & inhibitors , Animals , Bone Morphogenetic Protein 7/agonists , Bone Morphogenetic Protein 7/antagonists & inhibitors , Bone Morphogenetic Protein 7/physiology , Fibrosis , Humans , Kidney/drug effects , Kidney/physiopathology , Renal Insufficiency, Chronic/physiopathology , Signal Transduction , Transforming Growth Factor alpha/physiology
14.
Nat Methods ; 9(10): 1021-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22983457

ABSTRACT

A single-format method to detect multiple G protein-coupled receptor (GPCR) signaling, especially Gα(12/13) signaling, presently has limited throughput and sensitivity. Here we report a transforming growth factor-α (TGFα) shedding assay, in which GPCR activation is measured as ectodomain shedding of a membrane-bound proform of alkaline phosphatase-tagged TGFα (AP-TGFα) and its release into conditioned medium. AP-TGFα shedding response occurred almost exclusively downstream of Gα(12/13) and Gα(q) signaling. Relying on chimeric Gα proteins and promiscuous Gα(16) protein, which can couple with Gα(s)- and Gα(i)-coupled GPCRs and induce Gα(q) signaling, we used the TGFα shedding assay to detect 104 GPCRs among 116 human GPCRs. We identified three orphan GPCRs (P2Y10, A630033H20 and GPR174) as Gα(12/13)-coupled lysophosphatidylserine receptors. Thus, the TGFα shedding assay is useful for studies of poorly characterized Gα(12/13)-coupled GPCRs and is a versatile platform for detecting GPCR activation including searching for ligands of orphan GPCRs.


Subject(s)
Receptors, G-Protein-Coupled/analysis , Transforming Growth Factor alpha/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , HEK293 Cells , Humans , Lysophospholipids/metabolism , Receptors, G-Protein-Coupled/physiology , Receptors, Purinergic P2/metabolism , Signal Transduction
16.
Mol Pharmacol ; 82(5): 948-57, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22909796

ABSTRACT

Reactive astrogliosis is beneficial in many aspects; however, it is also detrimental in some pathological states such as the development of lethal brain tumors. It is therefore crucial to understand the mechanisms regulating astrocyte proliferation. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor family, was shown to stimulate astrocyte proliferation in vitro. Herein, we further characterize the mitogenic potential of TWEAK on central nervous system cells. Among these cells, astrocytes express the highest level of TWEAK and Fn14 transcripts, suggesting that they are particularly sensitive to TWEAK stimulation. Using in vitro model systems, we found that TWEAK was as potent as epidermal growth factor (EGF) (a prototypical astrocyte mitogen) in mediating astrocyte proliferation. However, its mitogenic activity was delayed compared with that of EGF, suggesting distinct mechanisms of action. Using cell signaling pathway inhibitors, neutralizing antibodies, and protein assays, we further show that the mitogenic activity of TWEAK on primary astrocytes requires stimulation of the transforming growth factor-α (TGF-α) and of the epidermal growth factor receptor (EGFR) signaling pathway through extracellular signal-regulated kinase and p38 mitogen-activated protein kinase activation. In aggregates, our data demonstrate that TWEAK acts as a potent astrocyte mitogen through the induction of a TGF-α/EGFR signaling pathway. We anticipate that description of such a mechanism may allow novel approaches to human pathologies associated with astrocyte proliferation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Astrocytes/cytology , ErbB Receptors/physiology , Membrane Proteins/metabolism , Transforming Growth Factor alpha/physiology , Tumor Necrosis Factors/metabolism , Animals , Apoptosis Regulatory Proteins/pharmacology , Astrocytes/metabolism , Cell Proliferation , Cytokine TWEAK , Embryo, Mammalian , Enzyme Activation , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Membrane Proteins/pharmacology , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Primary Cell Culture , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , TWEAK Receptor , Tumor Necrosis Factors/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
FEBS J ; 279(18): 3290-313, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22443451

ABSTRACT

During liver regeneration, quiescent hepatocytes re-enter the cell cycle to proliferate and compensate for lost tissue. Multiple signals including hepatocyte growth factor, epidermal growth factor, tumor necrosis factor α, interleukin-6, insulin and transforming growth factor ß orchestrate these responses and are integrated during the G(1) phase of the cell cycle. To investigate how these inputs influence DNA synthesis as a measure for proliferation, we established a large-scale integrated logical model connecting multiple signaling pathways and the cell cycle. We constructed our model based upon established literature knowledge, and successively improved and validated its structure using hepatocyte-specific literature as well as experimental DNA synthesis data. Model analyses showed that activation of the mitogen-activated protein kinase and phosphatidylinositol 3-kinase pathways was sufficient and necessary for triggering DNA synthesis. In addition, we identified key species in these pathways that mediate DNA replication. Our model predicted oncogenic mutations that were compared with the COSMIC database, and proposed intervention targets to block hepatocyte growth factor-induced DNA synthesis, which we validated experimentally. Our integrative approach demonstrates that, despite the complexity and size of the underlying interlaced network, logical modeling enables an integrative understanding of signaling-controlled proliferation at the cellular level, and thus can provide intervention strategies for distinct perturbation scenarios at various regulatory levels.


