Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Sci Rep ; 12(1): 17661, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36271272

ABSTRACT

Fasciola hepatica, one of the agents that causes fasciolosis, modulates the host immune system to allow parasite survival in the host. F. hepatica expresses carbohydrate-containing glycoconjugates that are decoded by C-type lectin receptors, such as Dectin-1, mannose receptor, DC-SIGN and MGL, that are mainly present on myeloid antigen presenting cells (APCs) and can mediate immunoregulatory properties on T cells. In particular, Macrophage Gal/GalNAc lectin 2 (MGL2) expands modified Th2 immune responses, while suppressing Th1 polarization, upon recognition of GalNAc-glycosylated parasite components. In this study, by using MGL2-DTR transgenic mice that encode human diphtheria toxin receptor in MGL2+ cells, we demonstrate the role of peritoneal APCs during F. hepatica infection in favoring parasite survival. This process might be mediated by the induction of splenic Tregs in vivo, since the depletion of MGL2+ cells conferred mice with partial resistance to the infection and abrogated the increase of CD4+/CD25+ FoxP3+ Tregs induced by the parasite. Therefore, MGL2+ cells are critical determinants of F. hepatica infection and could constitute immune checkpoints to control parasite infection.


Subject(s)
Fasciola hepatica , Fascioliasis , Humans , Mice , Animals , Heparin-binding EGF-like Growth Factor , T-Lymphocytes, Regulatory , Fascioliasis/parasitology , Lectins, C-Type/genetics , Antigen-Presenting Cells , Glycoconjugates , Macrophages , Forkhead Transcription Factors
2.
Cells ; 8(7)2019 07 17.
Article in English | MEDLINE | ID: mdl-31319505

ABSTRACT

Background: The odontogenic keratocyst (OKC) is an odontogenic cyst that shows aggressive and intriguing biological behavior. It is suggested that a hypoxic environment occurs in OKC, which led us to investigate the immunoexpression and location of hypoxia-inducible factor 1-alpha (HIF-1α) and other hypoxia-related proteins. Methods: Twenty cases of OKC were evaluated for the expression of Notch homolog 1 (NOTCH1), HIF-1α, disintegrin and metalloproteinase domain-containing protein 12 (ADAM-12), and heparin-binding epidermal growth factor-like growth factor (HBEGF) by immunohistochemistry and compared to eight control cases of calcifying odontogenic cystic (COC), orthokeratinized odontogenic cyst (OOC), and normal oral mucosa (OM) in basal and parabasal layers. Results: In OKC, all the proteins tested were expressed significantly higher in both basal (except for NOTCH1 and HBEGF in OOC) and suprabasal epithelial layers compared to controls. Looking at the epithelial layers within OKC, we observed an increased NOTCH1 and HIF-1α expression in parabasal layers. Conclusions: These results suggest that hypoxia occurs more intensively in OKC compared to COC, OM, and OOC. Hypoxia appeared to be stronger in parabasal layers as observed by higher HIF-1α expression in upper cells. Overexpression of NOTCH1, ADAM-12, and HBEGF in OKC was observed, which suggests that microenvironmental hypoxia could potentially regulate the expression of hypoxia-related proteins, and consequently, its clinical and biological behavior.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Odontogenic Cysts/metabolism , Receptor, Notch1/metabolism , Signal Transduction , ADAM12 Protein/genetics , ADAM12 Protein/metabolism , Cell Hypoxia , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Odontogenic Cysts/pathology , Oxygen/metabolism , Receptor, Notch1/genetics , Up-Regulation
3.
J Cell Physiol ; 233(3): 2247-2256, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28703301

ABSTRACT

The xenoestrogens bisphenol-A (BPA) and nonylphenol (NP) are endocrine disruptors used in the plastic polymer industry to manufacture different products for human use. Previous studies have suggested a role of these compounds in the shedding of signaling molecules, such as tumor necrosis factor α (TNF-α). The aim of this work was to evaluate the effect of BPA and NP on the sheddase ADAM17 and its newly discovered regulators iRhom1 and iRhom2 in the release of EGFR-ligands. We report that BPA and NP can stimulate the release of the ADAM17-substrates HB-EGF and TGF-α. In cells lacking ADAM17 (Adam17-/- mEFs) BPA-stimulated release of HB-EGF, but not TGF-α, was strongly reduced, whereas NP-stimulated shedding of HB-EGF and TGF-α was completely abolished. Inactivation of both ADAM17 and the related ADAM10 (Adam10/17-/- mEFs) completely prevented the release of these substrates. In the absence of iRhom1, BPA- or NP-stimulated release of HB-EGF or TGF-α was comparable to wild-type control mEFs, conversely the BPA-induced release of HB-EGF was abolished in iRhom2-/- mEFs. The defect in shedding of HB-EGF in iRhom2-/- mEF cells could be rescued by overexpressing iRhom2. Interestingly, the NP-stimulated release of HB-EGF was not affected by the absence of iRhom2, suggesting that NP could potentially activate both ADAM10 and ADAM17. We tested this hypothesis using betacellulin (BTC), an EGFR-ligand that is a substrate for ADAM10. We found that NP, but not BPA stimulated the release of BTC in Adam17-/- , iRhom2-/- , or iRhom1/2-/- , but not in Adam10/17-/- cells. Taken together, our results suggest that BPA and NP stimulate the release of EGFR-ligands by differentially activating ADAM17 or ADAM10. The identification of specific effects of these endocrine disruptors on ADAM10 and ADAM17 will help to provide a better understanding of their roles in cell signaling and proinflammatory processes, and provide new potential targets for treatment of reproductive or inflammatory diseases such as asthma or breast cancer that are promoted by xenoestrogens.


Subject(s)
ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Benzhydryl Compounds/pharmacology , Endocrine Disruptors/pharmacology , ErbB Receptors/metabolism , Estrogens/pharmacology , Fibroblasts/drug effects , Membrane Proteins/metabolism , Phenols/pharmacology , ADAM10 Protein/genetics , ADAM17 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Fibroblasts/enzymology , Heparin-binding EGF-like Growth Factor/metabolism , Ligands , Membrane Proteins/genetics , Mice, Knockout , Transfection , Tumor Necrosis Factor-alpha/metabolism
4.
JBRA Assist Reprod ; 21(3): 196-202, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28837027

ABSTRACT

OBJECTIVE: Alterations in endometrial receptivity may be involved in the etiopathogenesis of endometriosis-related infertility. The literature has suggested that patients with endometriosis present progestin resistance, which could affect embryo implantation. We question the presence of alterations in the expression of the progesterone receptor gene (PGR) and the genes related to endometrium-embryo interaction regulated by progesterone. This pilot study compared the expression of PGR, HBEGF, ITGAV, ITGB3, and SPP1 genes in eutopic endometrium during the implantation window (IW) in infertile women with endometriosis with that observed in the endometrium of fertile and infertile controls. METHODS: In this prospective case-control study, endometrial biopsies were performed during the IW in patients aged between 18 and 45 years old, with regular cycles and without endocrine/systemic dysfunctions, divided into endometriosis (END), infertile control (IC) and fertile control (FC) groups. Total RNA extraction, cDNA synthesis, and gene expression analysis by Real-Time PCR were performed. We assessed the size of the difference that our series was powered to detect. RESULTS: From the 687 patients who underwent diagnostic videolaparoscopy or tubal ligation at the University Hospital, 130 were eligible. Of these, 32 had endometrial samples collected, with 17 confirmed in the IW. Fifteen samples (5 END, 5 IC and 5 FC) were analyzed. There was no significant difference in the expression of any studied gene. Our sample size allowed us to identify or discard large differences (two standard deviations) among the groups. CONCLUSION: Endometriosis doesn't cause large changes in the endometrial expression of PGR, HBEGF, ITGAV, ITGB3 and SPP1 during the IW.


Subject(s)
Embryo Implantation , Endometriosis/epidemiology , Endometrium/metabolism , Infertility, Female/epidemiology , Adult , Endometriosis/metabolism , Endometriosis/therapy , Female , Heparin-binding EGF-like Growth Factor/analysis , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Infertility, Female/metabolism , Infertility, Female/therapy , Integrin beta3/analysis , Integrin beta3/genetics , Integrin beta3/metabolism , Osteopontin/analysis , Osteopontin/genetics , Osteopontin/metabolism , Pilot Projects , Real-Time Polymerase Chain Reaction , Receptors, Progesterone/analysis , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
5.
J Crit Care ; 42: 162-167, 2017 12.
Article in English | MEDLINE | ID: mdl-28746898

ABSTRACT

PURPOSE: Endothelial barrier dysfunction is a hallmark of sepsis, and is at least partially mediated by pathways that regulate endothelial barrier assembly during angiogenesis. Not surprisingly, increased levels of key angiogenic proteins such as VEGF-A and Angiopoietin-2 have been described in sepsis. The purpose of this study was to investigate if additional pathways that regulate endothelial barrier integrity during angiogenesis could also be involved in the host response of sepsis. MATERIAL AND METHODS: We evaluated circulating levels of four proteins involved in angiogenesis, not previously studied in sepsis, in a cohort of 50 patients with severe sepsis and septic shock. RESULTS: Circulating levels of BMP-9 and FGF-2 were similar in patients and healthy volunteers. In contrast, patients with septic shock presented 1.5-fold higher levels of endoglin (P=0.004), and 2-fold lower levels of Heparin-Binding EGF-like growth factor (HB-EGF) (P=0.002) when compared to healthy individuals. Of note, HB-EGF deficiency has been recently demonstrated to be detrimental to survival in a murine model of sepsis. CONCLUSIONS: Endoglin and HB-EGF could be involved in the host response of sepsis. Additional studies are warrant to investigate their role as biomarker or therapeutic targets in sepsis.


Subject(s)
Endoglin/blood , Fibroblast Growth Factor 2/blood , Growth Differentiation Factors/blood , Heparin-binding EGF-like Growth Factor/blood , Sepsis/blood , Shock, Septic/blood , Adult , Aged , Animals , Female , Growth Differentiation Factor 2 , Humans , Male , Middle Aged , Sepsis/physiopathology , Shock, Septic/physiopathology , Signal Transduction
6.
Genet Mol Res ; 15(3)2016 Aug 26.
Article in English | MEDLINE | ID: mdl-27706598

ABSTRACT

Development of the eyelid requires coordination of the cellular processes involved in proliferation, cell size alteration, migration, and cell death. C57BL/6J-corneal opacity (B6-Co) mice are mutant mice generated by the administration of N-ethyl-N-nitrosourea (100 mg/kg). They exhibit the eyelids open at birth phenotype, abnormal round cell shape from tightened F-actin bundles in leading edge keratinocytes at E16.5, and gradual corneal opacity with neovessels. The tip of the leading edge in B6-Co mice did not move forward, and demonstrated a sharp peak shape without obvious directionality. Analysis of the biological characteristics of B6-Co mice demonstrated that abnormal migration of keratinocytes could affect eyelid development, but proliferation and apoptosis in B6-Co mice had no effect. Mutant gene mapping and sequence analysis demonstrated that in B6-Co mice, adenosine was inserted into the untranslated regions, between 3030 and 3031, in the mRNA 3'-terminal of Fgf10. In addition, guanine 7112 was substituted by adenine in the Mtap1B mRNA, and an A2333T mutation was identified in Mtap1B. Quantitative real-time polymerase chain reaction analysis showed that expression of the Hbegf gene was significantly down-regulated in the eyelids of B6- Co mice at E16.5, compared to B6 mice. However, the expression of Rock1, Map3k1, and Jnk1 genes did not show any significant changes. Abnormal keratinocyte migration and down-regulated expression of the Hbegf gene might be associated with impaired eyelid development in B6-Co mice.


Subject(s)
Cornea/metabolism , Corneal Neovascularization/genetics , Corneal Opacity/genetics , Eyelids/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Keratinocytes/metabolism , 3' Untranslated Regions , Actins/genetics , Actins/metabolism , Animals , Cell Movement , Cell Polarity , Cell Proliferation , Cell Shape , Cornea/abnormalities , Cornea/growth & development , Corneal Neovascularization/chemically induced , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Corneal Opacity/chemically induced , Corneal Opacity/metabolism , Corneal Opacity/pathology , Embryo, Mammalian , Ethylnitrosourea , Eyelids/abnormalities , Eyelids/growth & development , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation, Developmental , Heparin-binding EGF-like Growth Factor/metabolism , Keratinocytes/pathology , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutagens , Phenotype , Primary Cell Culture
7.
Biochem Biophys Res Commun ; 478(1): 39-45, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27462018

ABSTRACT

The epidermal growth factor receptor (EGFR) is activated through binding to specific ligands and generates signals for proliferation, differentiation, migration, and cell survival. Recent data show the role of nuclear EGFR in tumors. Although many EGFR ligands are upregulated in cancers, little is known about their effects on EGFR nuclear translocation. We have compared the effects of six EGFR ligands (EGF, HB-EGF, TGF-α, ß-Cellulin, amphiregulin, and epiregulin) on nuclear translocation of EGFR, receptor phosphorylation, migration, and proliferation. Cell fractionation and confocal immunofluorescence detected EGFR in the nucleus after EGF, HB-EGF, TGF-α and ß-Cellulin stimulation in a dose-dependent manner. In contrast, amphiregulin and epiregulin did not generate nuclear translocation of EGFR. EGF, HB-EGF, TGF-α and ß-Cellulin showed correlations between a higher rate of wound closure and increased phosphorylation of residues in the carboxy-terminus of EGFR, compared to amphiregulin and epiregulin. The data indicate that EGFR is translocated to the nucleus after stimulation with EGF, HB-EGF, TGF-α and ß-Cellulin, and that these ligands are related to increased phosphorylation of EGFR tyrosine residues, inducing migration of SkHep-1 cells.


Subject(s)
Cell Nucleus/metabolism , ErbB Receptors/metabolism , Active Transport, Cell Nucleus , Betacellulin/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epidermal Growth Factor/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Neoplasms/metabolism , Phosphorylation , Transforming Growth Factor alpha/metabolism
8.
Histopathology ; 69(1): 99-106, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26707922

ABSTRACT

AIMS: Ameloblastoma AME is a benign tumour characterized by local invasiveness, high recurrence rates, and diverse histological patterns. The oxygen concentration is reduced in specific areas of the tumour microenvironment, which leads to intratumoral hypoxia. Crosstalk between NOTCH1, a disintegrin and metalloproteinase 12 (ADAM-12), hypoxia-inducible factor 1α (HIF-1α) and heparin-binding epidermal growth factor (HB-EGF) under hypoxic conditions has been implicated in invadopodia formation, tumour invasiveness, and metastasis development. The aim of this study was to analyse the expression of these proteins, in order to further elucidate the mechanisms underlying AME invasiveness. METHODS AND RESULTS: Twenty cases of AME, eight calcifying cystic odontogenic tumours CCOTs and 10 samples of dental follicle were used to investigate the expression of these proteins by immunohistochemistry with the primary antibodies anti-NOTCH1, anti-ADAM-12, anti-HIF-1α, and anti-HB-EGF. Immunostaining results were expressed as the percentage of stained area in images acquired in an AxioScope microscope equipped with an AxioCamHRc camera and a × 40 objective. The results showed that immunoexpression of all proteins was higher in the AME samples than in the CCOT and dental follicle samples (P < 0.05). CONCLUSIONS: AME showed an increased presence of proteins associated with tumour invasiveness, which indicates a possible role of these proteins in the biological behaviour of this tumour.


Subject(s)
ADAM12 Protein/metabolism , Ameloblastoma/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mouth Neoplasms/metabolism , Odontogenic Cyst, Calcifying/metabolism , Receptor, Notch1/metabolism , Ameloblastoma/diagnosis , Cohort Studies , Dental Sac/metabolism , Dental Sac/pathology , Female , Humans , Immunohistochemistry , Male , Mouth Neoplasms/diagnosis , Neoplasm Invasiveness , Odontogenic Cyst, Calcifying/diagnosis , Tissue Array Analysis , Tumor Hypoxia , Tumor Microenvironment
9.
Neuroscience ; 279: 65-76, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25171790

ABSTRACT

In genetically-modified Lmx1b(f/f/p) mice, selective deletion of LMX1B in Pet-1 expressing cells leads to failure of embryonic development of serotonin (5-HT) neurons. As adults, these mice have a decreased hypercapnic ventilatory response and abnormal thermoregulation. This mouse model has been valuable in defining the normal role of 5-HT neurons, but it is possible that developmental compensation reduces the severity of observed deficits. Here we studied mice genetically modified to express diphtheria toxin receptors (DTR) on Pet-1 expressing neurons (Pet-1-Cre/floxed DTR or Pet1/DTR mice). These mice developed with a normal complement of 5-HT neurons. As adults, systemic treatment with 2-35µg of diphtheria toxin (DT) reduced the number of tryptophan hydroxylase-immunoreactive (TpOH-ir) neurons in the raphe nuclei and ventrolateral medulla by 80%. There were no effects of DT on minute ventilation (VE) or the ventilatory response to hypercapnia or hypoxia. At an ambient temperature (TA) of 24°C, all Pet1/DTR mice dropped their body temperature (TB) below 35°C after DT treatment, but the latency was shorter in males than females (3.0±0.37 vs. 4.57±0.29days, respectively; p<0.001). One week after DT treatment, mice were challenged by dropping TA from 37°C to 24°C, which caused TB to decrease more in males than in females (29.7±0.31°C vs. 33.0±1.3°C, p<0.01). We conclude that the 20% of 5-HT neurons that remain after DT treatment in Pet1/DTR mice are sufficient to maintain normal baseline breathing and a normal response to CO2, while those affected include some essential for thermoregulation, in males more than females. In comparison to models with deficient embryonic development of 5-HT neurons, acute deletion of 5-HT neurons in adults leads to a greater defect in thermoregulation, suggesting that significant developmental compensation can occur.


Subject(s)
Body Temperature Regulation/physiology , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/metabolism , Neurons/physiology , Respiration , Serotonin/metabolism , Animals , Female , Heparin-binding EGF-like Growth Factor/genetics , Hypercapnia/physiopathology , Hypoxia/physiopathology , Male , Medulla Oblongata/physiology , Mice, Transgenic , Raphe Nuclei/physiology , Sex Characteristics , Tryptophan Hydroxylase/metabolism
10.
J Biol Chem ; 287(51): 43071-82, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23105116

ABSTRACT

ADAM17, which is also known as TNFα-converting enzyme, is the major sheddase for the EGF receptor ligands and is considered to be one of the main proteases responsible for the ectodomain shedding of surface proteins. How a membrane-anchored proteinase with an extracellular catalytic domain can be activated by inside-out regulation is not completely understood. We characterized thioredoxin-1 (Trx-1) as a partner of the ADAM17 cytoplasmic domain that could be involved in the regulation of ADAM17 activity. We induced the overexpression of the ADAM17 cytoplasmic domain in HEK293 cells, and ligands able to bind this domain were identified by MS after protein immunoprecipitation. Trx-1 was also validated as a ligand of the ADAM17 cytoplasmic domain and full-length ADAM17 recombinant proteins by immunoblotting, immunolocalization, and solid phase binding assay. In addition, using nuclear magnetic resonance, it was shown in vitro that the titration of the ADAM17 cytoplasmic domain promotes changes in the conformation of Trx-1. The MS analysis of the cross-linked complexes showed cross-linking between the two proteins by lysine residues. To further evaluate the functional role of Trx-1, we used a heparin-binding EGF shedding cell model and observed that the overexpression of Trx-1 in HEK293 cells could decrease the activity of ADAM17, activated by either phorbol 12-myristate 13-acetate or EGF. This study identifies Trx-1 as a novel interaction partner of the ADAM17 cytoplasmic domain and suggests that Trx-1 is a potential candidate that could be involved in ADAM17 activity regulation.


Subject(s)
ADAM Proteins/metabolism , Thioredoxins/metabolism , ADAM Proteins/chemistry , ADAM17 Protein , Amino Acid Sequence , Animals , Chromatography, Liquid , Cross-Linking Reagents/pharmacology , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Heparin-binding EGF-like Growth Factor , Humans , Immunoblotting , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Models, Molecular , Molecular Sequence Data , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Recombinant Proteins/metabolism , Reproducibility of Results , Tetradecanoylphorbol Acetate/pharmacology , Thioredoxins/chemistry
11.
J Pediatr ; 156(2 Suppl): S31-5, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20105663

ABSTRACT

Maternal milk is a complex fluid, with multifunctional roles within the developing gastrointestinal tract. Epidermal growth factor (EGF) and heparin-binding EGF-like growth factor (HB-EGF) are members of the family of EGF-related peptides. Biological actions of these growth factors are mediated via interaction with the EGF-receptor (EGF-R). In the early postnatal period, breast milk is the major source of EGF for the developing intestinal mucosa. HB-EGF is also detected in breast milk, but in concentrations 2 to 3 times lower than EGF. With normal physiological conditions, the intestinal epithelium undergoes a continuing process of cell proliferation, differentiation, and maturation. EGF plays an important role in these processes. In pathophysiologic situations, EGF contributes to epithelial protection from injury and post-injury mucosal repair. Necrotizing enterocolitis (NEC) is a devastating disease affecting infants born prematurely. The pathogenesis of NEC is not known, and there is no effective treatment for this disease. In an experimental NEC model, oral administration of a physiological dose of EGF significantly reduces the incidence and severity of NEC. HB-EGF provides similar protection against NEC, but only when pharmacological doses are used. Further studies are necessary before EGF can be introduced as an efficient therapeutic approach of intestinal injury.


Subject(s)
Epidermal Growth Factor/physiology , Gastrointestinal Tract/growth & development , Intercellular Signaling Peptides and Proteins/physiology , Milk, Human/chemistry , Animals , Enterocolitis, Necrotizing/physiopathology , Enterocolitis, Necrotizing/therapy , Epidermal Growth Factor/therapeutic use , Female , Heparin-binding EGF-like Growth Factor , Humans , Infant , Infant, Newborn , Intestinal Mucosa/physiology
12.
J Periodontal Res ; 44(1): 73-80, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19515020

ABSTRACT

BACKGROUND AND OBJECTIVES: Membrane type 1-matrix metalloproteinase (MT1-MMP) is a collagenolytic enzyme involved in connective tissue remodeling. In periodontal tissues, either cytokines or growth factors regulate the production of proteolytic enzymes. Mice deficient in epidermal growth factor receptor (EGFR) show a reduced expression of MT1-MMP, suggesting that this receptor may play an important role in MT1-MMP production. The present study evaluated the role of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) and EGFR in the production of MT1-MMP in gingival fibroblasts. MATERIAL AND METHODS: Primary cultures of human gingival fibroblasts were cultured over plastic or a type I collagen matrix and stimulated with TNF-alpha and EGF. A selective EGFR inhibitor (AG1478) was used to interfere with this signaling pathway. Production of MT1-MMP and activation of proMMP-2 were studied using Western blot and gelatin zymography, respectively. Activation of EGFR signaling was assessed through immunoprecipitation and Western blot. Expression of EGFR ligands was determined through reverse transcriptase-polymerase chain reaction. RESULTS: Treatment of gingival fibroblasts cultured over a collagen matrix with TNF-alpha stimulated proMMP-2 activation and MT1-MMP production. However, after using AG1478, both responses were inhibited. Tumor necrosis factor-alpha induced EGFR transactivation and stimulated the expression of the mRNA for the EGFR ligands heparin binding-epidermal growth factor (HB-EGF) and transforming growth factor-alpha (TGF-alpha). CONCLUSIONS: The present study shows that TNF-alpha may stimulate MT1-MMP production through transactivation of EGFR. Tumor necrosis factor-alpha may also modulate the expression of the EGFR ligands TGF-alpha and HB-EGF. Production of MT1-MMP by TNF-alpha requires interaction with EGFR, suggesting that tissue remodeling is controlled by cross-communication between diverse signaling pathways in gingival fibroblasts.


Subject(s)
ErbB Receptors/drug effects , Fibroblasts/drug effects , Gingiva/drug effects , Matrix Metalloproteinase 14/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Collagen Type I/pharmacology , Culture Media , Enzyme Inhibitors/pharmacology , Enzyme Precursors/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Fibroblasts/enzymology , Gelatinases/drug effects , Gingiva/cytology , Gingiva/enzymology , Heparin/analysis , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/analysis , Ligands , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Quinazolines , Receptors, Cell Surface/analysis , Transcriptional Activation/drug effects , Transforming Growth Factor alpha/drug effects , Tyrphostins/pharmacology
13.
Oncol Rep ; 21(6): 1397-404, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19424616

ABSTRACT

The effects of estradiol (E2) and of an AFP-derived cyclized peptide (cP) on the proliferation of primary cultures of cancer cells isolated from spontaneous canine mammary tumors were studied. The cellular response to E2 and cP was related to the expression of estradiol receptor (isoforms alpha and beta). In ER-positive cells, 2 nM estradiol increased cell proliferation and the phosphorylation of ERK1/2; 2 microg/ml cP inhibited all these effects. Estradiol also increased HER2 immunoreactivity in ER-positive cells, an effect that was reverted to its basal values by cP. Estradiol stimulated in these cells the release of MMP2 and MMP9 and the shedding of HB-EGF, effects that the cP did not affect. ER-negative cells were refractory to estradiol or cP. All canine mammary tumor cells in culture responded to treatments analogously to human mammary cancer cells. Our results support the proposal of cP as a new, potentially effective therapeutic agent for the management of mammary cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Mammary Neoplasms, Animal/pathology , Peptides, Cyclic/pharmacology , alpha-Fetoproteins/pharmacology , Animals , Apoptosis/drug effects , Dogs , Epidermal Growth Factor/metabolism , Estradiol/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Female , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/metabolism , Mammary Neoplasms, Animal/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Staging , Phosphorylation , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Tamoxifen/pharmacology , Tumor Cells, Cultured
14.
Eur J Pharmacol ; 578(1): 1-10, 2008 Jan 06.
Article in English | MEDLINE | ID: mdl-17915215

ABSTRACT

The effect of insulin-like growth factor-I (IGF-I) on human alpha(1B)-adrenoceptor function, phosphorylation state and cellular location was studied. Rat-1 fibroblasts were transfected with a plasmid construction containing enhanced green fluorescent protein joined to the carboxyl terminus of the human alpha(1B)-adrenoceptor. Receptors were identified by radioligand binding and photoaffinity labeling, and were immunoprecipitated with an antiserum generated against the enhanced green fluorescent protein. The receptor was functional, as evidenced by noradrenaline action on intracellular calcium and inositol phosphate production. IGF-I had no significant effect by itself on these parameters but markedly reduced the effects of noradrenaline. IGF-I induced alpha(1B)-adrenoceptor phosphorylation, which was markedly reduced by the following agents: pertussis toxin, a metalloproteinase inhibitor, diphtheria toxin mutant CRM 197, an epidermal growth factor (EGF) receptor intrinsic kinase activity inhibitor, and by phosphoinositide 3-kinase and protein kinase C inhibitors. IGF-I action appears to involve activation of a pertussis toxin-sensitive G protein, shedding of heparin-binding EGF and autocrine activation of EGF receptors. G protein subunits and phosphotyrosine residues stimulate phosphoinositide 3-kinase activity leading to activation of protein kinase C, which in turn phosphorylates alpha(1B)-adrenoceptors. Confocal fluorescent microscopy showed that alpha(1B)-adrenoceptors fussed to the green fluorescent protein were located in plasma membrane and intracellular vesicles in the basal state. IGF-I induced receptor redistribution favoring the intracellular location; this effect was blocked by hypertonic sucrose and concanavalin A. Our data show that IGF-I induces alpha(1B)-adrenoceptor desensitization associated to receptor phosphorylation and internalization.


Subject(s)
GTP-Binding Proteins/metabolism , Insulin-Like Growth Factor I/physiology , Receptors, Adrenergic, alpha-1/metabolism , Animals , Cell Line , ErbB Receptors/metabolism , Fibroblasts/metabolism , Green Fluorescent Proteins , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Microscopy, Confocal , Pertussis Toxin/pharmacology , Phosphorylation , Photoaffinity Labels , Radioligand Assay , Rats
15.
J Cell Biochem ; 103(3): 986-93, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17654528

ABSTRACT

In this study, we demonstrated that tyrosine phosphorylation of EGFR and the autocrine expression of uPA and HB-EGF depend on the activity of c-jun amino-terminal kinase (JNK) in human prostatic DU-145 cells. These cells overexpress EGFR and produce a high amount of uPA. Treatment with either SP600125, a specific chemical inhibitor of JNK, or the expression of a dominant-negative JNK form inhibited autocrine production of uPA and HB-EGF, which block EGFR phosphorylation and mitigates invasive capacity. Our data provided evidence that in DU-145 cells, the maintenance of the activation level of EGFR, which determines the cellular invasive potential, operates through an autocrine loop involving the JNK-dependent production of uPA and HB-EGF activity. Moreover, we found that exogenously added uPA stimulates autocrine production of HB-EGF, and that blocking HB-EGF activity curbed DU-145 cell invasive potential.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/biosynthesis , Intercellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Urokinase-Type Plasminogen Activator/biosynthesis , Anthracenes/pharmacology , Autocrine Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Enzyme Activation , Epidermal Growth Factor/physiology , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Neoplasm Invasiveness , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Up-Regulation , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/pharmacology
16.
Braz J Med Biol Res ; 39(11): 1373-85, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17146550

ABSTRACT

The successful implantation of the blastocyst depends on adequate interactions between the embryo and the uterus. The development of the embryo begins with the fertilized ovum, a single totipotent cell which undergoes mitosis and gives rise to a multicellular structure named blastocyst. At the same time, increasing concentrations of ovarian steroid hormones initiate a complex signaling cascade that stimulates the differentiation of endometrial stromal cells to decidual cells, preparing the uterus to lodge the embryo. Studies in humans and in other mammals have shown that cytokines and growth factors are produced by the pre-implantation embryo and cells of the reproductive tract; however, the interactions between these factors that converge for successful implantation are not well understood. This review focuses on the actions of interleukin-1, leukemia inhibitory factor, epidermal growth factor, heparin-binding epidermal growth factor, and vascular endothelial growth factor, and on the network of their interactions leading to early embryo development, peri-implantatory endometrial changes, embryo implantation and trophoblast differentiation. We also propose therapeutical approaches based on current knowledge on cytokine interactions.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Endometrium/cytology , Intercellular Signaling Peptides and Proteins/physiology , Trophoblasts/cytology , Animals , Blastocyst/cytology , Blastocyst/physiology , Embryo Implantation/physiology , Embryo Transfer , Endometrium/metabolism , Epidermal Growth Factor/biosynthesis , Female , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-1/biosynthesis , Leukemia Inhibitory Factor/biosynthesis , Mice , Pregnancy , Vascular Endothelial Growth Factor A/biosynthesis
17.
Eur J Pharmacol ; 542(1-3): 31-6, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16828079

ABSTRACT

Phosphorylation of G protein-coupled receptors is one of the earliest events that regulate their function. Current evidence indicates that homologous desensitization of these receptors mainly involves G protein-coupled receptor kinases whereas in heterologous desensitization second messenger-activated kinases play key roles. Recent data show that transactivation of EGF (epidermal growth factor) receptors may also play a role in receptor phosphorylation. The role of this process was studied for the alpha1B-adrenoceptor phosphorylation induced by agents acting through different processes using inhibitors to block the EGF receptor transactivation process at different levels. Experiments were performed using transfected rat-1 fibroblasts that express alpha1B-adrenoceptors in a stably fashion. A metalloproteinase inhibitor, an anti-heparin-binding-EGF-selective antibody, and a selective EGF-receptor kinase inhibitor blocked the alpha1B-adrenoceptor phosphorylation induced by noradrenaline or endothelin-1. Our results indicate that shedding of heparin-binding-EGF, transactivation of EGF receptors plays a more general role in alpha1B-adrenoceptor phosphorylation than previously anticipated. It is possible that other receptors/channels could be modulated through a similar pathway.


Subject(s)
ErbB Receptors/physiology , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-Agonists/pharmacology , Androstadienes/pharmacology , Animals , Antibodies/pharmacology , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Endothelin-1/pharmacology , Epidermal Growth Factor/immunology , ErbB Receptors/immunology , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Norepinephrine/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Quinazolines , Rats , Receptors, Adrenergic, alpha-1/genetics , Tetradecanoylphorbol Acetate/pharmacology , Thiophenes/pharmacology , Tyrphostins/pharmacology , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL