Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Alzheimers Dis ; 55(3): 1249-1259, 2017.
Article in English | MEDLINE | ID: mdl-27792009

ABSTRACT

BACKGROUND: Several neurological and systemic diseases can cause dementia, beyond Alzheimer's disease. Rare genetic causes are often responsible for dementia with atypical features. Recently, mutations causative for Niemann-Pick type C disease (NPC) have also been implicated in neurodegenerative diseases. NPC is an autosomal recessive lipid storage disorder caused by mutations in NPC1 and NPC2 genes. In adults, clinical presentation mimicking other neurodegenerative diseases makes diagnosis difficult. Recent evidence suggests that heterozygous mutations in NPC genes may take on etiological significance. OBJECTIVE: To investigate the presence of NPC1 and NPC2 mutations in adults affected by neurodegenerative dementia plus. METHODS: We performed a genetic screening on 50 patients using a wide clinical and biochemical approach to characterize the phenotype of mutated patients. RESULTS: Sequencing analysis revealed four different and known heterozygous mutations in NPC1 and NPC2 genes. Patient 1 carried the p. F284LfsX26 in NPC1 and was affected by progressive supranuclear palsy-like syndrome. The remaining three patients showed a corticobasal syndrome and harbored the c.441+1G>A variant of NPC2 (patient 2), the missense p.N222 S mutation associated with the c.1947+8G>C variant in the splice region of intron 12 in NPC1 (patient 3), and the p.V30M mutation in NPC2 (patient 4), respectively. Filipin staining was abnormal in patients 1 and 2. mRNA analysis revealed an altered splicing of the NPC2 gene in patient 2. CONCLUSIONS: Heterozygous mutations of NPC1 and NPC2 genes could contribute to dementia plus, at least in a subset of patients. We highlight the occurrence of NPC1 and NPC2 heterozygous variants in dementia-plus as pathological event.


Subject(s)
Carrier Proteins/genetics , Dementia/genetics , Glycoproteins/genetics , Membrane Glycoproteins/genetics , Mutation/genetics , Aged , Animals , Brain/diagnostic imaging , DNA Mutational Analysis , Dementia/diagnostic imaging , Dementia/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins , Magnetic Resonance Imaging , Male , Middle Aged , Niemann-Pick C1 Protein , Tomography, Emission-Computed, Single-Photon , Vesicular Transport Proteins
2.
Clin Chim Acta ; 455: 39-45, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26790753

ABSTRACT

Niemann-Pick C disease (NPCD) is a rare autosomal recessive neurovisceral disorder with a heterogeneous clinical presentation. Cholestan-3ß,5α,6ß-triol and 7-ketocholesterol have been proposed as biomarkers for the screening of NPCD. In this work, we assessed oxysterols levels in a cohort of Italian patients affected by NPCD and analyzed the obtained results in the context of the clinical, biochemical and molecular data. In addition, a group of patients affected by Niemann-Pick B disease (NPBD) were also analyzed. NPC patients presented levels of both oxysterols way above the cut off value, except for 5 siblings presenting the variant biochemical phenotype who displayed levels of 3ß,5α,6ß-triol below or just above the cut-off value; 2 of them presented also normal levels of 7-KC. Both oxysterols were extremely high in a patient presenting the neonatal systemic lethal phenotype. All NPB patients showed increased oxysterols levels. In conclusion, the reported LC-MS/MS assay provides a robust non-invasive screening tool for NPCD. However, false negative results can be obtained in patients expressing the variant biochemical phenotype. These data strengthen the concept that the results should always be interpreted in the context of the patients' clinical picture and filipin staining and/or genetic studies might still be undertaken in patients with normal levels of oxysterols if symptoms are highly suggestive of NPCD. Both oxysterols are significantly elevated in NPB patients; thus a differential diagnosis should always be performed in patients presenting isolated hepatosplenomegaly, a common clinical sign of both NPCD and NPBD.


Subject(s)
Carrier Proteins/genetics , Cholestanes/blood , Ketocholesterols/blood , Membrane Glycoproteins/genetics , Mutation , Niemann-Pick Diseases/blood , Sphingomyelin Phosphodiesterase/genetics , Calibration , Cohort Studies , Humans , Intracellular Signaling Peptides and Proteins , Italy , Niemann-Pick C1 Protein , Niemann-Pick Diseases/genetics , Reproducibility of Results
3.
FASEB J ; 29(9): 3839-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26018676

ABSTRACT

Acid ß-glucosidase (GCase), the enzyme deficient in Gaucher disease (GD), is transported to lysosomes by the lysosomal integral membrane protein (LIMP)-2. In humans, LIMP-2 deficiency leads to action myoclonus-renal failure (AMRF) syndrome. GD and AMRF syndrome share some clinical features. However, they are different from clinical and biochemical points of view, suggesting that the role of LIMP-2 in the targeting of GCase would be different in different tissues. Besides, the role of LIMP-2 in the uptake and trafficking of the human recombinant (hr)GCase used in the treatment of GD is unknown. Thus, we compared GCase activity and intracellular localization in immortalized lymphocytes, fibroblasts, and a neuronal model derived from multipotent adult stem cells, from a patient with AMRF syndrome, patients with GD, and control subjects. In fibroblasts and neuronlike cells, GCase targeting to the lysosomes is completely dependent on LIMP-2, whereas in blood cells, GCase is partially targeted to lysosomes by a LIMP-2-independent mechanism. Although hrGCase cellular uptake is independent of LIMP-2, its trafficking to the lysosomes is mediated by this receptor. These data provide new insights into the mechanisms involved in the intracellular trafficking of GCase and in the pathogeneses of GD and AMRF syndrome.


Subject(s)
Adult Stem Cells/metabolism , Fibroblasts/metabolism , Glucosylceramidase , Lymphocytes/metabolism , Lysosomal Membrane Proteins/metabolism , Multipotent Stem Cells/metabolism , Receptors, Scavenger/metabolism , Adult , Adult Stem Cells/pathology , Fibroblasts/pathology , Glucosylceramidase/pharmacokinetics , Glucosylceramidase/pharmacology , Humans , Lymphocytes/pathology , Lysosomal Membrane Proteins/genetics , Lysosomes/metabolism , Lysosomes/pathology , Multipotent Stem Cells/pathology , Myoclonic Epilepsies, Progressive/drug therapy , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Receptors, Scavenger/genetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
4.
PLoS One ; 9(2): e89232, 2014.
Article in English | MEDLINE | ID: mdl-24586617

ABSTRACT

UNLABELLED: Low-to-moderate levels of reactive oxygen species (ROS) govern different steps of neurogenesis via molecular pathways that have been decrypted only partially. Although it has been postulated that redox-sensitive molecules are involved in neuronal differentiation, the molecular bases for this process have not been elucidated yet. The aim of this work was therefore to study the role played by the redox-sensitive, multifunctional protein APE1/Ref-1 (APE1) in the differentiation process of human adipose tissue-derived multipotent adult stem cells (hAT-MASC) and embryonic carcinoma stem cells (EC) towards a neuronal phenotype. METHODS AND RESULTS: Applying a definite protocol, hAT-MASC can adopt a neural fate. During this maturation process, differentiating cells significantly increase their intracellular Reactive Oxygen Species (ROS) levels and increase the APE1 nuclear fraction bound to chromatin. This latter event is paralleled by the increase of nuclear NF-κB, a transcription factor regulated by APE1 in a redox-dependent fashion. Importantly, the addition of the antioxidant N-acetyl cysteine (NAC) to the differentiation medium partially prevents the nuclear accumulation of APE1, increasing the neuronal differentiation of hAT-MASC. To investigate the involvement of APE1 in the differentiation process, we employed E3330, a specific inhibitor of the APE1 redox function. The addition of E3330, either to the neurogenic embryonic carcinoma cell line NT2-D1or to hAT-MASC, increases the differentiation of stem cells towards a neural phenotype, biasing the differentiation towards specific subtypes, such as dopaminergic cells. In conclusion, during the differentiation process of stem cells towards a neuroectodermic phenotype, APE1 is recruited, in a ROS-dependent manner, to the chromatin. This event is associated with an inhibitory effect of APE1 on neurogenesis that may be reversed by E3330. Therefore, E3330 may be employed both to boost neural differentiation and to bias the differentiation potential of stem cells towards specific neuronal subtypes. These findings provide a molecular basis for the redox-mediated hypothesis of neuronal differentiation program.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/physiology , Cell Differentiation/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Multipotent Stem Cells/physiology , Neurogenesis/physiology , Adult , Adult Stem Cells/metabolism , Benzoquinones , Blotting, Western , Chromatin/metabolism , Flow Cytometry , Humans , Microscopy, Fluorescence , Multipotent Stem Cells/metabolism , NF-kappa B/metabolism , Oxidation-Reduction , Propionates , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
5.
Bone ; 58: 81-91, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24120669

ABSTRACT

It is well established that osteoblasts, the key cells involved in bone formation during development and in adult life, secrete a number of glycoproteins harboring autocrine and paracrine functions. Thus, investigating the osteoblastic secretome could yield important information for the pathophysiology of bone. In the present study, we characterized for the first time the secretome of human Hobit osteoblastic cells. We discovered that the secretome comprised 89 protein species including the powerful growth factor progranulin. Recombinant human progranulin (6nM) induced phosphorylation of mitogen-activated protein kinase in both Hobit and osteocytic cells and induced cell proliferation and survival. Notably, risedronate, a nitrogen-containing bisphosphonate widely used in the treatment of osteoporosis, induced the expression and secretion of progranulin in the Hobit secretome. In addition, our proteomic study of the Hobit secretome revealed that risedronate induced the expression of ERp57, HSP60 and HSC70, three proteins already shown to be associated with the prevention of bone loss in osteoporosis. Collectively, our findings unveil novel targets of risedronate-evoked biological effects on osteoblast-like cells and further our understanding of the mechanisms of action of this currently used compound.


Subject(s)
Etidronic Acid/analogs & derivatives , Intercellular Signaling Peptides and Proteins/metabolism , Osteoblasts/metabolism , Proteome/metabolism , Animals , Blotting, Western , Cell Line , Cell Survival/drug effects , Electrophoresis, Polyacrylamide Gel , Etidronic Acid/pharmacology , Humans , Mass Spectrometry , Mice , Osteoblasts/drug effects , Progranulins , Reproducibility of Results , Risedronic Acid , Time Factors
6.
Nucleic Acids Res ; 42(2): 1291-302, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24150945

ABSTRACT

Glycogen storage disease type II is a lysosomal storage disorder due to mutations of the GAA gene, which causes lysosomal alpha-glucosidase deficiency. Clinically, glycogen storage disease type II has been classified in infantile and late-onset forms. Most late-onset patients share the leaky splicing mutation c.-32-13T>G. To date, the mechanism by which the c.-32-13T>G mutation affects the GAA mRNA splicing is not fully known. In this study, we demonstrate that the c.-32-13T>G mutation abrogates the binding of the splicing factor U2AF65 to the polypyrimidine tract of exon 2 and that several splicing factors affect exon 2 inclusion, although the only factor capable of acting in the c.-32-13 T>G context is the SR protein family member, SRSF4 (SRp75). Most importantly, a preliminary screening using small molecules described to be able to affect splicing profiles, showed that resveratrol treatment resulted in a significant increase of normal spliced GAA mRNA, GAA protein content and activity in cells transfected with a mutant minigene and in fibroblasts from patients carrying the c-32-13T>G mutation. In conclusion, this work provides an in-depth functional characterization of the c.-32-13T>G mutation and, most importantly, an in vitro proof of principle for the use of small molecules to rescue normal splicing of c.-32-13T>G mutant alleles.


Subject(s)
Glycogen Storage Disease Type II/genetics , Mutation , RNA Splicing , alpha-Glucosidases/genetics , Cells, Cultured , Exons , HeLa Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Nuclear Proteins/metabolism , RNA Splicing/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors , Splicing Factor U2AF , alpha-Glucosidases/metabolism
7.
Mol Biol Cell ; 23(20): 4079-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918947

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) is the main abasic endonuclease in the base excision repair (BER) pathway of DNA lesions caused by oxidation/alkylation in mammalian cells; within nucleoli it interacts with nucleophosmin and rRNA through N-terminal Lys residues, some of which (K(27)/K(31)/K(32)/K(35)) may undergo acetylation in vivo. Here we study the functional role of these modifications during genotoxic damage and their in vivo relevance. We demonstrate that cells expressing a specific K-to-A multiple mutant are APE1 nucleolar deficient and are more resistant to genotoxic treatment than those expressing the wild type, although they show impaired proliferation. Of interest, we find that genotoxic treatment induces acetylation at these K residues. We also find that the charged status of K(27)/K(31)/K(32)/K(35) modulates acetylation at K(6)/K(7) residues that are known to be involved in the coordination of BER activity through a mechanism regulated by the sirtuin 1 deacetylase. Of note, structural studies show that acetylation at K(27)/K(31)/K(32)/K(35) may account for local conformational changes on APE1 protein structure. These results highlight the emerging role of acetylation of critical Lys residues in regulating APE1 functions. They also suggest the existence of cross-talk between different Lys residues of APE1 occurring upon genotoxic damage, which may modulate APE1 subnuclear distribution and enzymatic activity in vivo.


Subject(s)
Cell Nucleolus/enzymology , DNA Damage , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Lysine/metabolism , Acetylation , Cell Proliferation , Enzyme Stability , HeLa Cells , Humans , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Protein Binding , Protein Conformation , Protein Transport , RNA, Ribosomal/metabolism , Sirtuin 1/metabolism , Structure-Activity Relationship
8.
J Cell Biochem ; 112(10): 2920-30, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21630325

ABSTRACT

Parathyroid hormone (PTH) promotes osteoblast survival through a mechanism that depends on cAMP-mediated signaling downstream of the G protein-coupled receptor PTHR1. We present evidence herein that PTH-induced survival signaling is impaired in cells lacking connexin43 (Cx43). Thus, expression of functional Cx43 dominant negative proteins or Cx43 knock-down abolished the expression of cAMP-target genes and anti-apoptosis induced by PTH in osteoblastic cells. In contrast, cells lacking Cx43 were still responsive to the stable cAMP analog dibutyril-cAMP. PTH survival signaling was rescued by transfecting wild type Cx43 or a truncated dominant negative mutant of ßarrestin, a PTHR1-interacting molecule that limits cAMP signaling. On the other hand, Cx43 mutants lacking the cytoplasmic domain (Cx43(Δ245)) or unable to be phosphorylated at serine 368 (Cx43(S368A)), a residue crucial for Cx43 trafficking and function, failed to restore the anti-apoptotic effect of PTH in Cx43-deficient cells. In addition, overexpression of wild type ßarrestin abrogated PTH survival signaling in Cx43-expressing cells. Moreover, ßarrestin physically associated in vivo to wild type Cx43 and to a lesser extent to Cx43(S368A) ; and this association and the phosphorylation of Cx43 in serine 368 were reduced by PTH. Furthermore, induction of Cx43(S368) phosphorylation or overexpression of wild type Cx43, but not Cx43(Δ245) or Cx43(S368A) , reduced the interaction between ßarrestin and the PTHR1. These studies demonstrate that ßarrestin is a novel Cx43-interacting protein and suggest that, by sequestering ßarrestin, Cx43 facilitates cAMP signaling, thereby exerting a permissive role on osteoblast survival induced by PTH.


Subject(s)
Arrestins/metabolism , Connexin 43/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Immunoprecipitation , Mice , Microscopy, Confocal , Osteoblasts/cytology , Protein Binding/drug effects , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
9.
J Proteomics ; 74(7): 1113-22, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21504803

ABSTRACT

Nitrogen-containing bisphosphonates (N-BPs) are therapeutic agents used to treat osteoporosis and promote osteoblast and osteocyte survival. The molecular mechanisms underlying this effect have been extensively studied, but the global changes induced by N-BPs at the protein level are not known. In this context, we investigated the effect of 10(-7)M Risedronate for 1h and 48h on MLO-Y4 osteocytic cells, through a quantitative, label free shotgun proteomic analysis. We described herein a preliminary proteome map of untreated MLO-Y4 cells, composed of 353 protein species. Moreover, we identified 10 and 15 differentially expressed proteins after 1h and 48h of Risedronate treatment, respectively. Among these, PARK7/DJ-1 protein levels were induced up to 3 times and this event was associated with the activation of the pro-survival Akt pathway that we propose as a novel player in the effect of N-BPs on osteocytes. Risedronate was also able to induce the expression and the secretion of the growth factor pro-granulin. In addition, protein prenylation inhibition appeared to be involved in the modulation of MLO-Y4 proteome by RIS in a protein-specific manner. In conclusion, these findings unveil novel functions targeted by N-BPs in osteocytes and could be useful to design novel pharmaceutical compounds.


Subject(s)
Bone Density Conservation Agents/pharmacology , Etidronic Acid/analogs & derivatives , Osteocytes/drug effects , Animals , Blotting, Western , Diphosphonates/pharmacology , Etidronic Acid/pharmacology , Gene Expression Profiling , Mice , Protein Prenylation/drug effects , Proteomics/methods , Proto-Oncogene Proteins c-akt/physiology , Risedronic Acid , Tandem Mass Spectrometry
10.
Endocr Pathol ; 22(1): 18-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21258971

ABSTRACT

Nucleophosmin (NPM) is a multifunctional nucleolar protein that, depending on the context, can act as oncogene or tumour suppressor. Mutations of the NPM1 gene induce delocalization of NPM in acute myeloid leukaemia. Differently, in solid tumours, only NPM overexpression, but not delocalization, has been so far reported. Here, NPM localization in thyroid tumours was investigated. By using immunohistochemistry, we show increase of NPM cytoplasmic localization in follicular adenomas and papillary carcinomas compared to normal thyroid tissue (p = 0.0125 and <0.0001, respectively). NPM1 mutations commonly found in human leukaemia are not present in thyroid tumours. Immunofluorescence in cultured cell lines was utilized to discriminate between nucleolar and nuclear localization. We show that in thyroid cancer cell lines NPM localizes both in the nucleolus and in nucleus, while in non-tumorigenic thyroid cell lines localizes only in nucleolus. Either presence of the histone deacetylase inhibitor trichostatin A or absence of thyroid-stimulating hormone induces NPM nuclear localization in non-tumorigenic thyroid cell lines.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Adenoma/metabolism , Carcinoma, Papillary/metabolism , Nuclear Proteins/metabolism , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/drug therapy , Adenocarcinoma, Follicular/pathology , Adenoma/pathology , Biomarkers, Tumor/metabolism , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cell Nucleolus/metabolism , Cell Nucleolus/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , DNA Mutational Analysis , DNA, Neoplasm/analysis , Humans , Hydroxamic Acids/pharmacology , Nucleophosmin , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Thyrotropin/deficiency , Thyrotropin/pharmacology
11.
Nucleic Acids Res ; 38(22): 8239-56, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20699270

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1), an essential protein in mammals, is involved in base excision DNA repair (BER) and in regulation of gene expression, acting as a redox co-activator of several transcription factors. Recent findings highlight a novel role for APE1 in RNA metabolism, which is modulated by nucleophosmin (NPM1). The results reported in this article show that five lysine residues (K24, K25, K27, K31 and K32), located in the APE1 N-terminal unstructured domain, are involved in the interaction of APE1 with both RNA and NPM1, thus supporting a competitive binding mechanism. Data from kinetic experiments demonstrate that the APE1 N-terminal domain also serves as a device for fine regulation of protein catalytic activity on abasic DNA. Interestingly, some of these critical lysine residues undergo acetylation in vivo. These results suggest that protein-protein interactions and/or post-translational modifications involving APE1 N-terminal domain may play important in vivo roles, in better coordinating and fine-tuning protein BER activity and function on RNA metabolism.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Lysine/metabolism , Acetylation , Amino Acid Sequence , Binding Sites , DNA-(Apurinic or Apyrimidinic Site) Lyase/classification , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nucleophosmin , Peptides/metabolism , Phylogeny , Protein Structure, Tertiary , RNA/metabolism , Sequence Analysis, Protein
12.
Hematol Oncol ; 28(3): 151-5, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19593743

ABSTRACT

Nucleophosmin 1 (NPM1) is an abundant phosphoprotein mainly located in the nucleolus but also shuttling between the nucleus and cytoplasm. NPM1 has been proposed to be involved in synthesis and processing of ribosomal RNA, regulation of chromatin structure and transport of rRNA and ribosomal proteins. NPM1 gene is considered to be implicated in human cancer as it is a frequent target of genetic alterations, primarily in haematopoietic neoplasms. We describe a case of a therapy-responder acute myeloid leukaemia (AML) patient bearing two novel NPM1 mutations. Cells' transfection studies indicate that the presence of one of these mutations is associated to an abnormal nucleolar structure, suggesting that NPM1 may contribute to the control of nucleolar integrity.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Nuclear Proteins/genetics , Cell Nucleolus/genetics , Exons , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Middle Aged , Nucleophosmin , Transfection
13.
Genome Biol ; 10(9): R93, 2009.
Article in English | MEDLINE | ID: mdl-19744312

ABSTRACT

BACKGROUND: Nitrogen-containing bisphosphonates are the elected drugs for the treatment of diseases in which excessive bone resorption occurs, for example, osteoporosis and cancer-induced bone diseases. The only known target of nitrogen-containing bisphosphonates is farnesyl pyrophosphate synthase, which ensures prenylation of prosurvival proteins, such as Ras. However, it is likely that the action of nitrogen-containing bisphosphonates involves additional unknown mechanisms. To identify novel targets of nitrogen-containing bisphosphonates, we used a genome-wide high-throughput screening in which 5,936 Saccharomyces cerevisiae heterozygote barcoded mutants were grown competitively in the presence of sub-lethal doses of three nitrogen-containing bisphosphonates (risedronate, alendronate and ibandronate). Strains carrying deletions in genes encoding potential drug targets show a variation of the intensity of their corresponding barcodes on the hybridization array over the time. RESULTS: With this approach, we identified novel targets of nitrogen-containing bisphosphonates, such as tubulin cofactor B and ASK/DBF4 (Activator of S-phase kinase). The up-regulation of tubulin cofactor B may explain some previously unknown effects of nitrogen-containing bisphosphonates on microtubule dynamics and organization. As nitrogen-containing bisphosphonates induce extensive DNA damage, we also document the role of DBF4 as a key player in nitrogen-containing bisphosphonate-induced cytotoxicity, thus explaining the effects on the cell-cycle. CONCLUSIONS: The dataset obtained from the yeast screen was validated in a mammalian system, allowing the discovery of new biological processes involved in the cellular response to nitrogen-containing bisphosphonates and opening up opportunities for development of new anticancer drugs.


Subject(s)
Cell Cycle Proteins/genetics , Diphosphonates/pharmacology , Mutation , Saccharomyces cerevisiae/genetics , Alendronate/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Cell Movement/drug effects , DNA Breaks, Double-Stranded , DNA Damage , Etidronic Acid/analogs & derivatives , Etidronic Acid/pharmacology , Gene Deletion , Humans , Ibandronic Acid , Microscopy, Confocal , Microscopy, Electron , Microtubules/drug effects , Microtubules/metabolism , Polyisoprenyl Phosphates/pharmacology , RNA Interference , Risedronic Acid , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
15.
Mol Cell Biol ; 29(7): 1834-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19188445

ABSTRACT

APE1/Ref-1 (hereafter, APE1), a DNA repair enzyme and a transcriptional coactivator, is a vital protein in mammals. Its role in controlling cell growth and the molecular mechanisms that fine-tune its different cellular functions are still not known. By an unbiased proteomic approach, we have identified and characterized several novel APE1 partners which, unexpectedly, include a number of proteins involved in ribosome biogenesis and RNA processing. In particular, a novel interaction between nucleophosmin (NPM1) and APE1 was characterized. We observed that the 33 N-terminal residues of APE1 are required for stable interaction with the NPM1 oligomerization domain. As a consequence of the interaction with NPM1 and RNA, APE1 is localized within the nucleolus and this localization depends on cell cycle and active rRNA transcription. NPM1 stimulates APE1 endonuclease activity on abasic double-stranded DNA (dsDNA) but decreases APE1 endonuclease activity on abasic single-stranded RNA (ssRNA) by masking the N-terminal region of APE1 required for stable RNA binding. In APE1-knocked-down cells, pre-rRNA synthesis and rRNA processing were not affected but inability to remove 8-hydroxyguanine-containing rRNA upon oxidative stress, impaired translation, lower intracellular protein content, and decreased cell growth rate were found. Our data demonstrate that APE1 affects cell growth by directly acting on RNA quality control mechanisms, thus affecting gene expression through posttranscriptional mechanisms.


Subject(s)
Cell Nucleolus/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Nuclear Proteins/metabolism , RNA, Ribosomal/metabolism , Binding, Competitive , Cell Cycle , Cell Proliferation , DNA/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Electrophoresis, Gel, Two-Dimensional , HeLa Cells , Humans , Nuclear Proteins/chemistry , Nucleophosmin , Oxidation-Reduction , Peptide Mapping , Protein Binding , Protein Biosynthesis , Protein Interaction Mapping , Protein Multimerization , Protein Structure, Tertiary , RNA, Ribosomal/genetics , RNA, Ribosomal, 18S/metabolism , RNA, Ribosomal, 28S/metabolism , Transcription, Genetic
16.
J Proteome Res ; 8(3): 1131-42, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19226166

ABSTRACT

We combined high-throughput screening of differential mRNAs with mass spectrometric characterization of proteins obtained from osteocytes untreated and treated with Risedronate. Microarray analysis revealed, upon treatment, a marked upregulation of messengers encoding zinc-proteins. MS analysis identified 84 proteins in the osteocytes proteome map. Risedronate affected the expression of 10 proteins, associated with cytoskeleton, stress-response and metabolism. Data validated using gel imaging in combination with the GLaD post digestion isotopic labeling method provide the molecular basis for understanding the role of bisphosphonates as antiapoptotic drugs for osteocytes.


Subject(s)
Diphosphonates/pharmacology , Etidronic Acid/analogs & derivatives , Osteocytes/metabolism , Proteome/metabolism , RNA, Messenger/metabolism , Animals , Bone Density Conservation Agents/pharmacology , Cells, Cultured , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Etidronic Acid/pharmacology , Gene Expression Profiling , Isotope Labeling/methods , Osteocytes/drug effects , Risedronic Acid , Spectrometry, Mass, Electrospray Ionization
17.
Proteomics ; 9(4): 1058-74, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19180539

ABSTRACT

Apurinic apyrimidinic endonuclease/redox effector factor 1 (APE1/Ref-1) protects cells from oxidative stress by acting as a central enzyme in base excision repair pathways of DNA lesions and through its independent activity as a redox transcriptional co-activator. Dysregulation of this protein has been associated with cancer development. At present, contrasting data have been published regarding the biological relevance of the two functions as well as the molecular mechanisms involved. Here, we combined both mRNA expression profiling and proteomic analysis to determine the molecular changes associated with APE1 loss-of-expression induced by siRNA technology. This approach identified a role of APE1 in cell growth, apoptosis, intracellular redox state, mitochondrial function, and cytoskeletal structure. Overall, our data show that APE1 acts as a hub in coordinating different and vital functions in mammalian cells, highlighting the molecular determinants of the multifunctional nature of APE1 protein.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Proteomics/methods , Apoptosis , Cell Cycle , Cytoskeleton/metabolism , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Data Interpretation, Statistical , Down-Regulation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Mitochondria/metabolism , Oxidative Stress , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
18.
Cell Calcium ; 43(5): 457-68, 2008 May.
Article in English | MEDLINE | ID: mdl-17825906

ABSTRACT

Reactive oxygen species (ROS) have long been considered as toxic by-products of aerobic metabolism and appear involved in the pathogenesis of degenerative diseases. The physiological role of ROS as second messengers in cell signal transduction is, on the other hand, increasingly recognized. Here we investigated the effects of H(2)O(2) and extracellular nucleotides on calcium signalling in four osteoblastic cell lines. In the highly differentiated HOBIT cells, sensitive to nanomolar concentrations of ADP and UTP, millimolar H(2)O(2) induced oscillatory increases of the cytosolic calcium concentration followed by a steady and sustained calcium increase. Long lasting rhythmic calcium activity was induced by micromolar H(2)O(2) doses. The H(2)O(2)-induced calcium signals, due to both release from intracellular stores and influx from the extracellular milieu, were totally prevented by incubating the cells with the P2 receptor antagonist suramin or with the ATP/ADP hydrolyzing enzyme apyrase. In the osteosarcoma SaOS-2 cells micromolar H(2)O(2) failed to evoke calcium signals and millimolar H(2)O(2) induced a slowly developing calcium influx which was unaffected by suramin and apyrase. These cells responded to micromolar concentrations of ATP and ADP, but were largely insensitive to UTP. ROS 17/2.8 osteosarcoma cells were totally insensitive to ATP, ADP and UTP in keeping with the evidence that these cells lack functional purinergic receptors. In these cells, H(2)O(2) up to 1mM did not increase the cytosolic calcium concentration. In ROS/P2Y(2) cells, stably expressing the P2Y(2) receptor, spontaneous calcium oscillations were observed in 38% of the population and nanomolar concentration of extracellular ATP or UTP activated oscillations in quiescent cells. Spontaneous calcium signals were inhibited by suramin and apyrase. In these cells H(2)O(2) induced oscillatory calcium activity that was blocked by suramin and apyrase. The sensitivity of ROS/P2Y(2) cells to UTP decreased significantly in the presence of DTT, which was effective also in inhibiting spontaneous calcium oscillations. On the other hand, the membrane-impermeant thiol oxidant DTNB induced calcium oscillations that were inhibited by incubating the cells with suramin or apyrase. Since peroxide did not increase extracellular ATP in these cell lines, we propose that, in osteoblasts, mild oxidative conditions could activate purinergic signalling through the sensitization of P2Y(2) receptor.


Subject(s)
Adenine Nucleotides/pharmacology , Calcium Signaling , Hydrogen Peroxide/pharmacology , Osteoblasts/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Cell Line , Cell Line, Tumor , Humans , Osteoblasts/drug effects , Receptors, Purinergic P2Y2 , Uridine Triphosphate/pharmacology
19.
Proc Natl Acad Sci U S A ; 104(38): 15087-92, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17848519

ABSTRACT

Increased production of tumor necrosis factor alpha (TNF) in the bone marrow (BM) in response to both oxidative stress and T cell activation contributes to the bone loss induced by estrogen deficiency, but it is presently unknown whether oxidative stress causes bone loss through T cells. Here we show that ovariectomy causes an accumulation in the BM of reactive oxygen species, which leads to increased production of TNF by activated T cells through up-regulation of the costimulatory molecule CD80 on dendritic cells. Accordingly, bone loss is prevented by treatment of ovariectomized mice with either antioxidants or CTLA4-Ig, an inhibitor of the CD80/CD28 pathway. In summary, reactive oxygen species accumulation in the BM is an upstream consequence of ovariectomy that leads to bone loss by activating T cells through enhanced activity of BM dendritic cells, and these findings suggest that the CD80/CD28 pathway may represent a therapeutic target for postmenopausal bone loss.


Subject(s)
Bone Marrow Cells/immunology , Bone Resorption/immunology , Dendritic Cells/immunology , Estrogens/deficiency , Oxidative Stress , Abatacept , Animals , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Female , Flow Cytometry , Immunoconjugates/pharmacology , Mice , Mice, Inbred C57BL , Ovariectomy , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
J Cell Physiol ; 209(1): 44-55, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16741950

ABSTRACT

Nucleotide receptors signaling affects cell proliferation, with possible implications on tumorigenic processes. However, molecular targets and action mechanisms of the extracellular nucleotides are still poorly elucidated. We have previously shown in ARO cells that APE1/Ref-1, a transcriptional coactivator responsible for the maintenance of the cellular proliferative rate, is functionally controlled by P2-mediated signaling. Here, we demonstrate that extracellular ATP has a mitogenic effect on ARO cells, increasing ERK phosphorylation, AP1 activation, and cyclin D1 expression. Using the ATP/ADPase apyrase and the P2 receptor antagonist suramin, we show that the extracellular ATP, physiologically released by ARO cells, exerts mitogenic effects. A differential proteomic approach was used to identify molecular events associated with the ATP-induced cell proliferation. Among other proteins, Hsp90 was found upregulated upon ATP stimulation. Pretreatment with suramin completely blocked the ATP-induced Hsp90 activation, confirming the involvement of cell-surface P2 nucleotide receptors in the ATP-mediated activation of ARO cells. Treatment of proliferating ARO cells with suramin and apyrase significantly reduced the intracellular levels of Hsp90, suggesting an autocrine/paracrine mechanism of control on Hsp90 expression by extracellular ATP. The influence of Hsp90 on ATP-induced cell proliferation was also demonstrated by its specific inhibition with 17-AAG. The molecular pathway by which ATP stimulates cell proliferation was further investigated by siRNA strategies showing that Hsp90 is a target of APE1/Ref-1 functional activation. Stimulation of ARO cells with specific nucleotide receptors agonists evidenced a major involvement of P2Y1 and P2Y2 receptors in controlling the Hsp90 activation. Accordingly, these two receptors resulted significantly upregulated in sample biopsies from different thyroid tumors.


Subject(s)
Adenosine Triphosphate/pharmacology , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , HSP90 Heat-Shock Proteins/metabolism , Receptors, Purinergic P2/metabolism , Thyroid Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclin D , Cyclins/metabolism , Down-Regulation , Humans , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factor AP-1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...