Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Biochem Biophys Res Commun ; 533(3): 424-428, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32972751

ABSTRACT

Nutrient stress driven by glutamine deficiency activates EGFR signaling in a subset of KRAS-mutant pancreatic ductal adenocarcinoma (PDAC) cells. EGFR signaling in the context of glutamine starvation is thought to be instigated by the transcriptional upregulation of EGFR ligands and functions as an adaptation mechanism to allow PDAC cells to maintain metabolic fitness. Having a clear view of the intricate signaling cascades potentiated by the metabolic induction of EGFR is important in understanding how these effector pathways influence cancer progression. In this study, we examined the complex signaling that occurs in PDAC cells when EGFR is activated by glutamine deprivation. We elucidate that the metabolic activation of EGFR is principally mediated by HB-EGF, and that other members of the ErbB receptor tyrosine kinase family are not activated by glutamine starvation. Additionally, we determine that glutamine depletion-driven EGFR signaling is associated with a specific receptor phosphorylation known to participate in a feedback loop, a process that is dependent on Erk. Lastly, we determine that K-Ras is required for glutamine depletion-induced Erk activation, as well as EGFR feedback phosphorylation, but is dispensable for Akt activation. These data provide important insights into the regulation of EGFR signaling in the context of metabolic stresses.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/physiology , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , ErbB Receptors/metabolism , Feedback, Physiological , Glutamine/physiology , Heparin-binding EGF-like Growth Factor/physiology , Humans , MAP Kinase Signaling System , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism
2.
Genes Cells ; 24(2): 112-125, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30474194

ABSTRACT

Type II alveolar epithelial cells (AEC2s) play a crucial role in the regeneration of type I AECs after acute lung injury. The mechanisms underlying the regeneration of AEC2s are not fully understood. To address this issue, here, we investigated a murine model of acute lung injury using mice expressing human Diphtheria Toxin Receptor (DTR) under the control of Lysozyme M promoter (LysM-DTR). DT injection induced the depletion of AEC2s, alveolar macrophages, and bone marrow (BM)-derived myeloid cells in LysM-DTR mice, and the mice died within 6 days after DT injection. Apoptotic AEC2s and bronchiolar epithelial cells appeared at 24 hr, whereas Ki67-positive proliferating cells appeared in the alveoli and bronchioles in the lung of LysM-DTR mice at 72-96 hr after DT injection. Transfer of wild-type BM cells into LysM-DTR mice accelerated the regeneration of AEC2s along with the up-regulation of several growth factors. Moreover, several metabolites were significantly decreased in the sera of LysM-DTR mice compared with WT mice after DT injection, suggesting that these metabolites might be biomarkers to predict AEC2s injury. Together, LysM-DTR mice might be useful to identify growth factors to promote lung repair and the metabolites to predict the severity of lung injury.


Subject(s)
Acute Lung Injury/prevention & control , Alveolar Epithelial Cells/cytology , Biomarkers/metabolism , Bone Marrow Transplantation , Heparin-binding EGF-like Growth Factor/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Metabolome , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Diphtheria Toxin/toxicity , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Promoter Regions, Genetic , Wound Healing
3.
Lab Invest ; 99(2): 150-157, 2019 02.
Article in English | MEDLINE | ID: mdl-30451982

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a progressive and devastating chronic lung condition that has a significant global burden, both medically and financially. Currently there are no medications that can alter the course of disease. At best, the drugs in clinical practice provide symptomatic relief to suffering patients by alleviating acute exacerbations. Most of current clinical research activities are in late severe disease with lesser attention given to early disease manifestations. There is as yet, a lack of understanding of the underlying mechanisms of disease progression and the molecular switches that are involved in their manifestation. Small airway fibrosis and obliteration are known to cause fixed airflow obstruction in COPD, and the consequential damage to the lung has an early onset. So far, there is little evidence of the mechanisms that underlie this aspect of pathology. However, emerging research confirms that airway epithelial reprogramming or epithelial to mesenchymal transition (EMT) is a key mechanism that drives fibrotic remodelling changes in smokers and patients with COPD. A recent study by Lai et al. further highlights the importance of EMT in smoking-related COPD pathology. The authors identify HB-EGF, an EGFR ligand, as a key driver of EMT and a potential new therapeutic target for the amelioration of EMT and airway remodelling. There are also wider implications in lung cancer prophylaxis, which is another major comorbidity associated with COPD. We consider that improved molecular understanding of the intricate pathways associated with epithelial cell plasticity in smokers and patients with COPD will have major therapeutic implications.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Heparin-binding EGF-like Growth Factor , Pulmonary Disease, Chronic Obstructive , Airway Remodeling/physiology , Heparin-binding EGF-like Growth Factor/metabolism , Heparin-binding EGF-like Growth Factor/physiology , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology
4.
Proc Natl Acad Sci U S A ; 112(7): E796-805, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646445

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), caused by dominant mutations in the NOTCH3 receptor in vascular smooth muscle, is a genetic paradigm of small vessel disease (SVD) of the brain. Recent studies using transgenic (Tg)Notch3(R169C) mice, a genetic model of CADASIL, revealed functional defects in cerebral (pial) arteries on the surface of the brain at an early stage of disease progression. Here, using parenchymal arterioles (PAs) from within the brain, we determined the molecular mechanism underlying the early functional deficits associated with this Notch3 mutation. At physiological pressure (40 mmHg), smooth muscle membrane potential depolarization and constriction to pressure (myogenic tone) were blunted in PAs from TgNotch3(R169C) mice. This effect was associated with an ∼ 60% increase in the number of voltage-gated potassium (KV) channels, which oppose pressure-induced depolarization. Inhibition of KV1 channels with 4-aminopyridine (4-AP) or treatment with the epidermal growth factor receptor agonist heparin-binding EGF (HB-EGF), which promotes KV1 channel endocytosis, reduced KV current density and restored myogenic responses in PAs from TgNotch3(R169C) mice, whereas pharmacological inhibition of other major vasodilatory influences had no effect. KV1 currents and myogenic responses were similarly altered in pial arteries from TgNotch3(R169C) mice, but not in mesenteric arteries. Interestingly, HB-EGF had no effect on mesenteric arteries, suggesting a possible mechanistic basis for the exclusive cerebrovascular manifestation of CADASIL. Collectively, our results indicate that increasing the number of KV1 channels in cerebral smooth muscle produces a mutant vascular phenotype akin to a channelopathy in a genetic model of SVD.


Subject(s)
Brain/physiopathology , Cerebrovascular Disorders/genetics , Potassium Channels/genetics , 4-Aminopyridine/pharmacology , Animals , Brain/blood supply , Cerebrovascular Disorders/physiopathology , Disease Models, Animal , Heparin-binding EGF-like Growth Factor/physiology , Membrane Potentials , Mice , Mice, Transgenic , Receptor, Notch3 , Receptors, Notch/genetics , Receptors, Notch/physiology
5.
Pediatr Res ; 82(3): 490-500, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28422949

ABSTRACT

BackgroundNeonatal necrotizing enterocolitis (NEC) is associated with alterations of the enteric nervous system (ENS), with loss of neuronal nitric oxide synthase (nNOS)-expressing neurons in the intestine. The aim of this study was to investigate the roles of heparin-binding EGF-like growth factor (HB-EGF) in neural stem cell (NSC) differentiation, nNOS expression, and effects on ENS integrity during experimental NEC.MethodsThe effects of HB-EGF on NSC differentiation and nNOS production were determined using cultured enteric NSCs. Myenteric neuronal subpopulations were examined in HB-EGF knockout mice. Rat pups were exposed to experimental NEC, and the effects of HB-EGF treatment on nNOS production and intestinal neuronal apoptosis were determined.ResultsHB-EGF promotes NSC differentiation, with increased nNOS production in differentiated neurons and glial cells. Moreover, loss of nNOS-expressing neurons in the myenteric plexus and impaired neurite outgrowth were associated with absence of the HB-EGF gene. In addition, administration of HB-EGF preserves nNOS expression in the myenteric plexus and reduces enteric neuronal apoptosis during experimental NEC.ConclusionHB-EGF promotes the differentiation of enteric NSCs into neurons in a nitric oxide (NO)-dependent manner, and protects the ENS from NEC-induced injury, providing new insights into potential therapeutic strategies for the treatment of NEC in the future.


Subject(s)
Enteric Nervous System/metabolism , Enterocolitis, Necrotizing/metabolism , Heparin-binding EGF-like Growth Factor/physiology , Nitric Oxide Synthase Type I/metabolism , Animals , Cell Differentiation , Female , Heparin-binding EGF-like Growth Factor/genetics , Intestinal Mucosa/metabolism , Mice , Neurons/metabolism , Nitric Oxide/biosynthesis , Pregnancy , Rats , Rats, Sprague-Dawley
6.
Osteoarthritis Cartilage ; 23(9): 1523-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25937027

ABSTRACT

OBJECTIVE: We determined if the epidermal growth factor receptor ligand HB-EGF is produced in cartilage and if it regulates chondrocyte anabolic or catabolic activity. METHODS: HB-EGF expression was measured by quantitative PCR using RNA isolated from mouse knee joint tissues and from normal and osteoarthritis (OA) human chondrocytes. Immunohistochemistry was performed on normal and OA human cartilage and meniscus sections. Cultured chondrocytes were treated with fibronectin fragments (FN-f) as a catabolic stimulus and osteogenic protein 1 (OP-1) as an anabolic stimulus. Effects of HB-EGF on cell signaling were analyzed by immunoblotting of selected signaling proteins. MMP-13 was measured in conditioned media, proteoglycan synthesis was measured by sulfate incorporation, and matrix gene expression by quantitative PCR. RESULTS: HB-EGF expression was increased in 12-month old mice at 8 weeks after surgery to induce OA and increased amounts of HB-EGF were noted in human articular cartilage from OA knees. FN-f stimulated chondrocyte HB-EGF expression and HB-EGF stimulated chondrocyte MMP-13 production. However, HB-EGF was not required for FN-f stimulation of MMP-13 production. HB-EGF activated the ERK and p38 MAP kinases and stimulated phosphorylation of Smad1 at an inhibitory serine site which was associated with inhibition of OP-1 mediated proteoglycan synthesis and reduced aggrecan (ACAN) but not COL2A1 expression. CONCLUSION: HB-EGF is a new factor identified in OA cartilage that promotes chondrocyte catabolic activity while inhibiting anabolic activity suggesting it could contribute to the catabolic-anabolic imbalance seen in OA cartilage.


Subject(s)
Chondrocytes/metabolism , Heparin-binding EGF-like Growth Factor/biosynthesis , Heparin-binding EGF-like Growth Factor/physiology , Osteoarthritis/metabolism , Aggrecans/analysis , Animals , Bone Morphogenetic Protein 7/pharmacology , Cartilage/metabolism , Cartilage, Articular/metabolism , Chondrocytes/drug effects , Collagen Type II/analysis , Fibronectins/pharmacology , Heparin-binding EGF-like Growth Factor/analysis , Humans , Immunoblotting , Immunohistochemistry , In Vitro Techniques , Knee Joint/metabolism , Matrix Metalloproteinase 13/analysis , Mice , Osteoarthritis, Knee/metabolism , Phosphorylation , Proteoglycans/biosynthesis , Real-Time Polymerase Chain Reaction , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
7.
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
8.
Metab Syndr Relat Disord ; 18(4): 186-196, 2020 05.
Article in English | MEDLINE | ID: mdl-32077785

ABSTRACT

Heparin-binding EGF-like growth factor (HB-EGF) is an EGF family member that interacts with epidermal growth factor receptor (EGFR) and ERBB4. Since HB-EGF was first identified as a novel growth factor secreted from a human macrophage cell line, numerous pathological and physiological functions related to cell proliferation, migration, and inflammation have been reported. Notably, the expression of HB-EGF is sensitively upregulated by oxidative stress in the endothelial cells and functions for auto- and paracrine-EGFR signaling. Overnutrition and obesity cause elevation of HB-EGF expression and EGFR signaling in the hepatic and vascular systems. Modulations of HB-EGF signaling showed a series of protections against phenotypes related to metabolic syndrome and advanced metabolic diseases, suggesting HB-EGF as a potential target against metabolic diseases.


Subject(s)
Heparin-binding EGF-like Growth Factor/physiology , Metabolic Diseases/etiology , Oxidative Stress/physiology , Animals , Atherosclerosis/epidemiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cardiometabolic Risk Factors , Dyslipidemias/epidemiology , Dyslipidemias/genetics , Dyslipidemias/metabolism , Humans , Insulin Resistance/genetics , Metabolic Diseases/epidemiology , Metabolic Diseases/metabolism , Phenotype , Signal Transduction/physiology
9.
Hear Res ; 374: 49-57, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30710792

ABSTRACT

The blood-perilymph barrier serves a critical role by separating the components of blood from inner ear fluids, limiting traffic of cells, proteins and other solutes into the labyrinth, and allowing gas (O2-CO2) exchange. Inflammation produces changes in the blood-perilymph barrier resulting in increased vascular permeability. It is commonly thought that compromise of the blood-inner ear barrier would lead to hearing impairment through loss of the endocochlear potential (EP). In fact, the effect of increasing cochlear vascular permeability on hearing function and EP is poorly understood. We used a novel method to measure the integrity of the blood-perilymph barrier and demonstrated the effects of barrier compromise on ABR threshold and EP. We also investigated the contribution of CX3CR1 cochlear macrophages and CCR2 inflammatory monocytes to barrier function after systemic exposure to lipopolysaccharide (LPS). We found that systemic LPS induced a profound change in vascular permeability, which correlated with minimal change in ABR threshold and EP. Macrophage depletion using CX3CR1-DTR mice did not alter the baseline permeability of cochlear vessels and resulted in preservation of barrier function in LPS-treated animals. We conclude that cochlear macrophages are not required to maintain the barrier in normal mice and activated macrophages are a critical factor in breakdown of the barrier after LPS. CCR2 null mice demonstrated that LPS induction of barrier leakiness occurs in the absence of CCR2 expression. Thus, enhanced aminoglycoside ototoxicity after LPS can be linked to the expression of CCR2 in inflammatory monocytes, and not to preservation of the blood-perilymph barrier in CCR2 knockout mice.


Subject(s)
Cochlea/blood supply , Cochlea/physiology , Macrophages/physiology , Monocytes/physiology , Perilymph/cytology , Perilymph/physiology , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cochlea/cytology , Diphtheria Toxin/toxicity , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/physiology , Lipopolysaccharides/toxicity , Macrophage Activation/physiology , Macrophages/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Ototoxicity/pathology , Ototoxicity/physiopathology , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/physiology
10.
Methods Mol Biol ; 1748: 203-228, 2018.
Article in English | MEDLINE | ID: mdl-29453574

ABSTRACT

Testis development and function is regulated by intricate cell-cell cross talk. Characterization of the mechanisms underpinning this has been derived through a wide variety of approaches including pharmacological manipulation, transgenics, and cell-specific ablation of populations. The removal of all or a proportion of a specific cell type has been achieved through a variety of approaches. In this paper, we detail a combined transgenic and pharmacological approach to ablate the Sertoli or germ cell populations using diphtheria toxin in mice. We describe the key steps in generation, validation, and use of the models and also describe the caveats and cautions necessary. We also provide a detailed description of the methodology applied to characterize testis development and function in models of postnatal Sertoli or germ cell ablation.


Subject(s)
Diphtheria Toxin/pharmacology , Germ Cells/metabolism , Heparin-binding EGF-like Growth Factor/physiology , Poisons/pharmacology , Sertoli Cells/metabolism , Animals , Cells, Cultured , Germ Cells/cytology , Germ Cells/drug effects , Male , Mice , Mice, Transgenic , Sertoli Cells/cytology , Sertoli Cells/drug effects , Spermatogenesis
11.
Domest Anim Endocrinol ; 58: 90-96, 2017 01.
Article in English | MEDLINE | ID: mdl-27769009

ABSTRACT

In feedlot steers, estradiol-17ß (E2) and combined E2 and trenbolone acetate (a testosterone analog) implants enhance rate and efficiency of muscle growth; and, consequently, these compounds are widely used as growth promoters in several countries. Treatment with E2 stimulates protein synthesis rate and suppresses protein degradation rate in fused bovine satellite cell (BSC) cultures; however, the mechanisms involved in these effects are not known with certainty. Although the genomic effects of E2 mediated through the classical estrogen receptors have been characterized, recent studies indicate that binding of E2 to the G protein-coupled estrogen receptor (GPER)-1 mediates nongenomic effects of E2 on cellular function. Our current data show that inhibition of GPER-1, matrix metalloproteinases 2 and 9 (MMP2/9), or heparin binding epidermal growth factor-like growth factor (hbEGF) suppresses E2 stimulate protein synthesis rate in cultured BSCs (P < 0.001) suggesting that all of these are required in order for E2 to stimulate protein synthesis in these cultures. In contrast, inhibition of GPER-1, MMP2/9, or hbEGF has no effect on the ability of E2 to suppress protein degradation rates in fused BSC cultures indicating that these factors are not required in order for E2 to suppress protein degradation rate in these cells. Furthermore, treatment of fused BSC cultures with E2 increased (P < 0.05) pAKT levels indicating that the pAKT pathway may play a role in E2-stimulated effects on cultured BSC. In summary, our current data show that active GPER-1, MMP2/9, and hbEGF are necessary for E2-stimulated protein synthesis but not for E2-simulated suppression of protein degradation in cultured BSC. In addition, E2 treatment increases pAKT levels in cultured BSC.


Subject(s)
Cattle , Estradiol/pharmacology , Estrogen Receptor alpha/physiology , Proteins/metabolism , Receptors, G-Protein-Coupled/physiology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Fusion , Cells, Cultured , Estrogen Receptor alpha/antagonists & inhibitors , GTP-Binding Proteins/physiology , Heparin-binding EGF-like Growth Factor/physiology , Male , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/physiology , Matrix Metalloproteinase Inhibitors , Receptors, Estrogen , Receptors, G-Protein-Coupled/antagonists & inhibitors , Satellite Cells, Skeletal Muscle/drug effects
12.
Oncogene ; 36(21): 2946-2956, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28092674

ABSTRACT

Aberrant epidermal growth factor (EGF) receptor (EGFR) signaling contributes to neoplastic initiation and progression in lung. Mutated EGFR has become as an important therapeutic target in lung cancer, whereas targeted treatment is not available for wild-type EGFR or its ligands. In this study, we found that heparin-binding (HB)-EGF, a member of the EGF family, was highly expressed in a subset of lung cancer, proliferation of which was dependent on HB-EGF signaling. Silencing of HB-EGF with RNA interference inhibited cell cycle progression in lung cancer cells. We observed that, upon HB-EGF induction, CITED4 was induced through a signal transducer and activator of transcription 3 (STAT3)-dependent pathway, regulating cell proliferation. CITED4 interacted with MYC and potentiated MYC-mediated transactivation of the CCND1 promoter, leading to cell cycle progression. Correlation analysis revealed that HB-EGF and CITED4 were significantly positively associated in primary lung tumors, and expression of HB-EGF predicted a poor survival outcome in patients. In vitro and in vivo experiments revealed that pharmacological inhibition of HB-EGF with CRM197 significantly attenuated tumor cell growth. Thus, CITED4 functions as a molecular switch in HB-EGF-induced growth control, and HB-EGF provides a novel therapeutic target for lung cancer intervention.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Heparin-binding EGF-like Growth Factor/physiology , Lung Neoplasms/genetics , Transcription Factors/physiology , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Heparin-binding EGF-like Growth Factor/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Xenograft Model Antitumor Assays
13.
Oncotarget ; 7(52): 86608-86620, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27888810

ABSTRACT

In ovarian cancer, a high ratio of anti-inflammatory M2 to pro-inflammatory M1 macrophages correlates with poor patient prognosis. The mechanisms driving poor tumor outcome as a result of the presence of M2 macrophages in the tumor microenvironment remain unclear and are challenging to study with current techniques. Therefore, in this study we utilized a micro-culture device previously developed by our lab to model concentrated paracrine signaling in order to address our hypothesis that interactions between M2 macrophages and ovarian cancer cells induce tumor cell proliferation. Using the micro-culture device, we determined that co-culture with M2-differentiated primary macrophages or THP-1 increased OVCA433 proliferation by 10-12%. This effect was eliminated with epidermal growth factor receptor (EGFR) or heparin-bound epidermal growth factor (HB-EGF) neutralizing antibodies and HBEGF expression in peripheral blood mononuclear cells from ovarian cancer patients was 9-fold higher than healthy individuals, suggesting a role for HB-EGF in tumor progression. However, addition of HB-EGF at levels secreted by macrophages or macrophage-conditioned media did not induce proliferation to the same extent, indicating a role for other factors in this process. Matrix metalloproteinase-9, MMP-9, which cleaves membrane-bound HB-EGF, was elevated in co-culture and its inhibition decreased proliferation. Utilizing inhibitors and siRNA against MMP9 in each population, we determined that macrophage-secreted MMP-9 released HB-EGF from macrophages, which increased MMP9 in OVCA433, resulting in a positive feedback loop to drive HB-EGF release and increase proliferation in co-culture. Identification of multi-cellular interactions such as this may provide insight into how to most effectively control ovarian cancer progression.


Subject(s)
Heparin-binding EGF-like Growth Factor/physiology , Macrophages/physiology , Matrix Metalloproteinase 9/physiology , Ovarian Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , ErbB Receptors/physiology , Feedback, Physiological , Female , Humans
14.
Oncogene ; 34(50): 6040-54, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-25798835

ABSTRACT

Mechanisms underlying ovarian cancer initiation and progression are unclear. Herein, we report that the Yes-associated protein (YAP), a major effector of the Hippo tumor suppressor pathway, interacts with ERBB signaling pathways to regulate the initiation and progression of ovarian cancer. Immunohistochemistry studies indicate that YAP expression is associated with poor clinical outcomes in patients. Overexpression or constitutive activation of YAP leads to transformation and tumorigenesis in human ovarian surface epithelial cells, and promotes growth of cancer cells in vivo and in vitro. YAP induces the expression of epidermal growth factor (EGF) receptors (EGFR, ERBB3) and production of EGF-like ligands (HBEGF, NRG1 and NRG2). HBEGF or NRG1, in turn, activates YAP and stimulates cancer cell growth. Knockdown of ERBB3 or HBEGF eliminates YAP effects on cell growth and transformation, whereas knockdown of YAP abrogates NRG1- and HBEGF-stimulated cell proliferation. Collectively, our study demonstrates the existence of HBEGF & NRGs/ERBBs/YAP/HBEGF & NRGs autocrine loop that controls ovarian cell tumorigenesis and cancer progression.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Ovarian Neoplasms/etiology , Phosphoproteins/physiology , Receptor, ErbB-3/physiology , Signal Transduction , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , ErbB Receptors/physiology , Female , Heparin-binding EGF-like Growth Factor/physiology , Humans , Mice , Prognosis , Transcription Factors , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL