Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Curr Alzheimer Res ; 10(7): 697-705, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23905995

ABSTRACT

The soluble amyloid ß precursor protein α (sAßPPα) released after α-secretase cleavage of the amyloid ß precursor protein (AßPP) has several functions including modulation of neuronal excitability and synaptic plasticity; it has been suggested that some of these effects are mediated by activation of NF-κB via induction of PI3K/Akt signaling pathway. We have recently described the presence of several consensus binding sites of c-Rel transcription factor in the promoter region of the GNB2L1 gene, coding for the Receptor for Activated C Kinase -1 (RACK-1). We investigated whether sAßPPα could influence the expression of RACK-1 through NF-κB involvement. Our data demonstrate that sAßPPα regulates RACK-1 gene expression through PI3K/Akt-dependent pathway, inducing c-Rel nuclear translocation and NF-κB activation. Since RACK-1 is the scaffold of protein kinase C ßII (PKCßII), we turned our attention to this kinase in order to evaluate whether sAßPPα could also influence PKCßII signalling demonstrating that sAßPPα induces PKCßII translocation and interaction with its scaffold with consequent RACK-1/PKCßII complex increase in membrane. Altogether these results suggest the existence of an interesting loop between the functions of the metabolic products of AßPP and the role of PKC and that the impact of a dysregulated AßPP metabolism occurring in several conditions (from physiological aging to injury response) may have consequences on the potential protective functions of the non amyloidogenic sAßPPα.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , GTP-Binding Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Protein Kinase C beta/biosynthesis , Receptors, Cell Surface/biosynthesis , Signal Transduction/physiology , Cell Line, Tumor , Humans , Protein Binding/physiology , Receptors for Activated C Kinase
2.
J Alzheimers Dis ; 30(2): 393-405, 2012.
Article in English | MEDLINE | ID: mdl-22451313

ABSTRACT

The amyloid-ß protein precursor (AßPP) can be processed by either the amyloidogenic or the non-amyloidogenic pathway; both pathways lead to release of the AßPP intracellular C-terminal domain (AICD). AICD involvement in signal transduction within Fe65/Tip60 complex is one of the most discussed mechanisms, and different models have been hypothesized to explain the role of AICD within this complex. The analysis of these models in relation to the degradation processes highlights the discrepancy among AICD localization, function, and degradation, leading to the hypothesis that a signaling mechanism may exist which allows AßPP proteolysis to generate either a transcriptionally active fragment or an inactive one with different involvement of proteasome and IDE (insulin-degrading enzyme). Our work aimed to analyze the functional role of AICD within the Fe65/Tip60 complex considering the AICD degradation processes. Our data suggest a correlation between the role of AICD in gene regulation and its removal operated by proteasome activity. Moreover, treatments with IDE inhibitor underlined the presence of an alternative mechanism involved in AICD removal when the latter is not exerting nuclear activity, thus providing clearer support for the existence of at least two mechanisms as previously suggested.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Amyloidosis/genetics , Amyloidosis/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Insulysin/genetics , Insulysin/metabolism , Lysine Acetyltransferase 5 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary/physiology , Signal Transduction/physiology , Structure-Activity Relationship , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
J Neurochem ; 115(6): 1299-308, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21039524

ABSTRACT

In recent decades, the study of the amyloid precursor protein (APP) and of its proteolytic products carboxy terminal fragment (CTF), APP intracellular C-terminal domain (AICD) and amyloid beta has been mostly focussed on the role of APP as a producer of the toxic amyloid beta peptide. Here, we reconsider the role of APP suggesting, in a provocative way, the protein as a central player in a putative signalling pathway. We highlight the presence in the cytosolic tail of APP of the YENPTY motif which is typical of tyrosine kinase receptors, the phosphorylation of the tyrosine, serine and threonine residues, the kinases involved and the interaction with intracellular adaptor proteins. In particular, we examine the interaction with Shc and Grb2 regulators, which through the activation of Ras proteins elicit downstream signalling events such as the MAPK pathway. The review also addresses the interaction of APP, CTFs and AICD with other adaptor proteins and in particular with Fe65 for nuclear transcriptional activity and the importance of phosphorylation for sorting the secretases involved in the amyloidogenic or non-amyloidogenic pathways. We provide a novel perspective on Alzheimer's disease pathogenesis, focussing on the perturbation of the physiological activities of APP-CTFs and AICD as an alternative perspective from that which normally focuses on the accumulation of neurotoxic proteolytic fragments.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Intracellular Space/chemistry , Intracellular Space/physiology , Peptide Fragments/metabolism , Phosphorylation/physiology , Protein Structure, Tertiary/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic/methods
4.
Pharmacol Res ; 62(4): 308-17, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20561999

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that represents the most common type of dementia in the elderly. One of the hallmarks of this disease is a progressive accumulation of amyloid fibrils in senile plaques (SPs), which are composed principally of amyloid-beta peptides (Abeta). The 4-kDa beta-amyloid peptides are produced from the beta-amyloid precursor protein (APP) through sequential processing by beta- and gamma-secretase enzymes in the amyloidogenic pathway. By an alternative non-amyloidogenic pathway, mediated by alpha- and gamma-secretases enzymes, APP is processed within the Abeta domain. Both processing pathways may result in the generation of a fragment called APP intracellular C-terminal domain (AICD) which is hypothesized to contribute to the pathophysiology of AD. Experimental evidence highlights that biological functions of AICD are mediated by interactions between its YENPTY motif and specific binding factors. We critically reviewed literature concerning physiological function of this proteolitic fragment, mainly focusing on their degradation by the two best characterized systems, proteasome and IDE (insulin degrading enzyme). Our work is aimed to analyse the functional role of AICD, integrating also the AICD degradation processes, to better define a potential role of AICD in signal transduction.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Insulysin/metabolism , Proteasome Endopeptidase Complex/genetics , Signal Transduction , Alzheimer Disease/metabolism , Animals , Humans , Protein Structure, Tertiary
5.
Clin Cancer Res ; 14(16): 5022-32, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18698020

ABSTRACT

PURPOSE: Hypothalamic or locally produced growth factors and cytokines control pituitary development, functioning, and cell division. We evaluated the expression of the chemokine stromal cell-derived factor 1 (SDF1) and its receptor CXCR4 in human pituitary adenomas and normal pituitary tissues and their role in cell proliferation. EXPERIMENTAL DESIGN: The expression of SDF1 and CXCR4 in 65 human pituitary adenomas and 4 human normal pituitaries was determined by reverse transcription-PCR, immunohistochemistry, and confocal immunofluorescence. The proliferative effect of SDF1 was evaluated in eight fibroblast-free human pituitary adenoma cell cultures. RESULTS: CXCR4 mRNA was expressed in 92% of growth hormone (GH)-secreting pituitary adenomas (GHoma) and 81% of nonfunctioning pituitary adenomas (NFPA), whereas SDF1 was identified in 63% and 78% of GHomas and NFPAs, respectively. Immunostaining for CXCR4 and SDF1 showed a strong homogenous labeling in all tumoral cells in both GHomas and NFPAs. In normal tissues, CXCR4 and SDF1 were expressed only in a subset of anterior pituitary cells, with a lower expression of SDF1 compared with its cognate receptor. CXCR4 and SDF1 were not confined to a specific cell population in the anterior pituitary but colocalized with discrete subpopulations of GH-, prolactin-, and adrenocorticorticotropic hormone-secreting cells. Conversely, most of the SDF1-containing cells expressed CXCR4. In six of eight pituitary adenoma primary cultures, SDF1 induced a statistically significant increase in DNA synthesis that was prevented by the treatment with the CXCR4 antagonist AMD3100 or somatostatin. CONCLUSIONS: CXCR4 and SDF1 are overexpressed in human pituitary adenomas and CXCR4 activation may contribute to pituitary cell proliferation and, possibly, to adenoma development in humans.


Subject(s)
Chemokine CXCL12/biosynthesis , Pituitary Neoplasms/metabolism , Receptors, CXCR4/biosynthesis , Cell Proliferation , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Hybridization , Microscopy, Confocal , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
6.
Ann N Y Acad Sci ; 1096: 249-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17405936

ABSTRACT

The mayor pathologic hallmarks of Alzheimer's disease (AD) are senile plaque and neurofibrillary tangles. Senile plaque are primarily made up of deposits of amyloid-beta protein, a proteolytic product derived from the amyloid precursor protein (APP). APP is a transmembrane protein detected into the endoplasmic reticulum, in the Golgi apparatus, at the cell surface, recycled by endocytosis to endosomes, whose physiological function is unclear. Presenilins (PS), are a component of gamma-secretase complex that cleave alpha-CTFs (carboxy-terminal fragment), or beta-CTFs, leaving 40 or 42 amino acids amyloid-beta peptides and 58 or 56 amino acids intracellular domains (AICD). Where the amyloid-beta peptides is generated is not clear. The study of APP-PS interaction in specific cell compartments provides a good opportunity to light upon the molecular mechanisms regulating the activity of the "gamma-secretase complex," and where beta-amyloid is generated. In our study we used a biophysical assay of protein proximity: fluorescence resonance energy transfer (FRET), that can provide information about molecular interactions when two proteins are in the close proximity (<10 nm), to examine the subcellular localization and interaction between APP and PS1 in human neuroglioma cells (H4). Confocal microscopic analysis reveals extensive colocalization in different cells' compartment, and centrosomal or microtubule organizing center (MTOC) localization of APP and PS1, but not necessarily a close molecular interaction. We used FRET to determine if APP and PS1 interact at the cell centrosome. FRET data suggest a close interaction between APP and PS1 in subcellular compartments and at the centrosome of H4 cells. Using this approach we show that APP and PS1 are closely associated in the centrosomes of the H4 cell.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Centrosome/ultrastructure , Fluorescence Resonance Energy Transfer/instrumentation , Fluorescence Resonance Energy Transfer/methods , Presenilin-1/biosynthesis , Amyloid beta-Protein Precursor/chemistry , Cell Line, Tumor , Centrosome/metabolism , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Presenilin-1/chemistry , Protein Binding
7.
Ann N Y Acad Sci ; 1096: 258-70, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17405937

ABSTRACT

The conversion of the prion protein (PrP) into a protease-resistant isoform (PrP(Res)) is considered the pathogenic event responsible for prion encephalopathies. Microglia activation accompanies PrP(Res) deposition representing an early event in the progression of these diseases. It is now believed that microglial cells play a worsening, if not causative, role in prion-induced neuronal death, through the release of proinflammatory and neurotoxic molecules. Indeed, in vitro observations have demonstrated that PrP(Res) and the synthetic prion fragment PrP106-126 induce neuronal death by activating microglial to migrate in the lesion area and secrete cytokines. Recently, we and others have demonstrated that the recombinant peptide, corresponding to the protease-resistant portion of PrP encompassing the amino acids 90-231 (PrP90-231), when beta-structured, is toxic for neuronal cells, in vitro. Here we report that PrP90-231 induces activation of N9 microglial cells, characterized by cell proliferation arrest and increased secretion of different cytokines (RANTES, GCSF, and IL-12). Moreover, the treatment of N9 cells with PrP90-231 elicited inducible nitric oxide synthase (i-NOS) expression, nitric oxide release, and a delayed (15 min to 1 h of treatment) extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation/activation. Although ERK1/2 is known to regulate proliferative and differentiative events, we show that its blockade, using the specific MEK inhibitor PD98059, did not prevent PrP90-231-induced inhibition of N9 cell proliferation. To our knowledge, this is the first evidence that a recombinant PrP(Res)-like peptide elicits microglial activation in vitro, thus representing a potentially important tool to develop possible therapeutic strategies to target prion-induced brain inflammation.


Subject(s)
Microglia/metabolism , Prions/chemistry , Animals , Cell Line , Cell Proliferation , Cytokines/metabolism , Inflammation , Mice , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitrites/chemistry , Peptides/chemistry , Protein Structure, Tertiary , Time Factors
8.
J Mol Endocrinol ; 38(3): 383-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17339401

ABSTRACT

Chemokines are key factors involved in the regulation of immune response, through the activation and control of leukocyte traffic, lymphopoiesis and immune surveillance. However, a large number of chemokines and their receptors are expressed in central nervous system (CNS) cells, either constitutively or induced by inflammatory stimuli, playing a role in many neuropathological processes. Stromal cell-derived factor 1 (SDF1) is a chemokine whose extra-immunological localization and functions have been extensively studied. SDF1 and its receptor CXCR4 were identified in both neurons and glia of many brain areas, including the hypothalamus, as well as at the pituitary level. Importantly, SDF1 and CXCR4 expression is increased in brain tumors in which their activity induced tumor cell proliferation and brain parenchyma invasion. Despite their localization, to date very few reports addressed the role of CXCR4 and SDF1 in the modulation of the hypothalamus/pituitary axis and their possible involvement in the development of pituitary adenomas. In this review, we discuss previous literature data on the role of chemokines in normal and adenomatous pituitary cells, focusing on recent data from our group showing that CXCR4 activation controls proliferation and both prolactin and GH release in the pituitary adenoma cell line GH4C1 through a complex network of intracellular signals. Thus, the SDF1/CXCR4 system together with other chemokinergic ligand-receptor pairs, may represent a novel regulatory pathway for pituitary function and, possibly, be involved in pituitary adenoma development. These lines of evidence suggest that the inhibition of chemokine receptors may represent a novel pharmacological target for the treatment of pituitary adenomas.


Subject(s)
Chemokines, CXC/physiology , Pituitary Gland, Anterior/physiology , Adenoma/pathology , Adenoma/physiopathology , Cell Proliferation , Chemokine CXCL12 , Humans , Neurosecretory Systems/physiology , Pituitary Neoplasms/pathology , Pituitary Neoplasms/physiopathology
9.
J Biol Chem ; 282(18): 13833-44, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17314098

ABSTRACT

The amyloid precursor protein (APP) and the presenilins 1 and 2 are genetically linked to the development of familial Alzheimer disease. APP is a single-pass transmembrane protein and precursor of fibrillar and toxic amyloid-beta peptides, which are considered responsible for Alzheimer disease neurodegeneration. Presenilins are multipass membrane proteins, involved in the enzymatic cleavage of APP and other signaling receptors and transducers. The role of APP and presenilins in Alzheimer disease development seems to be related to the formation of amyloid-beta peptides; however, their physiological function, reciprocal interaction, and molecular mechanisms leading to neurodegeneration are unclear. APP and presenilins are also involved in multiple interactions with intracellular proteins, the significance of which is under investigation. Among the different APP-interacting proteins, we focused our interest on the GRB2 adaptor protein, which connects cell surface receptors to intracellular signaling pathways. In this study we provide evidence by co-immunoprecipitation experiments, confocal and electron microscopy, and by fluorescence resonance energy transfer experiments that both APP and presenilin1 interact with GRB2 in vesicular structures at the centrosome of the cell. The final target for these interactions is ERK1,2, which is activated in mitotic centrosomes in a PS1- and APP-dependent manner. These data suggest that both APP and presenilin1 can be part of a common signaling pathway that regulates ERK1,2 and the cell cycle.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , GRB2 Adaptor Protein/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Presenilin-1/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Line , Centromere/metabolism , Centromere/ultrastructure , Fluorescence Resonance Energy Transfer , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission , Presenilin-2/metabolism , Protein Binding
10.
Chem Biol Interact ; 165(2): 138-45, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17196952

ABSTRACT

Current therapies for Alzheimer's disease treatment rely mainly on acetylcholinesterase inhibitors, improving central cholinergic neurotransmission. Among these molecules, galantamine (GAL) has an interesting pharmacological profile as it is both a reversible acetylcholinesterase inhibitor and an allosteric potentiator of nicotinic cholinergic receptors. We investigated the effect of GAL on the metabolism of the amyloid precursor protein (APP) in differentiated SH-SY5Y neuroblastoma cells. The rationale was based on the suggestion that cholinergic activity may also be involved in the regulation of APP metabolism. We studied the acute effect on APP metabolism measuring the secretion of sAPPalpha in the conditioned medium of cells. Following 2h treatment, GAL 10microM promoted a strong increase in the release of sAPPalpha, the maximal effect approaching on average three-fold baseline value. The compound appeared to increase the release of sAPPalpha, with a mechanism dependent upon an indirect cholinergic stimulation. The effect of GAL was prevented by pre-treatment with alpha-bungarotoxin (40nM) but not low (nanomolar) atropine concentrations, suggesting the specific involvement of nicotinic cholinergic receptors.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cholinesterase Inhibitors/pharmacology , Galantamine/pharmacology , Neurons/drug effects , Receptors, Nicotinic/metabolism , Acetylcholinesterase/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neurons/metabolism
11.
Neuro Oncol ; 9(1): 3-11, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17108064

ABSTRACT

Chemokines participate in cellular processes associated with tumor proliferation, migration, and angiogenesis. We previously demonstrated that stromal cell-derived factor 1 (SDF1) exerts a mitogenic activity in glioblastomas through the activation of its receptor CXCR4. Here we studied the expression of this chemokine in human meningiomas and its possible role in cell proliferation. Reverse transcriptase-PCR analysis for CXCR4 and SDF1 was performed on 55 human meningiomas (47 WHO grade I, 5 WHO II, and 3 WHO III). Immunolabeling for CXCR4 and SDF1 was performed on paraffin-embedded sections of these tumors. [(3)H]Thymidine uptake and Western blot analyses were performed on primary meningeal cell cultures of tumors to evaluate the proliferative activity of human SDF1alpha (hSDF1alpha) in vitro and the involvement of extracellular signal-regulated kinase 1/2 (ERK1/2) activation in this process. CXCR4 mRNA was expressed by 78% of the tumor specimens and SDF1 mRNA by 53%. CXCR4 and SDF1 were often detected in the same tumor tissues and colocalized with epithelial membrane antigen immunostaining. In 9 of 12 primary cultures from meningiomas, hSDF1alpha induced significant cell proliferation that was strongly reduced by the mitogen-activated protein kinase kinase inhibitor PD98059, involving ERK1/2 activation in the proliferative signal of hSDF1alpha. In fact, CXCR4 stimulation led to ERK1/2 phosphorylation/activation. In addition, the hSDF1alpha-induced cell proliferation was significantly correlated with the MIB1 staining index in the corresponding surgical specimen. In conclusion, we found that human meningiomas express CXCR4 and SDF1 and that hSDF1alpha induces proliferation in primary meningioma cell cultures through the activation of ERK1/2.


Subject(s)
Cell Proliferation , Chemokines, CXC/metabolism , Gene Expression Regulation, Neoplastic , Meningeal Neoplasms/metabolism , Meningioma/metabolism , Receptors, CXCR4/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Chemokine CXCL12 , Chemokines, CXC/genetics , Female , Humans , Immunoenzyme Techniques , In Vitro Techniques , Male , Meningeal Neoplasms/genetics , Meningioma/genetics , Meningioma/pathology , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, CXCR4/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stromal Cells/metabolism , Tumor Cells, Cultured
12.
Neurochem Int ; 49(5): 423-32, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16621164

ABSTRACT

Chemokines have been involved in cellular processes associated to malignant transformation such as proliferation, migration and angiogenesis. The expression of five CXC chemokine receptors and their main ligands was analysed by RT-PCR in 31 human astrocytic neoplasms. The mRNAs for all the receptors analysed were identified in a high percentage of tumours, while their ligands showed lower expression. CXCR4 and SDF1 were the most frequently mRNA identified (29/31 and 13/31 of the gliomas studied, respectively). Thus, we further analysed the cell localization of CXCR4 and SDF1 in immunohistochemistry experiments. We show a marked co-localization of CXCR4 and SDF1 in tumour cells, mainly evident in psudolpalisade and microcystic degeneration areas and in the vascular endothelium. In addition, hSDF1alpha induced a significant increase of DNA synthesis in primary human glioblastoma cell cultures and chemotaxis in a glioblastoma cell line. These results provide evidence of the expression of multiple CXC chemokines and their receptors in brain tumours and that in particular CXCR4 and SDF1 sustain proliferation and migration of glioma cells to promote malignant progression.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement/physiology , Cell Proliferation , Chemokines, CXC/physiology , Glioma/metabolism , Receptors, Chemokine/metabolism , Base Sequence , Brain Neoplasms/pathology , Cell Line, Tumor , Chemokines, CXC/metabolism , DNA Primers , Glioma/pathology , Humans , Immunohistochemistry , Ligands , Receptors, Chemokine/physiology , Reverse Transcriptase Polymerase Chain Reaction
13.
Mol Pharmacol ; 69(2): 539-46, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16258074

ABSTRACT

We used GH4C1 cells as a model to study the effects of the chemokine stromal cell-derived factor 1 (SDF1) in pituitary functions. In these cells, SDF1alpha induced proliferation and growth hormone secretion, suggesting a possible regulatory role for this chemokine at pituitary level. We evaluated the intracellular signaling involved in these effects: SDF1alpha increased cytosolic [Ca(2+)] and activated Pyk2, extracellular signal-regulated kinases 1 and 2 (ERK1/2), and large-conductance Ca(2+)-activated K(+) channels (BK(Ca)) channels. To correlate these intracellular effectors with the proliferative and secretory effects, we inhibited their activity using BAPTA-AM (Ca(2+) chelator), 2'-amino-3'-methoxyflavone (PD98059; a mitogen-activated protein kinase kinase inhibitor), salicylate (Pyk2 inhibitor), and tetraethyl ammonium (K(+) channel blocker). All of these compounds reverted SDF1alpha-induced proliferation, suggesting the involvement of multiple intracellular pathways. Conversely, only BAPTA-AM reverted growth hormone secretion. To identify a possible cross-talk and a molecular ordering among these pathways, we tested these antagonists on SDF1alpha-dependent activation of ERK1/2, Pyk2, and BK(Ca) channels. From these experiments, we observed that the inhibition of [Ca(2+)](i) increase or BK(Ca) channel activity did not affect ERK1/2 activation by SDF1alpha; Pyk2 activation was purely Ca(2+)-dependent, not involving ERK1/2 or BK(Ca) channels; and BK(Ca) channel activity was antagonized by Pyk2 but not by ERK1/2 inhibitors. These data suggest that an SDF1alpha-dependent increase of [Ca(2+)](i) activates Pyk2, which in turn regulates BK(Ca) channel activity. Conversely, ERK1/2 activation is an independent phenomenon. In conclusion, we demonstrate that SDF1alpha causes both proliferation and growth hormone release from pituitary adenoma cells, suggesting that the activation of CXCR4 may represent a novel regulatory mechanism for growth hormone secretion and pituitary cell proliferation, which may contribute to pituitary adenoma development.


Subject(s)
Adenoma/metabolism , Chemokines, CXC/pharmacology , Growth Hormone/metabolism , Pituitary Gland/drug effects , Pituitary Neoplasms/metabolism , Receptors, CXCR4/agonists , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12 , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Pituitary Gland/metabolism , Pituitary Gland/physiology , Rats , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction/drug effects
14.
Ann N Y Acad Sci ; 1090: 276-91, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17384271

ABSTRACT

Prion diseases comprise a group of fatal neurodegenerative disorders that affect both animals and humans. The transition of the prion protein (PrP) from a mainly alpha-structured isoform (PrPC) to a prevalent beta-sheet-containing protein (PrPSc) is believed to represent a major pathogenetic mechanism in prion diseases. To investigate the linkage between PrP neurotoxicity and its conformation, we used a recombinant prion protein fragment corresponding to the amino acidic sequence 90-231 of human prion protein (hPrP90-231). Using thermal denaturation, we set up an experimental model to induce the process of conversion from PrPC to PrPSc. We report that partial thermal denaturation converts hPrP90-231 into a beta-sheet-rich isoform, displaying a temperature- and time-dependent conversion into oligomeric structures that share some physico-chemical characteristics with brain PrPSc. SH-SY5Y cells were chosen to characterize the potential neurotoxic effect of hPrP90-231 in its different structural conformations. We demonstrated that hPrP90-231 in beta-conformation, but not when alpha-structured, powerfully affected the survival of these cells. hPrP90-231 beta-structured caused DNA fragmentation and a significant increase in caspase-3 proteolytic activity (maximal effects+170%), suggesting the occurrence of apoptotic cell death. Finally, we investigated the involvement of MAP kinases in the regulation of beta-hPrP90-231-dependent apoptosis. We observed that the p38 MAP kinase blocker SB203580 prevented the apoptotic cell death evoked by hPrP90-231, and Western blot analysis revealed that the exposure of the cells to the peptide induced p38 phosphorylation. In conclusion, we demonstrate that the hPrP90-231 elicits proapoptotic activity when in beta-sheet-rich conformation and that this effect is mediated by p38 and caspase-3 activation.


Subject(s)
Apoptosis , Prions/metabolism , Caspases/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Prions/chemistry , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Ann N Y Acad Sci ; 1090: 332-43, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17384278

ABSTRACT

Recent evidence indicates that cancer cells express chemokine (CK) receptors and that their signaling is crucial for tumor proliferation, migration, and angiogenesis. The profiles of expression of CXC CK receptors (CXCR1-5) and their main ligands (growth-related oncogene, GRO1-2-3/CXCL1-2-3; interleukin 8, IL-8/CXCL8; monokine-induced gamma-interferon MIG/CXCL9; gamma-interferon-inducible-protein-10, IP-10/CXCL10; stromal cell-derived factor-1, SDF1/CXCL12; B-cell activating CK-1, BCA-1/CXCL13) were analyzed by reverse transcription polymerase chain reaction (RT-PCR) in surgical samples of human meningiomas. All the five receptors displayed high percentages of positive cases: 92% CXCR1, 89% CXCR2, 83% CXCR3, 78% CXCR4, and 94% CXCR5. Conversely, their ligands showed a lower pattern of expression: 40% IL-8, 42% GRO1-3, 42% IP-10, 28% MIG, 53% SDF1, and 3% BCA-1. SDF1/CXCR4 interaction plays a pivotal role in cancer proliferation. Thus, the signaling mechanisms activated by the exclusive binding between SDF1 and CXCR4 was investigated in 12 primary cultures from meningioma tissues. CXCR4 was functionally coupled as demonstrated by the significant increase of DNA synthesis in meningioma cells in response to SDF1, measured by [3H]-thymidine uptake. In three primary cultures, the SDF1-dependent mitogenic activity was associated with a marked phosphorylation of extracellular signal-regulated kinase (ERK1/2) as evaluated by Western blots. PD98059 (a MEK inhibitor) significantly reduced ERK1/2 activation, thus linking the SDF1/CXCR4 pathway to meningioma cell proliferation via ERK1/2 signal transduction. We demonstrate, for the first time in human meningiomas, the simultaneous expression of CXCR1-5 and their CKs and the mitogenic activity of SDF1/CXCR4, suggesting a pivotal role of these receptor-ligand pairs in meningeal tumors.


Subject(s)
Cell Proliferation , Chemokines, CXC/metabolism , Meningioma/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, CXCR4/metabolism , Adult , Aged , Aged, 80 and over , Base Sequence , Chemokine CXCL12 , DNA Primers , Enzyme Activation , Female , Humans , Male , Meningioma/enzymology , Meningioma/pathology , Middle Aged , Tumor Cells, Cultured
16.
Ann N Y Acad Sci ; 1090: 385-98, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17384283

ABSTRACT

Stromal cell-derived factor-1 (SDF-1) is a chemokine of the CXC subfamily that exerts its effects via CXCR4, a G-protein-coupled receptor. CXCR4 is often expressed by tumor cells, and its activation causes tumor cell proliferation. Using GH4C1 cells, here we show that SDF-1 induced cell proliferation in a dose-dependent manner. Thus, we evaluated the intracellular signaling involved in this effect. SDF-1 increased cytosolic [Ca2+] and activated Pyk2, ERK1/2, and BKCa channels. To correlate these intracellular effectors with the proliferative activity of SDF-1, we inhibited their activity using BAPTA-AM (Ca2+ chelator), PD98059 (MEK inhibitor), salicylate (Pyk2 inhibitor), and TEA (K+ channel blocker). All these compounds reverted SDF-1-induced proliferation, suggesting the involvement of multiple intracellular pathways. To identify a possible crosstalk and a molecular ordering among these pathways, we tested these antagonists on SDF-1-dependent activation of ERK1/2, Pyk2, and BKCa channels. We report that the inhibition of [Ca2+]i increase or the blockade of BKCa channel activity did not affect ERK1/2 activation by SDF-1; Pyk2 activation was purely Ca2+-dependent, not involving ERK1/2 or BKCa channels; and BKCa channel activity was antagonized by Pyk2 but not by ERK1/2 inhibitors. These data suggest that SDF-1-dependent increase of [Ca2+]i activates Pyk2, which, in turn, regulates BKCa channel activity. Conversely, ERK1/2 activation is an independent phenomenon. In conclusion, we demonstrate that SDF-1 induces proliferation of GH4C1 cells, suggesting that the activation of CXCR4 may represent a novel regulatory mechanism for pituitary cell proliferation which may contribute to pituitary adenoma development.


Subject(s)
Cell Proliferation , Chemokines, CXC/physiology , Focal Adhesion Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pituitary Neoplasms/pathology , Adenoma/enzymology , Adenoma/metabolism , Adenoma/pathology , Calcium/metabolism , Cell Line , Chemokine CXCL12 , Enzyme Activation , GTP-Binding Proteins/metabolism , Humans , Pituitary Neoplasms/enzymology , Pituitary Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Ann N Y Acad Sci ; 1090: 455-65, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17384289

ABSTRACT

The amyloid precursor protein (APP) is a transmembrane protein with a short cytoplasmic tail whose physiological function is unclear, although it is well documented that the proteolytic processing of APP could influence the development of Alzheimer's disease (AD) through the formation of membrane-bound C-terminal fragments (CTFs) and of beta-amyloid peptides (Abeta). We have recently shown that tyrosine-phosphorylated APP and CTFs may interact with Grb2 and ShcA adaptor proteins and that this coupling occurs at a higher extent in AD subjects only. To study the interaction between APP or CTFs and ShcA/Grb2 and to investigate their molecular target we have used as experimental model two different cell lines: H4 human neuroglioma cells and APP/APLP null mouse embryonic fibroblast cells (MEFs). Here we show that in H4 cells APP interacts with Grb2; conversely in APP/APLP-null MEF cells this interaction is possible only after the reintroduction of human APP by transfection. We have also shown that in MEF cells the transfection of a plasmid encoding for human APP wild-type enhances the phosphorylation of ERK-1 and -2 as revealed by Western blotting and immunofluorescence experiments. Finally, also in H4 cells the overexpression of APP upregulates the levels of phospho-ERK-1 and -2. In summary our data suggest that APP may influence phospho-ERK-1 and -2 signaling through its binding with Grb2 and ShcA adaptors. The meaning of this event is not clear, but APP interaction with these adaptors could be relevant to regulate mitogenic pathway.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Signal Transduction/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line , Fluorescent Antibody Technique , Mice , Microscopy, Confocal
18.
Exp Cell Res ; 308(2): 241-53, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15921680

ABSTRACT

Ovarian cancer (OC) is the leading cause of death in gynecologic diseases in which there is evidence for a complex chemokine network. Chemokines are a family of proteins that play an important role in tumor progression influencing cell proliferation, angiogenic/angiostatic processes, cell migration and metastasis, and, finally, regulating the immune cells recruitment into the tumor mass. We previously demonstrated that astrocytes and glioblastoma cells express both the chemokine receptor CXCR4 and its ligand stromal cell-derived factor-1 (SDF-1), and that SDF-1alpha treatment induced cell proliferation, supporting the hypothesis that chemokines may play an important role in tumor cells' growth in vitro. In the present study, we report that CXCR4 and SDF-1 are expressed in OC cell lines. We demonstrate that SDF-1alpha induces a dose-dependent proliferation in OC cells, by the specific interaction with CXCR4 and a biphasic activation of ERK1/2 and Akt kinases. Our results further indicate that CXCR4 activation induces EGF receptor (EGFR) phosphorylation that in turn was linked to the downstream intracellular kinases activation, ERK1/2 and Akt. In addition, we provide evidence for cytoplasmic tyrosine kinase (c-Src) involvement in the SDF-1/CXCR4-EGFR transactivation. These results suggest a possible important "cross-talk" between SDF-1/CXCR4 and EGFR intracellular pathways that may link signals of cell proliferation in ovarian cancer.


Subject(s)
Carcinoma/metabolism , Chemokines, CXC/metabolism , ErbB Receptors/metabolism , Ovarian Neoplasms/metabolism , Receptors, CXCR4/metabolism , Transcriptional Activation , Carcinoma/physiopathology , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Dose-Response Relationship, Drug , Female , Genes, src/genetics , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Ovarian Neoplasms/physiopathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor Cross-Talk/physiology
19.
Brain Res Brain Res Rev ; 48(2): 257-64, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15850665

ABSTRACT

The amyloid precursor protein (APP) is an ubiquitous receptor-like molecule involved in the pathogenesis of Alzheimer's disease that generates beta-amyloid peptides and causes plaque formation. APP and some of its C-terminal proteolytic fragments (CTFs) have also been shown to be in the center of a complex protein-protein network, where selective phosphorylation of APP C-terminus may regulate the interaction with cytosolic phosphotyrosine binding (PTB) domain or Src homology 2 (SH2) domain containing proteins involved in cell signaling. We have recently described an interaction between tyrosine-phosphorylated CTFs and ShcA adaptor protein which is highly enhanced in AD brain, and a new interaction between APP and the adaptor protein Grb2 both in human brain and in neuroblastoma cultured cells. These data suggest a possible role in cell signaling for APP and its CTFs, in a manner similar to that previously reported for other receptors, through a tightly regulated coupling with intracellular adaptors to control the signaling of the cell. In this review, we discuss the significance of these novel findings for AD development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases , Brain/metabolism , Brain/pathology , Endopeptidases/classification , Endopeptidases/metabolism , GRB2 Adaptor Protein , Humans , Models, Neurological , Phosphorylation , Protein Binding , Tyrosine/metabolism , src Homology Domains/physiology
20.
Oncogene ; 24(19): 3187-95, 2005 Apr 28.
Article in English | MEDLINE | ID: mdl-15735685

ABSTRACT

The expression of the receptor protein tyrosine phosphatase r-PTPeta is drastically reduced in rat and human malignant thyroid cells, whereas its restoration reverts the neoplastic phenotype of retrovirally transformed rat thyroid cells. Moreover, reduced levels and loss of heterozygosity of DEP-1, the human homolog of r-PTPeta, have been found in many human neoplasias. Here, we report that the r-PTPeta protein binds to c-Src in living cells and dephosphorylates the c-Src inhibitory tyrosine phosphorylation site (Tyr 529), thereby increasing c-Src tyrosine kinase activity in malignant rat thyroid cells stably transfected with r-PTPeta. Tyrosine phosphorylation of focal adhesion kinase (FAK) and paxillin was enhanced in r-PTPeta-expressing cells. This was associated with increased adhesion of malignant r-PTPeta-transfected thyroid cells vs both untransfected cells and cells stably transfected with an inactive r-PTPeta mutant. Treatment of rat thyroid cells with the c-Src inhibitor PP2 decreased cell adhesion to a higher extent in r-PTPeta-transfected cells than in mock-transfected or stably transfected cells with the inactive r-PTPeta mutant, indicating that r-PTPeta regulates cell-substratum adhesion by activating c-Src. Interestingly, the extent of both c-Src dephosphorylation at Tyr 529, FAK and paxillin phosphorylation, and the increased cell adhesion were associated with the degree of r-PTPeta expression.


Subject(s)
Phosphotyrosine/chemistry , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases/metabolism , Animals , Blotting, Western , CSK Tyrosine-Protein Kinase , Cell Adhesion , Cytoskeletal Proteins/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Glutathione Transferase/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mutation , Neoplasms/metabolism , Paxillin , Peptides/chemistry , Phenotype , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 3 , Thyroid Gland/cytology , Thyroid Neoplasms/metabolism , Transfection , Tyrosine/chemistry , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...