Subject(s)
DNA Replication , Hepatocytes/metabolism , Signal Transduction/physiology , Animals , Cell Cycle/physiology , Cell Proliferation , DNA Replication/drug effects , Epidermal Growth Factor/physiology , Hepatocyte Growth Factor/physiology , Insulin/physiology , Interleukin-6/physiology , Liver Regeneration/physiology , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Systems Biology , Transforming Growth Factor alpha/physiology , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor-alpha/physiology
18.
Dev Biol ; 365(1): 71-81, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22366186

ABSTRACT

Regulation of intracellular pH (pHi) and protection against cytosolic acidification is primarily a function of the ubiquitous plasma membrane Na+/H+exchanger-1 (NHE1), which uses a highly conserved process to transfer cytosolic hydrogen ions (H+) across plasma membranes in exchange for extracellular sodium ions (Na+). Growth factors, which are essential regulators of morphogenesis, have also been found to be key activators of NHE1 exchanger activity; however, the crosstalk between both has not been fully evaluated during organ development. Here we report that mammary branching morphogenesis induced by transforming growth factor-alpha (TGFα) requires PI3K-dependent NHE1-activation and subsequent pHi alkalization. Inhibiting NHE1 activity after TGFα stimulation with 10 µM of the NHE1-specific inhibitor N-Methyl-N-isobutyl Amiloride (MIA) dramatically disrupted branching morphogenesis, induced extensive proliferation, ectopic expression of the epithelial hyper-proliferative marker Keratin-6 and sustained activation of MAPK. Together these findings indicate a novel developmental signaling cascade involving TGFα>PI3K>NHE1>pHi alkalization, which leads to a permissible environment for MAPK negative feedback inhibition and thus regulated mammary branching morphogenesis.


Subject(s)
Cation Transport Proteins/physiology , Mammary Glands, Animal/embryology , Sodium-Hydrogen Exchangers/physiology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Female , Hydrogen-Ion Concentration , Keratin-6 , Mammary Glands, Animal/physiology , Mice , Morphogenesis/drug effects , Morphogenesis/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Sodium-Hydrogen Exchanger 1 , Transforming Growth Factor alpha/physiology
19.
Trans Am Clin Climatol Assoc ; 123: 126-33; discussion 133-4, 2012.
Article in English | MEDLINE | ID: mdl-23303980

ABSTRACT

In 1888, Pierre Ménétrier first described the disease that bears his name. Many of the findings he reported then remain accepted features of the disease. Based on studies performed in our laboratory over the past 20 years, we have implicated increased transforming growth factor-α (TGFα) expression and heightened epidermal growth factor receptor (EGFR) activity in the pathogenesis of Ménétrier's disease. Herein, we provide a historical perspective of this rare disorder, review our experience with Ménétrier's disease, and discuss future challenges and opportunities posed by this disorder.


Subject(s)
Gastritis, Hypertrophic/history , Gastritis, Hypertrophic/physiopathology , ErbB Receptors/physiology , France , Gastritis, Hypertrophic/etiology , History, 19th Century , History, 20th Century , History, 21st Century , Transforming Growth Factor alpha/physiology
20.
Dev Neurobiol ; 72(9): 1229-42, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22021126

ABSTRACT

Synaptic target selection is critical for establishing functional neuronal circuits. The mechanisms regulating target selection remain incompletely understood. We describe a role for the EGF receptor and its ligand Gurken in target selection of octopaminergic Type II neurons in the Drosophila neuromuscular system. Mutants in happyhour, a regulator of EGFR signaling, form ectopic Type II neuromuscular junctions. These ectopic innervations are due to inappropriate target selection. We demonstrate that EGFR signaling is necessary and sufficient to inhibit synaptic target selection by these octopaminergic Type II neurons, and that the EGFR ligand Gurken is the postsynaptic, muscle-derived repulsive cue. These results identify a new pathway mediating cell-type and branch-specific synaptic repulsion, a novel role for EGFR signaling in synaptic target selection, and an unexpected role for Gurken as a muscle-secreted repulsive ligand.


Subject(s)
Drosophila Proteins/physiology , ErbB Receptors/physiology , Receptors, Invertebrate Peptide/physiology , Signal Transduction/physiology , Synaptic Transmission/physiology , Transforming Growth Factor alpha/physiology , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , Transforming Growth Factor alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL