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










Publication year range
1.
J Immunol ; 189(10): 4748-58, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23071284

ABSTRACT

The TNF superfamily ligands APRIL and BAFF bind with different affinity to two receptors, BCMA and TACI, and induce cell survival and/or proliferation, whereas BAFF also binds specifically to BAFFR. These molecules were considered specific for the immune system. Recently, however, they were also found in epithelial and mesenchymal noncancerous and cancerous tissues and cell lines. In this article, we report that hepatocellular carcinoma (HCC) cell lines HepG2 and Hep3B and HCC specimens express APRIL and BAFF and their receptors BCMA and BAFFR, but not TACI; APRIL/BCMA is enhanced in HCC, compared with normal liver tissue. In contrast to previous reports, APRIL binding to BCMA decreases cell proliferation by inducing G(2)/M cell cycle arrest, whereas BAFF has no effect on cell growth. HCC cells therefore represent a rare system in which these two ligands (APRIL and BAFF) exert a differential effect and may serve as a model for specific APRIL/BCMA actions. We show that the effect of APRIL is mediated via BCMA, which does not activate the classical NF-κB pathway, whereas it induces a novel signaling pathway, which involves JNK2 phosphorylation, FOXO3A activation, and GADD45 transcription. In addition, JNK2 mediates the phosphorylation of Akt, which is activated but does not participate in the antiproliferative effect of APRIL. Furthermore, transcriptome analysis revealed that APRIL modifies genes specifically related to cell cycle modulation, including MCM2/4/5/6, CDC6, PCNA, and POLE2. Our data, therefore, identify a novel APRIL/BCMA signaling pathway in HCC and suggest that APRIL could have a pleiotropic role in tumor biology.


Subject(s)
B-Cell Maturation Antigen/immunology , Cell Cycle Proteins/immunology , DNA-Binding Proteins/immunology , Forkhead Transcription Factors/immunology , G2 Phase Cell Cycle Checkpoints/immunology , Liver/immunology , M Phase Cell Cycle Checkpoints/immunology , MAP Kinase Kinase 7/immunology , Nuclear Proteins/immunology , Transcription Factors/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Cell Activating Factor/metabolism , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Hep G2 Cells , Humans , Liver/cytology , M Phase Cell Cycle Checkpoints/genetics , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/genetics , Phosphorylation/immunology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/immunology , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/immunology
2.
Cell Transplant ; 21(11): 2441-54, 2012.
Article in English | MEDLINE | ID: mdl-22507764

ABSTRACT

Epidermal organization and homeostasis are regulated by mesenchymal influences through paracrine actions. Until today, dermal fibroblasts (DFs) are used in the "dermal" layer to support keratinocyte growth in vitro in dermal and skin substitutes. In the present work, we used human adipose tissue-derived mesenchymal cells (ADMCs) as a support of keratinocyte growth in vitro (in monolayer culture and in 3D skin cell culture models) and in vivo (mouse wound healing models) and compared our findings with those obtained using dermal fibroblasts. ADMCs induce reepithelialization during wound healing more efficiently than DFs, by enhancing keratinocyte proliferation through cell cycle progression, and migration. This effect is mediated (at least partially) by a paracrine action of KGF-1 and PDGF-BB, which are more prominently expressed in ADMCs than in DFs. Furthermore, replacement of DFs by ADMCs in the dermal compartment of organotypic skin cultures leads to an artificial epidermis resembling to that of normal skin, concerning the general histology, although with a higher expression of cytokeratins 5 and 19. In Rag1 knockout mice, ADMCs induced a more rapid reepithelialization and a more effective wound healing, compared to dermal fibroblasts. In conclusion, we provide evidence that ADMCs can serve as supportive cells for primary keratinocyte cultures. In addition, because of their abundance and the great cell yield achieved during ADMC isolation, they represent an interesting cell source, with potential aspects for clinical use.


Subject(s)
Adipose Tissue/cytology , Fibroblast Growth Factor 7/metabolism , Fibroblasts/cytology , Mesenchymal Stem Cells/cytology , Proto-Oncogene Proteins c-sis/metabolism , Skin/cytology , Skin/metabolism , Becaplermin , Blotting, Western , Cell Cycle/physiology , Cell Line , Cells, Cultured , Humans , Immunohistochemistry , Keratinocytes/cytology , Polymerase Chain Reaction , Wound Healing/physiology
3.
PLoS One ; 7(3): e33609, 2012.
Article in English | MEDLINE | ID: mdl-22438963

ABSTRACT

The soluble TNF-like weak inducer of apoptosis (TWEAK, TNFSF12) binds to the fibroblast growth factor-inducible 14 receptor (FN14, TNFRSF12A) on the cell membrane and induces multiple biological responses, such as proliferation, migration, differentiation, angiogenesis and apoptosis. Previous reports show that TWEAK, which does not contain a death domain in its cytoplasmic tail, induces the apoptosis of tumor cell lines through the induction of TNFα secretion. TWEAK induces apoptosis in human keratinocytes. Our experiments clearly demonstrate that TWEAK does not induce the secretion of TNFα or TRAIL proteins. The use of specific inhibitors and the absence of procaspase-3 cleavage suggest that the apoptosis of keratinocytes follows a caspase- and cathepsin B-independent pathway. Further investigation showed that TWEAK induces a decrease in the mitochondrial membrane potential of keratinocytes. Confocal microscopy showed that TWEAK induces the cleavage and the translocation of apoptosis inducing factor (AIF) from the mitochondria to the nucleus, thus initiating caspase-independent apoptosis. Moreover, TWEAK induces FOXO3 and GADD45 expression, cdc2 phosphorylation and cdc2 and cyclinB1 degradation, resulting in the arrest of cell growth at the G2/M phase. Finally, we report that TWEAK and FN14 are normally expressed in the basal layer of the physiological epidermis and are greatly enhanced in benign (psoriasis) and malignant (squamous cell carcinoma) skin pathologies that are characterized by an inflammatory component. TWEAK might play an essential role in skin homeostasis and pathology.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/physiology , G2 Phase Cell Cycle Checkpoints/physiology , Keratinocytes/cytology , Keratinocytes/metabolism , Tumor Necrosis Factors/metabolism , Active Transport, Cell Nucleus , CDC2 Protein Kinase , Caspases/metabolism , Cathepsin B/metabolism , Cell Line , Cyclin B/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases , Cytokine TWEAK , Humans , Inflammation/metabolism , Inflammation/pathology , Psoriasis/metabolism , Psoriasis/pathology , Receptors, Tumor Necrosis Factor/metabolism , Skin/cytology , Skin/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TWEAK Receptor , Tumor Necrosis Factor-alpha/biosynthesis
5.
Endocrinology ; 153(2): 739-49, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22166983

ABSTRACT

TNFα is known to be expressed in human skin, regulating immune-related responses. Here we report that human normal skin keratinocytes express the members of the TNF superfamily members A proliferation-inducing ligand (APRIL; TNFSF13), B cell-activating factor (BAFF; TNFSF13B), and their receptors, B cell maturation antigen (BCMA; TNFRSF17) and transmembrane activator, calcium-modulator, and cyclophilin ligand interactor (TACI; TNFRSF13B), in a distinct spatial pattern. Our data show a differential expression of these molecules within epidermal layers and skin appendages, whereas the BAFF-specific receptor BAFFR (TNFRSF13C) is absent. Importantly, APRIL and BCMA but not BAFF or TACI are up-regulated in inflammatory skin lesions of psoriasis and squamous cell carcinomas. To explore the functional significance of this system in the skin, we assayed these receptors and ligands in cultured primary keratinocytes and HaCaT cells. We show that both cell types express BAFF, APRIL, BCMA, and TACI. Furthermore, APRIL and/or BAFF trigger nuclear factor-κB activation and IL-6 and granulocyte macrophage colony-stimulating factor (GM-CSF) expression through functional BCMA receptors, an activation inhibited by anti-BCMA short hairpin RNA. However, BAFF and/or APRIL do not induce IL-8 or TNFα production. Our data advance BCMA as an inflammation-related TNFSFR member in keratinocytes, of potential importance in the management of inflammatory skin conditions.


Subject(s)
B-Cell Maturation Antigen/metabolism , Dermatitis/metabolism , Gene Expression Regulation/physiology , Keratinocytes/metabolism , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Dermatitis/pathology , Epidermis/metabolism , Epidermis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
6.
PLoS Biol ; 9(4): e1001051, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21541365

ABSTRACT

The neurosteroid dehydroepiandrosterone (DHEA), produced by neurons and glia, affects multiple processes in the brain, including neuronal survival and neurogenesis during development and in aging. We provide evidence that DHEA interacts with pro-survival TrkA and pro-death p75(NTR) membrane receptors of neurotrophin nerve growth factor (NGF), acting as a neurotrophic factor: (1) the anti-apoptotic effects of DHEA were reversed by siRNA against TrkA or by a specific TrkA inhibitor; (2) [(3)H]-DHEA binding assays showed that it bound to membranes isolated from HEK293 cells transfected with the cDNAs of TrkA and p75(NTR) receptors (K(D): 7.4 ± 1.75 nM and 5.6 ± 0.55 nM, respectively); (3) immobilized DHEA pulled down recombinant and naturally expressed TrkA and p75(NTR) receptors; (4) DHEA induced TrkA phosphorylation and NGF receptor-mediated signaling; Shc, Akt, and ERK1/2 kinases down-stream to TrkA receptors and TRAF6, RIP2, and RhoGDI interactors of p75(NTR) receptors; and (5) DHEA rescued from apoptosis TrkA receptor positive sensory neurons of dorsal root ganglia in NGF null embryos and compensated NGF in rescuing from apoptosis NGF receptor positive sympathetic neurons of embryonic superior cervical ganglia. Phylogenetic findings on the evolution of neurotrophins, their receptors, and CYP17, the enzyme responsible for DHEA biosynthesis, combined with our data support the hypothesis that DHEA served as a phylogenetically ancient neurotrophic factor.


Subject(s)
Apoptosis , Dehydroepiandrosterone/metabolism , Nerve Tissue Proteins/metabolism , Neurons/pathology , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurogenesis , PC12 Cells , Phosphorylation , Phylogeny , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Rats , Receptor, trkA/genetics , Receptors, Nerve Growth Factor/genetics , Signal Transduction , Transfection
7.
Anal Cell Pathol (Amst) ; 34(1-2): 49-60, 2011.
Article in English | MEDLINE | ID: mdl-21483105

ABSTRACT

In advanced renal cell carcinoma (RCC), surgery combined with systemic chemotherapy and immunotherapy have had limited effectiveness. Therapeutic modalities targeting VEGF, PDGF, and c-kit using tyrosine kinase inhibitors and m-TOR using specific biologic factors are in development. Therapeutic approaches targeting TNF-alpha have shown limited efficacy, while anti-TRAIL (TNFSF10) antibodies have shown enhanced activity. The presence and potential significance of other members of the TNFSF has not been investigated. Here, we assayed the TNFSF members APRIL, BAFF, TWEAK and their receptors (BCMA, TACI, BAFFR, Fn14) in 86 conventional type clear cell RCC, using immunohistochemistry and correlated our findings with histological data and, in a limited series, follow-up of patients. We observed a differential expression of these TNFSF ligands and receptors in cancerous and non-cancerous structures. BAFF was found in all RCC; APRIL expression is associated with an aggressive phenotype, correlating negatively with patients' disease-free survival, while TWEAK and its receptor Fn14 are heterogeneously expressed, correlating negatively with the grade and survival of RCC patients. This is the first study, presenting together the TNFSF members APRIL, BAFF, TWEAK and their receptors in different areas of normal renal tissue and RCC, suggesting a potential role of these TNFSF members in renal tumor biology.


Subject(s)
B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/metabolism , Carcinoma, Renal Cell/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Tumor Necrosis Factors/metabolism , B-Cell Activating Factor/genetics , B-Cell Activation Factor Receptor/genetics , B-Cell Maturation Antigen/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cytokine TWEAK , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Ligands , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/genetics , TWEAK Receptor , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factors/genetics
8.
J Immunol ; 183(9): 5948-56, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19828625

ABSTRACT

Adipose tissue represents a rich source of multipotent stem cells. Mesenchymal cells, isolated from this source, can differentiate to other cell types in vitro and therefore can be used for a number of regenerative therapies. Our view of adipose tissue has recently changed, establishing adipocytes as new members of the immune system, as they produce a number of proinflammatory cytokines (such as IL-6 and TNFalpha and chemokines, in addition to adipokines (leptin, adiponectin, resistin) and molecules associated with the innate immune system. In this paper, we report the differential expression of TNF-superfamily members B cell activating factor of the TNF Family (BAFF), a proliferation inducing ligand (APRIL), and TNF-like weak inducer of apoptosis (TWEAK) in immature-appearing and mature adipocytes and in benign and malignant adipose tissue-derived tumors. These ligands act through their cognitive receptors, BAFF receptor, transmembrane activator and calcium signal-modulating cyclophilic ligand (TACI), B cell maturation Ag (BCMA), and fibroblast growth factor-inducible 14 (Fn14), which are also expressed in these cells. We further report the existence of functional BCMA, TACI, and Fn14 receptors and their ligands BAFF, APRIL, and TWEAK on adipose tissue-derived mesenchymal cells, their interaction modifying the rate of adipogenesis. Our data integrate BAFF, APRIL, and TWEAK and their receptors BCMA, TACI, and Fn14 as novel potential mediators of adipogenesis, in addition to their specific role in immunity, and define immature and mature adipocytes as source of immune mediators.


Subject(s)
Adipocytes/immunology , Adipocytes/metabolism , B-Cell Activating Factor/biosynthesis , B-Cell Activation Factor Receptor/biosynthesis , B-Cell Maturation Antigen/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Transmembrane Activator and CAML Interactor Protein/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 13/biosynthesis , Tumor Necrosis Factors/biosynthesis , Adipocytes/cytology , Adipocytes/pathology , Adipose Tissue/cytology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Cell Differentiation/immunology , Cells, Cultured , Cytokine TWEAK , Humans , TWEAK Receptor , Tumor Cells, Cultured
9.
Exp Cell Res ; 315(13): 2275-83, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19379733

ABSTRACT

Although the epidermis is importantly affected by steroid hormones, little is known about the effects of dehydroepiandrosterone (DHEA) on human keratinocytes, in spite of its abundance in human serum. Here, we demonstrate for the first time a protective role of DHEA against apoptosis in keratinocytes, using non-cancerous immortalized human HaCaT cells. We show that DHEA transmits its signal via specific G protein-coupled, membrane binding sites and inhibits apoptosis, through prevention of mitochondrial disruption and altered balance of Bcl-2 proteins. DHEA conjugated to the membrane impermeable molecule BSA, as well as DHEA-S, the most abundant form of DHEA in human serum exhibit similar anti-apoptotic effect. Our data provide new insights in the treatment of the epidermis with steroid hormones in apoptosis-related conditions.


Subject(s)
Apoptosis/drug effects , Cell Membrane/metabolism , Cytoprotection , Dehydroepiandrosterone/pharmacology , Keratinocytes , Animals , Binding Sites , Cell Line , Dehydroepiandrosterone/chemistry , Estradiol/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Testosterone/metabolism
10.
Exp Cell Res ; 312(15): 2745-56, 2006 Sep 10.
Article in English | MEDLINE | ID: mdl-16822503

ABSTRACT

Sex steroids affect adrenal chromaffin cell function. In the present work, we have examined the expression and functional significance of membrane androgen receptor sites in normal rat adrenal chromaffin cells and in the PC12 rat pheochromocytoma cell line which can differentiate to either a neuronal or to an epithelial phenotype and expresses membrane estrogen receptor sites. Our data are as follows: (a) no cytosolic androgen receptors were found in both normal chromaffin and PC12 cells; (b) both types of chromaffin cells expressed high affinity membrane testosterone binding sites; (c) activation of these sites increased cytosolic Ca(2+), decreased catecholamine secretion and induced apoptosis; (d) NGF-induced neuronal differentiation of PC12 cells resulted in the suppression of the number of membrane testosterone sites. In conclusion, our data provide evidence for the existence of specific membrane testosterone receptors on adrenal chromaffin cells via which androgens, (some of them originating in the cortex) modulate their function. Neuronal differentiation of chromaffin cells results in a significant attenuation of these effects, via suppression of the expression of membrane androgen receptors suggesting, that the latter are specific for epithelioid chromaffin cells.


Subject(s)
Membrane Proteins/metabolism , Neurons/metabolism , Receptors, Androgen/metabolism , Adrenal Medulla/cytology , Adrenal Medulla/metabolism , Animals , Apoptosis/drug effects , Binding Sites/drug effects , Calcium/metabolism , Catecholamines/biosynthesis , Cell Differentiation , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Flow Cytometry , Membrane Proteins/drug effects , Microscopy, Confocal , Neurons/cytology , PC12 Cells , Rats , Testosterone/metabolism , Testosterone/pharmacology , Time Factors
11.
Eur J Pharmacol ; 530(1-2): 33-9, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16386243

ABSTRACT

Matrix metalloproteinases (MMP) are ubiquitous enzymes involved in extracellular matrix remodeling, and as a consequence in a number of physiological and pathological states, including development, wound healing and cancer. A crucial feature of cancer progression and metastasis is the disruption of extracellular matrix, and spreading of proliferating cancer cells. Modulation of MMP is a main target of cancer research. Using the mouse fibrosarcoma cell line WEHI 164, producing high amounts of MMP-2, we investigated whether we could modulate its production. We report that MMP-2 is under the control of nitric oxide (NO)/nitric oxide synthase (NOS) system. In addition, we show that NOS activity is controlled by opioids in a non-opioid receptor-related manner. Finally, we provide evidence that morphine, when administrated at low, non-toxic concentrations (<10(-9) M) attenuates MMP-2 activity. We conclude that, as morphine is able to decrease metalloproteinase activity via the NO/NOS system, it may have a place in the treatment of several sarcomas including fibrosarcoma.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Morphine/pharmacology , Nitric Oxide/metabolism , Animals , Cell Proliferation/drug effects , Diprenorphine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Flow Cytometry , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , NG-Nitroarginine Methyl Ester/pharmacology , Narcotic Antagonists/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Opioid/genetics , Reverse Transcriptase Polymerase Chain Reaction , S-Nitroso-N-Acetylpenicillamine/pharmacology , Tumor Cells, Cultured
12.
FASEB J ; 20(3): 577-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16407456

ABSTRACT

The neurosteroid dehydroepiandrosterone (DHEA) at 1 nM protects NMDA-/GABAA-receptor negative neural crest-derived PC12 cells from apoptosis. We now report that membrane-impermeable DHEA-BSA conjugate replaces unconjugated DHEA in protecting serum-deprived PC12 cells from apoptosis (IC50=1.5 nM). Protection involves phosphorylation of the prosurvival factor Src and induction of the anti-apoptotic protein Bcl-2 and is sensitive to pertussis toxin. Binding assays of [3H]DHEA on isolated PC12 cell membranes revealed saturation within 30 min and binding of DHEA with a Kd of 0.9 nM. A similar binding activity was detectable in isolated membranes from rat hippocampus and from normal human adrenal chromaffin cells. The presence of DHEA-specific membrane binding sites was confirmed by flow cytometry and confocal laser microscopy of DHEA-BSA-FITC stained cells. In contrast to estrogens and progestins, glucocorticoids and androgens displaced DHEA from its membrane binding sites but with a 10-fold lower affinity than DHEA (IC50=9.3 and 13.6 nM, respectively). These agents acted as pure antagonists, blocking the antiapoptotic effect of DHEA as well as the induction of Bcl-2 proteins and Src kinase activation. In conclusion, our findings suggest that neural crest-derived cells possess specific DHEA membrane binding sites coupled to G proteins. Binding to these sites confers neuroprotection.


Subject(s)
Dehydroepiandrosterone/pharmacology , Neuroprotective Agents/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, Steroid/drug effects , Adrenal Medulla/cytology , Androgens/pharmacology , Animals , Apoptosis/drug effects , Cell Membrane/drug effects , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Culture Media, Serum-Free/pharmacology , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/metabolism , Estrogens/pharmacology , Glucocorticoids/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Humans , Male , Microscopy, Confocal , Neuroprotective Agents/metabolism , PC12 Cells/drug effects , Pertussis Toxin/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, GABA-A/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Steroid/metabolism , Serum Albumin, Bovine/pharmacology , bcl-X Protein/physiology
13.
J Dairy Res ; 72 Spec No: 44-50, 2005.
Article in English | MEDLINE | ID: mdl-16180720

ABSTRACT

Experimental and epidemiological studies indicate that antioxidant food polyphenols could have antimitotic activities, interfering with cancer initiation, progression or mortality. Circulating polyphenols are far lower than the nominal value in foods. In the rare studies dealing with polyphenol bioavailability, it was noted that their active concentrations in the blood are <1% of their food concentration. In the present study we investigated the effect of four polyphenols (resveratrol, and the flavonoids quercetin, catechin and epicatechin, major constituents of wine) in the hormone-sensitive human cancer cell line T47D, at concentrations compatible with their calculated plasma concentrations after ingestion of a moderate quantity of wine (nM or pM). Our results indicate that cell growth was decreased, with cells being arrested at the S phase of the cycle. In addition, we provide evidence of a bimodal modulation of the NO/NOS system, affecting its activity and transcription. We show that modulation of this system is sufficient to explain polyphenol action on this cell line. This result suggests a potential importance of wine ingestion and possibly the consumption of other polyphenol-rich dietary foods and drinks in the control of breast cancer cell growth.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Flavonoids/pharmacology , Phenols/pharmacology , Catechin/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Humans , Kinetics , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Polyphenols , Quercetin/pharmacology , Resveratrol , S Phase/drug effects , Stilbenes/pharmacology , Wine/analysis
14.
Exp Cell Res ; 307(1): 41-51, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15922725

ABSTRACT

Classical steroid mode of action involves binding to intracellular receptors, the later acting as ligand-activated nuclear transcription factors. Recently, membrane sites for different steroids have been also identified, mediating rapid, non-genomic, steroid actions. Membrane sites for estrogen and androgen have been found in a number of different cell types, bearing or not classical intracellular receptors. In the present study, with the use of radioligand binding, flow cytometry and confocal laser microscopy, we report that T47D human breast cancer cells express specific and saturable membrane receptors for both estrogen (K(D) 4.06 +/- 3.31 nM) and androgen (K(D) 7.64 +/- 3.15 nM). Upon activation with BSA-conjugated, non-permeable ligands (E(2)-BSA and testosterone-BSA), membrane estrogen receptors protect cells from serum-deprivation-induced apoptosis, while androgen receptors induce apoptosis in serum-supplemented T47D cells. In addition, co-incubation of cells with a fixed concentration of one steroid and varying concentrations of the other reversed the abovementioned effect (apoptosis for androgen, and anti-apoptosis for E(2)), suggesting that the fate of the cell depends on the relative concentration of either steroid in the culture medium. We also report the identification of membrane receptors for E(2) and androgen in biopsy slides from breast cancer patients. Both sites are expressed, with the staining for membrane E(2) being strongly present in ER-negative, less differentiated, more aggressive tumors. These findings suggest that aromatase inhibitors may exert their beneficial effects on breast cancer by also propagating the metabolism of local steroids towards androgen, inducing thus cell apoptosis through membrane androgen receptor activation.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Estradiol/pharmacology , Protein Binding/drug effects , Receptors, Androgen/drug effects , Receptors, Estrogen/drug effects , Testosterone/pharmacology , Binding Sites , Biopsy , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Kinetics , Microscopy, Confocal , Radioligand Assay , Retrospective Studies
15.
Exp Cell Res ; 294(2): 434-45, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15023532

ABSTRACT

Neuropeptides influence cancer cell replication and growth. Opioid peptides, and opiergic neurons are found in the prostate gland, and they are proposed to exert a role in tumor regulation, influencing cancer cell growth, as opioid agonists inhibit cell growth in several systems, including the human prostate cancer cell line LNCaP. In the same cell line, the existence of membrane testosterone receptors was recently reported, which increase, in a non-genomic manner, the secretion of PSA, and modify actin cytoskeleton dynamics, through the signaling cascade FAK-->PI-3 kinase-->Cdc42/Rac1. In the present work, we present data supporting that the general opioid agonist Ethylketocyclazocine (EKC) decreases testosterone-BSA (a non-internalizable testosterone analog) induced PSA secretion. Furthermore, we report that this opioid affects this non-genomic testosterone action, by modifying the distribution of the actin cytoskeleton in the cells, disrupting the above signaling cascade. In addition, after long (>24 h) incubation, opioids decrease the number of membrane testosterone receptors, and reverse their effect on the signaling molecules. In conclusion, our results provide some new insights of a possible action of opioids in prostate cancer control by interfering with the action and the expression of membrane testosterone receptors and signaling.


Subject(s)
Carcinoma/metabolism , Cell Membrane/metabolism , Ethylketocyclazocine/pharmacology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Testosterone/analogs & derivatives , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Analgesics, Opioid/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Carcinoma/drug therapy , Cell Division/drug effects , Cell Division/physiology , Cell Membrane/drug effects , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Male , Opioid Peptides/agonists , Opioid Peptides/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Reaction Time/drug effects , Reaction Time/physiology , Receptors, Androgen/drug effects , Serum Albumin, Bovine/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Testosterone/metabolism , Testosterone/pharmacology , Tumor Cells, Cultured
16.
Breast Cancer Res ; 6(2): R63-74, 2004.
Article in English | MEDLINE | ID: mdl-14979919

ABSTRACT

INTRODUCTION: The oncoprotective role of food-derived polyphenol antioxidants has been described but the implicated mechanisms are not yet clear. In addition to polyphenols, phenolic acids, found at high concentrations in a number of plants, possess antioxidant action. The main phenolic acids found in foods are derivatives of 4-hydroxybenzoic acid and 4-hydroxycinnamic acid. METHODS: This work concentrates on the antiproliferative action of caffeic acid, syringic acid, sinapic acid, protocatechuic acid, ferulic acid and 3,4-dihydroxy-phenylacetic acid (PAA) on T47D human breast cancer cells, testing their antioxidant activity and a number of possible mechanisms involved (interaction with membrane and intracellular receptors, nitric oxide production). RESULTS: The tested compounds showed a time-dependent and dose-dependent inhibitory effect on cell growth with the following potency: caffeic acid > ferulic acid = protocatechuic acid = PAA > sinapic acid = syringic acid. Caffeic acid and PAA were chosen for further analysis. The antioxidative activity of these phenolic acids in T47D cells does not coincide with their inhibitory effect on tumoral proliferation. No interaction was found with steroid and adrenergic receptors. PAA induced an inhibition of nitric oxide synthase, while caffeic acid competes for binding and results in an inhibition of aryl hydrocarbon receptor-induced CYP1A1 enzyme. Both agents induce apoptosis via the Fas/FasL system. CONCLUSIONS: Phenolic acids exert a direct antiproliferative action, evident at low concentrations, comparable with those found in biological fluids after ingestion of foods rich in phenolic acids. Furthermore, the direct interaction with the aryl hydrocarbon receptor, the nitric oxide synthase inhibition and their pro-apoptotic effect provide some insights into their biological mode of action.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Hydroxybenzoates/pharmacology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Receptors, Aryl Hydrocarbon/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...